1
|
Saad KM, Elmasry K, Baban B, Livingston MJ, Dong Z, Abdelmageed ME, Abdelaziz RR, Suddek GM, Elmarakby AA. Protocatechuic Acid Ameliorates Cisplatin-Induced Inflammation and Apoptosis in Mouse Proximal Tubular Cells. Int J Mol Sci 2025; 26:4115. [PMID: 40362355 PMCID: PMC12071929 DOI: 10.3390/ijms26094115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 04/16/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Cisplatin is a highly cytotoxic drug used for the treatment of head, neck, and soft tissue cancers; however, it has nephrotoxic effects that can lead to acute kidney injury. Protocatechuic acid (PCA) is a natural widely available antioxidant found in many fruits such as kiwi, mango, and berries. We have recently shown that PCA reduced renal injury in a mouse model of unilateral ureteral obstruction. The current study aims to investigate the protective effects of PCA in Cisplatin-induced inflammation in vitro in Boston University Mouse Proximal Tubular (BUMPT) cells. BUMPT cells were cultured in complete DMEM. Confluent BUMPT cells were then treated with 20 μM Cisplatin ± PCA 50 or 100 μM for 24 h. PCA treatment showed a dose-depending increase in % cell viability in Cisplatin-treated BUMPT cells. PCA treatment also dose-dependently decreased Cisplatin-induced increases in oxidative stress (ROS and TBARS), inflammation (p-NF-κB and IL-6), and apoptosis (cleaved caspase-3 and % of TUNEL+ cells) compared to Cisplatin-only treatment. The reduction in oxidative stress, inflammation, and apoptosis with PCA treatment in Cisplatin-treated BUMPT cells was associated with decreases in tubular physical barrier resistance and the expression of the tight junction protein zonula occludens-1 (ZO-1) when compared to BUMPT cells treated with Cisplatin alone. The current findings suggest that PCA treatment improves tubular barrier function in Cisplatin-treated BUMPT cells via reductions in oxidative stress, inflammation, and apoptosis.
Collapse
Affiliation(s)
- Karim M. Saad
- Department of Oral Biology and Diagnostic Sciences, The Dental College of Georgia, Augusta University, 1450 Laney Walker Blvd, Augusta, GA 30912, USA; (K.M.S.); (K.E.); (B.B.)
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; (M.E.A.); (R.R.A.); (G.M.S.)
| | - Khaled Elmasry
- Department of Oral Biology and Diagnostic Sciences, The Dental College of Georgia, Augusta University, 1450 Laney Walker Blvd, Augusta, GA 30912, USA; (K.M.S.); (K.E.); (B.B.)
- Department of Cellular Biology and Anatomy, The Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (M.J.L.); (Z.D.)
- Department of Human Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Babak Baban
- Department of Oral Biology and Diagnostic Sciences, The Dental College of Georgia, Augusta University, 1450 Laney Walker Blvd, Augusta, GA 30912, USA; (K.M.S.); (K.E.); (B.B.)
| | - Man J. Livingston
- Department of Cellular Biology and Anatomy, The Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (M.J.L.); (Z.D.)
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, The Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (M.J.L.); (Z.D.)
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| | - Marwa E. Abdelmageed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; (M.E.A.); (R.R.A.); (G.M.S.)
| | - Rania R. Abdelaziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; (M.E.A.); (R.R.A.); (G.M.S.)
| | - Ghada M. Suddek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; (M.E.A.); (R.R.A.); (G.M.S.)
| | - Ahmed A. Elmarakby
- Department of Oral Biology and Diagnostic Sciences, The Dental College of Georgia, Augusta University, 1450 Laney Walker Blvd, Augusta, GA 30912, USA; (K.M.S.); (K.E.); (B.B.)
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; (M.E.A.); (R.R.A.); (G.M.S.)
| |
Collapse
|
2
|
Yazgan Y, Cinar R. Gallic Acid Enhances Cisplatin-induced Death of Human Laryngeal Cancer Cells by Activating the TRPM2 Channel. DOKL BIOCHEM BIOPHYS 2025; 521:221-231. [PMID: 40216719 DOI: 10.1134/s1607672924601276] [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: 10/15/2024] [Revised: 11/25/2024] [Accepted: 11/25/2024] [Indexed: 05/16/2025]
Abstract
Cisplatin (CIS) is widely used in the treatment of laryngeal cancer, one of the most common and lethal cancers. However, it is not a satisfactory chemotherapeutic agent. Therefore, there is a need to identify new agents, such as gallic acid (GAL), that can exert a synergistic effect to elucidate the pathophysiological mechanisms of the chemotherapeutic effects of CIS and to increase the effectiveness of treatment by preventing drug resistance. For this purpose, we investigated the stimulatory role of GAL on CIS-induced human laryngeal cancer (Hep-2) cell death via TRPM2 channel activation. For the study, four groups were formed from human laryngeal cancer (Hep-2) cells as Control, GAL (1OO μM), CIS (25 μM), and GAL + CIS. In the analyses made, cell viability, glutathione (GSH) and glutathione peroxidase (GSH-Px) enzyme activity, lipid peroxidation (LPx) levels, inflammation markers I-1β, IL-6, and TNF-α, Total Oxidant/Antioxidant (TOS and TAS) status, reactive oxygen species (ROS), caspase (Cas-3-9) activity, Transient Receptor Potential Melastatin 2 (TRPM2), and Poly Adp Ribose Polymerase-1, (PARP-1) levels in the cells were determined. CIS treatment caused laryngeal cancer cell cytotoxic and increased Cas-3-9, ROS, IL-1β, TNF-α, IL-6, TOS, LPx, TRPM2, and PARP-1 levels while decreasing cell viability, GSH-Px, GSH, and TAS levels. The combination of GAL and CIS treatment made the treatment even more effective. In conclusion, the increase in ROS and cell death levels mediated by TRPM2 activation in CIS Hep-2 cells was further enhanced by GAL treatment. Thus, CIS chemotherapy in Hep-2 cells may be enhanced by the synergistic effect of the GAL combination, and drug resistance may be reduced.
Collapse
Affiliation(s)
- Yener Yazgan
- Department of Biophysics, Faculty of Medicine, Kastamonu University, Kastamonu, Türkiye.
| | - Ramazan Cinar
- Department of Biophysics, Faculty of Medicine, Bilecik Seyh Edebali University, Bilecik, Türkiye
| |
Collapse
|
3
|
Eid BG, Binmahfouz LS, Shaik RA, Bagher AM, Sirwi A, Abdel-Naim AB. Icariin inhibits cisplatin-induced ovarian toxicity via modulating NF-κB and PTEN/AKT/mTOR/AMPK axis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:1949-1959. [PMID: 39212737 DOI: 10.1007/s00210-024-03395-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
Cisplatin (CP) is a highly effective broad-spectrum chemotherapeutic agent for several solid tumors. However, its clinical use is associated with ovarian toxicity. Icariin (ICA) is a bioactive flavonoid of Epimedium brevicornum with reported protective activities against inflammation, oxidative stress and ovarian failure. This study aimed to explore the protective effects of ICA against CP-associated ovarian toxicity in rats. Rats were randomized into five groups and treated for 17 days: control, ICA (10 mg/kg/day, for 17 days. p.o.), CP (6 mg/kg, i.p. on days 7 and 14), CP + ICA (CP 6 mg/kg i.p. on days 7 and 14 and ICA 5 mg/kg p.o. daily), and CP + ICA (CP 6 mg/kg i.p. on days 7 and 14 and ICA 10 mg/kg p.o. daily). Our results indicated that ICA effectively improved ovarian reserve as indicated by attenuating CP-induced histolopathological changes and enhancing serum anti-müllerian hormone (AMH). Furthermore, co-administration of ICA with CP showed restoration of the oxidant-anti-oxidant balance in ovarian tissues, evidenced by decreased malondialdehyde (MDA) concentrations and elevated superoxide dismutase (SOD) and catalase (CAT) activities. Also, ICA suppressed ovarian inflammation as evidenced by down-regulation of the expression of interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α) and nuclear factor kappa B (NF-κB). ICA inhibited ovarian apoptosis in CP-treated rats by down-regulation of CASP3 and Bax and up-regulation of Bcl-2 mRNA expression. Further, ICA enhanced PTEN, p-AKT, p-mTOR, and p-AMPKα expression. In conclusion, ICA possesses a protective activity against CP-induced ovarian toxicity in rats by exhibiting antioxidant, antiinflammatory, anti-apoptotic activities and modulating NF-κB expression and PTEN/AKT/mTOR/AMPK axis in ovarian tissues.
Collapse
Affiliation(s)
- Basma G Eid
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Lenah S Binmahfouz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Rasheed A Shaik
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Amina M Bagher
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Alaa Sirwi
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Ashraf B Abdel-Naim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, 21589, Jeddah, Saudi Arabia.
| |
Collapse
|
4
|
Chuang CH, Tai YA, Wu TJ, Ho YJ, Yeh SL. Quercetin attenuates cisplatin-induced fatigue through mechanisms associated with the regulation of the HPA axis and MCP-1 signaling. Front Nutr 2025; 12:1530132. [PMID: 39949542 PMCID: PMC11821496 DOI: 10.3389/fnut.2025.1530132] [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: 11/18/2024] [Accepted: 01/17/2025] [Indexed: 02/16/2025] Open
Abstract
Introduction Cancer-related fatigue (CRF) is a common symptom induced by chemotherapy. The main objective of the present study was to investigate whether quercetin regulates the hypothalamic-pituitary-adrenal (HPA) axis and chemoattractant protein-1 (MCP-1) signaling, two factors contributing to CRF in mice exposed to cisplatin. Methods Male BALB/c mice were randomly assigned to the following five groups for 15 weeks: Control, CDDP, CDDP+TAK779 (an antagonist of MCP-1 receptor, human CC chemokine receptor R2 (CCR2)), CDDP+OQ (a diet containing 1% quercetin) and CDDP+IQ (quercetin given by ip, 10 mg/kg, 3 times/week). Results The results first showed that OQ and IQ significantly increased grip strength and locomotor activity, decreased plasma cortisol/corticosterone levels, and decreased the corticotropin releasing hormone (CRH) mRNA level in the brain tissues in mice exposed to CDDP. OQ and IQ also decreased CDDP-induced plasma levels of MCP-1 as well as the mRNA expression of MCP-1 and CCR2 in the brain stem. TAK779 significantly increased grip strength and tended to decrease the cortisol/corticosterone levels in CDDP-exposed mice, indicating the association between the HPA axis and MCP-1 signaling. Conclusion Taken together, the study suggests that quercetin could attenuate CDDP-induced CRF through the mechanisms associated with downregulation of the HPA axis and MCP-1 signaling in mice.
Collapse
Affiliation(s)
| | - Yu-An Tai
- Department of Nutritional Science, Chung Shan Medical University, Taichung, Taiwan
| | - Ting-Jing Wu
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Ying-Jui Ho
- Department of Psychology, Chung Shan Medical University, Taichung, Taiwan
| | - Shu-Lan Yeh
- Department of Nutritional Science, Chung Shan Medical University, Taichung, Taiwan
- Department of Nutrition, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
5
|
Guo Y, Xue L, Tang W, Xiong J, Chen D, Dai Y, Wu C, Wei S, Dai J, Wu M, Wang S. Ovarian microenvironment: challenges and opportunities in protecting against chemotherapy-associated ovarian damage. Hum Reprod Update 2024; 30:614-647. [PMID: 38942605 PMCID: PMC11369228 DOI: 10.1093/humupd/dmae020] [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: 10/30/2023] [Revised: 04/27/2024] [Indexed: 06/30/2024] Open
Abstract
BACKGROUND Chemotherapy-associated ovarian damage (CAOD) is one of the most feared short- and long-term side effects of anticancer treatment in premenopausal women. Accumulating detailed data show that different chemotherapy regimens can lead to disturbance of ovarian hormone levels, reduced or lost fertility, and an increased risk of early menopause. Previous studies have often focused on the direct effects of chemotherapeutic drugs on ovarian follicles, such as direct DNA damage-mediated apoptotic death and primordial follicle burnout. Emerging evidence has revealed an imbalance in the ovarian microenvironment during chemotherapy. The ovarian microenvironment provides nutritional support and transportation of signals that stimulate the growth and development of follicles, ovulation, and corpus luteum formation. The close interaction between the ovarian microenvironment and follicles can determine ovarian function. Therefore, designing novel and precise strategies to manipulate the ovarian microenvironment may be a new strategy to protect ovarian function during chemotherapy. OBJECTIVE AND RATIONALE This review details the changes that occur in the ovarian microenvironment during chemotherapy and emphasizes the importance of developing new therapeutics that protect ovarian function by targeting the ovarian microenvironment during chemotherapy. SEARCH METHODS A comprehensive review of the literature was performed by searching PubMed up to April 2024. Search terms included 'ovarian microenvironment' (ovarian extracellular matrix, ovarian stromal cells, ovarian interstitial, ovarian blood vessels, ovarian lymphatic vessels, ovarian macrophages, ovarian lymphocytes, ovarian immune cytokines, ovarian oxidative stress, ovarian reactive oxygen species, ovarian senescence cells, ovarian senescence-associated secretory phenotypes, ovarian oogonial stem cells, ovarian stem cells), terms related to ovarian function (reproductive health, fertility, infertility, fecundity, ovarian reserve, ovarian function, menopause, decreased ovarian reserve, premature ovarian insufficiency/failure), and terms related to chemotherapy (cyclophosphamide, lfosfamide, chlormethine, chlorambucil, busulfan, melphalan, procarbazine, cisplatin, doxorubicin, carboplatin, taxane, paclitaxel, docetaxel, 5-fluorouraci, vincristine, methotrexate, dactinomycin, bleomycin, mercaptopurine). OUTCOMES The ovarian microenvironment shows great changes during chemotherapy, inducing extracellular matrix deposition and stromal fibrosis, angiogenesis disorders, immune microenvironment disturbance, oxidative stress imbalances, ovarian stem cell exhaustion, and cell senescence, thereby lowering the quantity and quality of ovarian follicles. Several methods targeting the ovarian microenvironment have been adopted to prevent and treat CAOD, such as stem cell therapy and the use of free radical scavengers, senolytherapies, immunomodulators, and proangiogenic factors. WIDER IMPLICATIONS Ovarian function is determined by its 'seeds' (follicles) and 'soil' (ovarian microenvironment). The ovarian microenvironment has been reported to play a vital role in CAOD and targeting the ovarian microenvironment may present potential therapeutic approaches for CAOD. However, the relation between the ovarian microenvironment, its regulatory networks, and CAOD needs to be further studied. A better understanding of these issues could be helpful in explaining the pathogenesis of CAOD and creating innovative strategies for counteracting the effects exerted on ovarian function. Our aim is that this narrative review of CAOD will stimulate more research in this important field. REGISTRATION NUMBER Not applicable.
Collapse
Affiliation(s)
- Yican Guo
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Liru Xue
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Weicheng Tang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Jiaqiang Xiong
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Dan Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Yun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Chuqing Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Simin Wei
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Jun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei, China
| |
Collapse
|
6
|
Abady MM, Saadeldin IM, Han A, Bang S, Kang H, Seok DW, Kwon HJ, Cho J, Jeong JS. Melatonin and resveratrol alleviate molecular and metabolic toxicity induced by Bisphenol A in endometrial organoids. Reprod Toxicol 2024; 128:108628. [PMID: 38848930 DOI: 10.1016/j.reprotox.2024.108628] [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: 04/02/2024] [Revised: 05/21/2024] [Accepted: 05/28/2024] [Indexed: 06/09/2024]
Abstract
Bisphenol A (BPA), a widespread environmental contaminant, poses concerns due to its disruptive effects on physiological functions of the uterine endometrium. In contrast, melatonin (MT) and Resveratrol (RSV) are under scrutiny for their potential protective roles against BPA-induced damage. For the efficacy and ethical concerns in the animal test, endometrial organoids, three-dimensional models mimicking endometrium, serve as crucial tools for unraveling the impact of environmental factors on reproductive health. This study aimed to comprehensively characterize the morphological, molecular and metabolic responses of porcine endometrial organoids to BPA and assess the potential protective effects of MT and RSV. Porcine uteri were prepared, digested with collagenase, mixed with Matrigel, and incubated at 38°C with 5 % CO2. Passaging involved dissociation through trypsin-EDTA treatment and subculturing. The culture medium was refreshed every 2-3 days. To investigate the environmental impact on reproductive health, endometrial organoids were treated with BPA (0.5 µM), MT (with/without BPA at 0.1 µM), and/or RSV (10 µM). Various molecular screening using gene expression, western blotting, immunofluorescence staining, and metabolites profiling were assessed the effects of BPA, MT, and RSV in terms of cell viability, morphology, reproductivity, and metabolism alteration in the endometrial organoids. As expected, BPA induced structural and molecular disruptions in organoids, affecting cytoskeletal proteins, Wnt/β-catenin signaling, and epithelial/mesenchymal markers. It triggered oxidative stress and apoptotic pathways, altered miRNA expression, and disrupted the endocannabinoid system. The level of glucose, galactose, and essential amino acids were increased or decreased by approximately 1.5-3 times in BPA-treated groups compared to the control groups (p-value < 0.05), indicating metabolic changes. Moreover, MT and RSV treated groups exhibited protective effects, mitigating BPA-induced disruptions across multiple pathways. For the first time, our study models endometrial organoids, advancing understanding of environmental impacts on reproductive health.
Collapse
Affiliation(s)
- Mariam M Abady
- Organic Metrology Group, Division of Chemical and Material Metrology, Korea Research Institute of Standards and Science, 267 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea; Department of Bio-Analytical Science, University of Science and Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea; Department of Nutrition and Food Science, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Islam M Saadeldin
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Ayeong Han
- College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea; College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Seonggyu Bang
- College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea; College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Heejae Kang
- College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea; College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Dong Wook Seok
- Organic Metrology Group, Division of Chemical and Material Metrology, Korea Research Institute of Standards and Science, 267 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea; Department of Bio-Analytical Science, University of Science and Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Ha-Jeong Kwon
- Organic Metrology Group, Division of Chemical and Material Metrology, Korea Research Institute of Standards and Science, 267 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Jongki Cho
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea.
| | - Ji-Seon Jeong
- Organic Metrology Group, Division of Chemical and Material Metrology, Korea Research Institute of Standards and Science, 267 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea; Department of Bio-Analytical Science, University of Science and Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea.
| |
Collapse
|
7
|
Onalan E, Erbay B, Buran İK, Erol D, Tektemur A, Kuloglu T, Ozercan IH. Effects and Mechanism of AP39 on Ovarian Functions in Rats Exposed to Cisplatin and Chronic Immobilization Stress. J Menopausal Med 2024; 30:104-119. [PMID: 39315502 PMCID: PMC11439572 DOI: 10.6118/jmm.23015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/02/2024] [Accepted: 08/03/2024] [Indexed: 09/25/2024] Open
Abstract
OBJECTIVES Premature ovarian failure (POF) rat models are essential for elucidating the hormonal and ovarian molecular mechanisms of human POF diseases and developing new therapeutic agents. This study aimed to compare the applicability of chronic immobilization stress (CIS) as a POF model with that of cisplatin and to examine the impact of AP39, a mitochondrial protective agent, on ovarian function in rats treated with cisplatin and CIS. METHODS Sixty Sprague-Dawley female rats were divided equally into six groups (10 per group): Control, Cisplatin, AP39, Cisplatin + AP39, CIS, and CIS + AP39. Ovarian dysfunction was induced with cisplatin (3 mg/kg) or CIS. Forced swim test, hormone concentrations, estrous cyclicity, histopathology, follicle counts, and molecular alterations in the ovary and mitochondria were analyzed. RESULTS In the CIS and cisplatin groups, mitochondrial biogenesis, egg quality, hormonal profile, estrous cycle, and folliculogenesis significantly declined. Nonetheless, most of the parameters with undesirable results did not normalize after AP39 administration. CONCLUSIONS The cisplatin- and CIS-treated rats exhibited unshared deteriorated hormonal pathways and similarly disrupted gene expression patterns. Our current CIS model did not meet the human POF criteria, which include decreased estradiol levels, despite having advantages in terms of ease of modeling and reproducibility and demonstrating pathological changes similar to those observed in human POF. Therefore, rather than using this model as an POF model, using it as a representation of stress-induced ovarian dysfunction would be more appropriate.
Collapse
Affiliation(s)
- Ebru Onalan
- Department of Medical Biology, Faculty of Medicine, Firat University, Elazığ, Türkiye
| | - Bilgi Erbay
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - İlay Kavuran Buran
- Department of Medical Biology, Faculty of Medicine, Firat University, Elazığ, Türkiye.
| | - Deniz Erol
- Department of Medical Genetics, Faculty of Medicine, Firat University, Elazığ, Türkiye
| | - Ahmet Tektemur
- Department of Medical Biology, Faculty of Medicine, Firat University, Elazığ, Türkiye
| | - Tuncay Kuloglu
- Department of Histology and Embryology, Faculty of Medicine, Firat University, Elazığ, Türkiye
| | | |
Collapse
|
8
|
Hassanin HM, Kamal AA, Ismail OI. Resveratrol ameliorates atrazine-induced caspase-dependent apoptosis and fibrosis in the testis of adult albino rats. Sci Rep 2024; 14:17743. [PMID: 39085279 PMCID: PMC11291673 DOI: 10.1038/s41598-024-67636-z] [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: 01/28/2024] [Accepted: 07/15/2024] [Indexed: 08/02/2024] Open
Abstract
Pesticides like atrazine which are frequently present in everyday surroundings, have adverse impacts on human health and may contribute to male infertility. The work aimed to analyze the histological and biochemical effects of atrazine on the testis in adult albino rats and whether co-administration with resveratrol could reverse the effect of atrazine. Forty adult male albino rats in good health participated in this study. They were categorized at random into four groups: the Group Ӏ received water through a gastric tube for two months every day, the Group ӀӀ received resveratrol (20 mg/kg body weight (b.w.)) through a gastric tube for two months every day, the Group ӀӀӀ received atrazine (50 mg/kg bw) through a gastric tube for two months every day, the Group ӀV received concomitant doses of atrazine and resveratrol for two months every day. The testes of the animals were then carefully removed and prepared for biochemical, immunohistochemical, light, and electron microscopic studies. Atrazine exposure led to a significant decrease in serum testosterone hormone level, upregulation of caspase 3 and iNOS mRNA levels, destructed seminiferous tubules with few sperms in their lumens, many collagen fibres accumulation in the tunica albuginea and the interstitium, abnormal morphology of some sperms as well as many vacuolations, and damaged mitochondria in the cytoplasm of many germ cells. Concomitant administration of resveratrol can improve these adverse effects. It was concluded that atrazine exposure is toxic to the testis and impairs male fertility in adult rat and coadministration of resveratrol guards against this toxicity.
Collapse
Affiliation(s)
- Hala Mohamed Hassanin
- Human Anatomy and Embryology Department, Faculty of Medicine, Assiut University, Assiut, 71515, Egypt
| | - Asmaa A Kamal
- Medical Biochemistry Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Omnia I Ismail
- Human Anatomy and Embryology Department, Faculty of Medicine, Assiut University, Assiut, 71515, Egypt.
| |
Collapse
|
9
|
Li F, Zhu F, Wang S, Hu H, Zhang D, He Z, Chen J, Li X, Cheng L, Zhong F. Icariin alleviates cisplatin-induced premature ovarian failure by inhibiting ferroptosis through activation of the Nrf2/ARE pathway. Sci Rep 2024; 14:17318. [PMID: 39068256 PMCID: PMC11283570 DOI: 10.1038/s41598-024-67557-x] [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: 02/04/2024] [Accepted: 07/12/2024] [Indexed: 07/30/2024] Open
Abstract
Cisplatin is a widely used chemotherapeutic drug that can induce ovarian damage. Icariin (ICA), a natural antioxidant derived from Epimedium brevicornum Maxim., has been found to protect against organ injury. The aim of the present study was to investigate whether ICA can exert an ovarian-protective effect on cisplatin induced premature ovarian failure (POF) and the underlying mechanism involved. The preventive effect of ICA was evaluated using body weight, the oestrous cycle, ovarian histological analysis, and follicle counting. ICA treatment increased body weight, ovarian weight, and the number of follicles and improved the oestrous cycle in POF mice. ICA reduced cisplatin-induced oxidative damage and upregulated the protein expression levels of Nrf2, GPX4 and HO-1. Moreover, ICA reduced the expression levels of Bax and γH2AX and inhibited ovarian apoptosis. In addition, ICA activated the Nrf2 pathway in vitro and reversed changes in the viability of cisplatin-induced KGN cells, reactive oxygen species (ROS) levels, lipid peroxidation, and apoptosis, and these effects were abrogated when Nrf2 was knocked down or inhibited. Molecular docking confirmed that ICA promotes the release of Nrf2 by competing with Nrf2 for binding to Keap1. The inhibitory effects of ICA on cisplatin-induced oxidative stress, ferroptosis, and apoptosis may be mediated by its modulatory effects on the Nrf2 pathway, providing a novel perspective on the potential mechanisms by which ICA prevents POF.
Collapse
Affiliation(s)
- Fangfang Li
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Fengyu Zhu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Shushan District, Hefei, 230022, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
- Ministry of Education of the People's Republic of China, Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Siyuan Wang
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Huiqing Hu
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Di Zhang
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Zhouying He
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Jiaqi Chen
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Xuqing Li
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Shushan District, Hefei, 230022, Anhui, China.
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China.
- Ministry of Education of the People's Republic of China, Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China.
| | - Linghui Cheng
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Shushan District, Hefei, 230022, Anhui, China.
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China.
- Ministry of Education of the People's Republic of China, Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China.
| | - Fei Zhong
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China.
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei, 230032, Anhui, China.
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China.
| |
Collapse
|
10
|
Ramadan OI, S. Ali L, M. Abd-Allah F, E. A. Ereba R, S. Humeda H, A. Damanhory A, E. Moustafa A, M. Younes A, M. Y. Awad M, A. A. Omar N. Co-administration of either curcumin or resveratrol with cisplatin treatment decreases hepatotoxicity in rats via anti-inflammatory and oxidative stress-apoptotic pathways. PeerJ 2024; 12:e17687. [PMID: 39056050 PMCID: PMC11271648 DOI: 10.7717/peerj.17687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 06/14/2024] [Indexed: 07/28/2024] Open
Abstract
Background Cisplatin (CIS) is a broad-spectrum anticancer drug, with cytotoxic effects on either malignant or normal cells. We aimed to evaluate the hepatotoxicity in rats caused by CIS and its amelioration by the co-administration of either curcumin or resveratrol. Materials and Methods Forty adult male rats divided into four equal groups: (control group): rats were given a saline solution (0.9%) once intraperitoneally, daily for the next 28 days; (cisplatin group): rats were given a daily oral dose of saline solution (0.9%) for 28 days after receiving a single dose of cisplatin (3.3 mg/kg) intraperitoneally for three successive days; (CIS plus curcumin/resveratrol groups): rats received the same previous dose of cisplatin (3.3 mg/kg) daily for three successive days followed by oral administration of either curcumin/resveratrol solution at a dose of (20 mg/kg) or (10 mg/kg) consequently daily for 28 days. Different laboratory tests (ALT, AST, ALP, bilirubin, oxidative stress markers) and light microscopic investigations were done. Results Administration of CIS resulted in hepatotoxicity in the form of increased liver enzymes, oxidative stress markers; degenerative and apoptotic changes, the co-administration of CIS with either curcumin or resveratrol improved hepatotoxicity through improved microscopic structural changes, reduction in liver enzymes activity, decreased oxidative stress markers, improved degenerative, and apoptotic changes in liver tissues. Conclusion Co-administration of either curcumin or resveratrol with cisplatin treatment could ameliorate hepatotoxicity caused by cisplatin in rats via anti-inflammatory and oxidative stress-apoptotic pathways.
Collapse
Affiliation(s)
- Osama I. Ramadan
- Department of Dental Basic Sciences, Faculty of Dentistry, Applied Science Private University, Amman, Jordan
- Histology Department, Damietta Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Lashin S. Ali
- Department of Basic Medical Science, Faculty of Dentistry, Al-Ahliyya Amman University, Amman, Jordan
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Fatma M. Abd-Allah
- Histology Department, Damietta Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Rafik E. A. Ereba
- Department of Pharmacology, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Humeda S. Humeda
- Department of Physiology, Faculty of Medicine, Alzaiem AlAzhari University, Khartoum North, Sudan
- Physiology Department, General Medicine Practice Program, Batterjee Medical College, Aseer, Saudi Arabia
| | - Ahmed A. Damanhory
- Department of Biochemistry, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
- Department of Biochemistry, General Medicine Practice Program, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Ahmed E. Moustafa
- Medical Biochemistry Department, Damietta Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Amr M. Younes
- Anatomy and Embryology Department, Damietta Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Moaaz M. Y. Awad
- Anatomy and Embryology Department, Damietta Faculty of Medicine, Al-Azhar University, Cairo, Egypt
- Anatomy Department, General Medicine Practice Program, Batterjee Medical Collage, Aseer, Saudi Arabia
| | - Nassar A. A. Omar
- Anatomy Department, General Medicine Practice Program, Batterjee Medical Collage, Aseer, Saudi Arabia
- Department of Anatomy, Faculty of Medicine, Sohag University, Sohag, Egypt
| |
Collapse
|
11
|
Yang Q, Meng D, Zhang Q, Wang J. Advances in the role of resveratrol and its mechanism of action in common gynecological tumors. Front Pharmacol 2024; 15:1417532. [PMID: 39086397 PMCID: PMC11288957 DOI: 10.3389/fphar.2024.1417532] [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: 04/15/2024] [Accepted: 06/26/2024] [Indexed: 08/02/2024] Open
Abstract
The incidence of common gynecological malignancies remains high, with current treatments facing multiple limitations and adverse effects. Thus, continuing the search for safe and effective oncologic treatment strategies continues. Resveratrol (RES), a natural non-flavonoid polyphenolic compound, is widely found in various plants and fruits, such as grapes, Reynoutria japonica Houtt., peanuts, and berries. RES possesses diverse biological properties, including neuroprotective, antitumor, anti-inflammatory, and osteoporosis inhibition effects. Notably, RES is broadly applicable in antitumor therapy, particularly for treating gynecological tumors (cervical, endometrial, and ovarian carcinomas). RES exerts antitumor effects by promoting tumor cell apoptosis, inhibiting cell proliferation, invasion, and metastasis, regulating tumor cell autophagy, and enhancing the efficacy of antitumor drugs while minimizing their toxic side effects. However, comprehensive reviews on the role of RES in combating gynecological tumors and its mechanisms of action are lacking. This review aims to fill this gap by examining the RES antitumor mechanisms of action in gynecological tumors, providing valuable insights for clinical treatment.
Collapse
Affiliation(s)
- Qian Yang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dandan Meng
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qingchen Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jin Wang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
12
|
Markowska A, Antoszczak M, Markowska J, Huczyński A. Gynotoxic Effects of Chemotherapy and Potential Protective Mechanisms. Cancers (Basel) 2024; 16:2288. [PMID: 38927992 PMCID: PMC11202309 DOI: 10.3390/cancers16122288] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Chemotherapy is one of the leading cancer treatments. Unfortunately, its use can contribute to several side effects, including gynotoxic effects in women. Ovarian reserve suppression and estrogen deficiency result in reduced quality of life for cancer patients and are frequently the cause of infertility and early menopause. Classic alkylating cytostatics are among the most toxic chemotherapeutics in this regard. They cause DNA damage in ovarian follicles and the cells they contain, and they can also induce oxidative stress or affect numerous signaling pathways. In vitro tests, animal models, and a few studies among women have investigated the effects of various agents on the protection of the ovarian reserve during classic chemotherapy. In this review article, we focused on the possible beneficial effects of selected hormones (anti-Müllerian hormone, ghrelin, luteinizing hormone, melatonin), agents affecting the activity of apoptotic pathways and modulating gene expression (C1P, S1P, microRNA), and several natural (quercetin, rapamycin, resveratrol) and synthetic compounds (bortezomib, dexrazoxane, goserelin, gonadoliberin analogs, imatinib, metformin, tamoxifen) in preventing gynotoxic effects induced by commonly used cytostatics. The presented line of research appears to provide a promising strategy for protecting and/or improving the ovarian reserve in the studied group of cancer patients. However, well-designed clinical trials are needed to unequivocally assess the effects of these agents on improving hormonal function and fertility in women treated with ovotoxic anticancer drugs.
Collapse
Affiliation(s)
- Anna Markowska
- Department of Perinatology and Women’s Health, Poznań University of Medical Sciences, 60-535 Poznań, Poland
| | - Michał Antoszczak
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, 61-614 Poznań, Poland
| | - Janina Markowska
- Gynecological Oncology Center, Poznańska 58A, 60-850 Poznań, Poland;
| | - Adam Huczyński
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, 61-614 Poznań, Poland
| |
Collapse
|
13
|
Sharma R, Ansari MM, Alam M, Fareed M, Ali N, Ahmad A, Sultana S, Khan R. Sophorin mitigates flutamide-induced hepatotoxicity in wistar rats. Toxicon 2024; 243:107722. [PMID: 38653393 DOI: 10.1016/j.toxicon.2024.107722] [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: 01/18/2024] [Revised: 04/06/2024] [Accepted: 04/12/2024] [Indexed: 04/25/2024]
Abstract
Flutamide is frequently used in the management of prostate cancer, hirsutism, and acne. It is a non-steroidal anti-androgenic drug and causes hepatotoxicity. The current study's objective is to evaluate sophorin's hepatoprotective effectiveness against flutamide-induced hepatotoxicity in Wistar rats. Sophorin is a citrus flavonoid glycoside, also known as rutin, which is a low molecular weight polyphenolic compound with natural antioxidant properties and reported to have promising hepatoprotective efficacy. In this study, sophorin was used at a dose of 100 mg/kg body weight in purified water via oral route for 4 week daily whereas, flutamide was used at a dose of 100 mg kg/b.wt for 4 weeks daily in 0.5% carboxy methyl cellulose (CMC) through the oral route for the induction of hepatotoxicity. Flutamide administration leads to enhanced reactive oxygen species (ROS) generation, an imbalance in redox homeostasis and peroxidation of lipid resulted in reduced natural antioxidant level in liver tissue. Our result demonstrated that sophorin significantly abrogate flutamide induced lipid peroxidation, protein carbonyl (PC), and also significantly increasesed in enzymatic activity/level of tissue natural antioxidant such as reduced glutathione(GSH), glutathione reductase(GR), catalase, and superoxide dismutase(SOD). Additionally, sophorin reduced the activity of cytochrome P450 3A1 in liver tissue which was elevated due to flutamide treatment. Furthermore, sophorin treatment significantly decreased the pro-inflammatory cytokines (TNF-α and IL-6) level. Immunohistochemical analysis for the expression of inflammatory proteins (iNOS and COX-2) in hepatic tissue was decreased after sophorin treatment against flutamide-induced hepatotoxicity. Moreover, sophorin suppressed the infiltration of mast cells in liver tissue which further showed anti-inflammatory potential of sophorin. Our histological investigation further demonstrated sophorin's hepatoprotective function by restoring the typical histology of the liver. Based on the aforementioned information, we are able to come to the conclusion that sophorin supplementation might benefit wistar rats with flutamide-induced hepatic damage by reducing oxidative stress and hepatocellular inflammation.
Collapse
Affiliation(s)
- Rishi Sharma
- Molecular Carcinogenesis and Chemoprevention Division, Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Md Meraj Ansari
- Heavy Metal and Clinical Toxicology Laboratory, Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Manzar Alam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Mohammad Fareed
- Department of Environmental Health and Clinical Epidemiology, Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamil Nadu, India
| | - Nemat Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Anas Ahmad
- Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Sarwat Sultana
- Molecular Carcinogenesis and Chemoprevention Division, Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, Mohali, 140306, Punjab, India.
| |
Collapse
|
14
|
Demir EA. Syringic acid alleviates cisplatin-induced ovarian injury through modulating endoplasmic reticulum stress, inflammation and Nrf2 pathway. J Trace Elem Med Biol 2024; 82:127356. [PMID: 38086229 DOI: 10.1016/j.jtemb.2023.127356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/29/2023] [Accepted: 12/04/2023] [Indexed: 01/10/2024]
Abstract
BACKGROUND Reproductive toxicity is one of the most important side effects of cisplatin (CIS) and leading to discontinuation of treatment. Syringic acid (SA) is a phenolic acid whose industrial use has increased in recent years due to its antioxidant properties. Recent reports highlight the importance of the supressed Nrf2 pathway in the molecular pathogenesis of CIS toxicity. Therefore, this study aimed to evaluate the therapeutic effect of SA on CIS-induced ovotoxicity through the Nrf2 pathway for the first time. MATERIAL AND METHODS Thirty female rats were divided into 5 groups: control, CIS, CIS+SA (5 and 10 mg/kg) and only SA (per se, 10 mg/kg). CIS was administered intraperitoneally at a dose of 5 mg/kg on the 1st day, injections of SA followed by three consecutive days in the rats. Serum anti-mullerian hormone (AMH) levels and ovarian oxidative stress (OS), inflammation, endoplasmic reticulum stress (ERS), apoptosis and Nrf2 pathway markers were determined colorimetrically. Histopathological examinations of the ovaries with hematoxylin and eosin staining were also used to evaluate CIS-induced ovotoxicity. RESULTS The CIS treatment depleted serum AMH levels, caused histopathological findings and increased OS, inflammation, ERS and apoptosis levels in ovarian tissue. However, treatments with SA significantly ameliorated CIS-induced biochemical and histopathological changes by activating Nrf2 pathway. CONCLUSION The promising adjuvant potential of SA to alleviate CIS-related ovarian damage should be supported by more comprehensive studies.
Collapse
Affiliation(s)
- Elif Ayazoglu Demir
- Department of Chemistry and Chemical Processing Technologies, Macka Vocational School, Karadeniz Technical University, 61750 Trabzon, Turkey.
| |
Collapse
|
15
|
Hu H, Zhang J, Xin X, Jin Y, Zhu Y, Zhang H, Fan R, Ye Y, Li D. Efficacy of natural products on premature ovarian failure: a systematic review and meta-analysis of preclinical studies. J Ovarian Res 2024; 17:46. [PMID: 38378652 PMCID: PMC10877904 DOI: 10.1186/s13048-024-01369-5] [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: 12/12/2023] [Accepted: 02/05/2024] [Indexed: 02/22/2024] Open
Abstract
OBJECTIVE This study aims to investigate the effects of natural products on animal models of premature ovarian failure (POF). METHODS We conducted comprehensive literature searches and identified relevant studies that examined the protective effects of natural products on experimental POF. We extracted quantitative data on various aspects such as follicular development, ovarian function, physical indicators, oxidative stress markers, inflammatory factors, and protein changes. The data was analyzed using random-effects meta-analyses, calculating pooled standardized mean differences and 95% confidence intervals. Heterogeneity was assessed using the I2 statistic, and bias was estimated using the SYRCLE tool. RESULTS Among the 879 reviewed records, 25 articles met our inclusion criteria. These findings demonstrate that treatment with different phytochemicals and marine natural products (flavonoids, phenols, peptides, and alkaloids, etc.) significantly improved various aspects of ovarian function compared to control groups. The treatment led to an increase in follicle count at different stages, elevated levels of key hormones, and a decrease in atretic follicles and hormone levels associated with POF. This therapy also reduced oxidative stress (specifically polyphenols, resveratrol) and apoptotic cell death (particularly flavonoids, chrysin) in ovarian granulosa cells, although it showed no significant impact on inflammatory responses. The certainty of evidence supporting these findings ranged from low to moderate. CONCLUSIONS Phytochemicals and marine natural product therapy (explicitly flavonoids, phenols, peptides, and alkaloids) has shown potential in enhancing folliculogenesis and improving ovarian function in animal models of POF. These findings provide promising strategies to protect ovarian reserve and reproductive health. Targeting oxidative stress and apoptosis pathways may be the underlying mechanism.
Collapse
Affiliation(s)
- Hangqi Hu
- Department of Traditional Chinese Medicine, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Jiacheng Zhang
- Department of Traditional Chinese Medicine, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Xiyan Xin
- Department of Traditional Chinese Medicine, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Yuxin Jin
- Department of Traditional Chinese Medicine, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Yutian Zhu
- Department of Traditional Chinese Medicine, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Haolin Zhang
- Department of Traditional Chinese Medicine, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Ruiwen Fan
- Department of Traditional Chinese Medicine, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Yang Ye
- Department of Traditional Chinese Medicine, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, China.
| | - Dong Li
- Department of Traditional Chinese Medicine, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing, 100191, China.
| |
Collapse
|
16
|
Yan H, Wang P, Yang F, Cheng W, Chen C, Zhai B, Zhou Y. Anticancer therapy-induced adverse drug reactions in children and preventive and control measures. Front Pharmacol 2024; 15:1329220. [PMID: 38425652 PMCID: PMC10902428 DOI: 10.3389/fphar.2024.1329220] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
In recent years, considerable achievements have been made in pediatric oncology with the innovation and development of antitumor drugs. However, compared to adults, children as a special group have not yet matured fully in terms of liver and kidney function. Moreover, pediatric patients are prone to more adverse drug reactions (ADRs) from the accumulation of antineoplastic drugs due to their smaller body size and larger body surface area. Chemotherapy-related ADRs have become a non-negligible factor that affects cancer remission. To date, studies on ADRs in pediatric cancer patients have emerged internationally, but few systematic summaries are available. Here, we reviewed the various systemic ADRs associated with antitumor drugs in children and adolescent patients, as well as the advances in strategies to cope with ADRs, which consisted of neurotoxicity, hematological toxicity, cardiotoxicity, ADRs of the respiratory system and gastrointestinal system and urinary system, ADRs of the skin and its adnexa, allergic reactions, and other ADRs. For clinicians and researchers, understanding the causes, symptoms, and coping strategies for ADRs caused by anticancer treatments will undoubtedly benefit more children.
Collapse
Affiliation(s)
- Hui Yan
- Henan Provincial Clinical Research Center for Pediatric Diseases, Henan Key Laboratory of Pediatric Genetics and Metabolic Diseases, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
- Department of Cardiothoracic Surgery, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Penggao Wang
- Department of Cardiothoracic Surgery, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Fang Yang
- Department of Cardiothoracic Surgery, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Weyland Cheng
- Henan Provincial Clinical Research Center for Pediatric Diseases, Henan Key Laboratory of Pediatric Genetics and Metabolic Diseases, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Congcong Chen
- Henan Provincial Clinical Research Center for Pediatric Diseases, Henan Key Laboratory of Pediatric Genetics and Metabolic Diseases, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Bo Zhai
- Department of Cardiothoracic Surgery, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Yang Zhou
- Henan Provincial Clinical Research Center for Pediatric Diseases, Henan Key Laboratory of Pediatric Genetics and Metabolic Diseases, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
- Department of Cardiothoracic Surgery, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| |
Collapse
|
17
|
Zaki NF, Orabi SH, Abdel-Bar HM, Elbaz HT, Korany RMS, Ismail AK, Daoush WM, Abduljabbar MH, Alosaimi ME, Alnemari RM, Mahboub HH, Ahmed MM. Zinc oxide resveratrol nanoparticles ameliorate testicular dysfunction due to levofloxacin-induced oxidative stress in rats. Sci Rep 2024; 14:2752. [PMID: 38307943 PMCID: PMC10837121 DOI: 10.1038/s41598-024-52830-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 01/24/2024] [Indexed: 02/04/2024] Open
Abstract
The present work is aimed to assess the protective influence of zinc oxide resveratrol nanoparticles against oxidative stress-associated testicular dysfunction. The number of 50 male albino rats were randomly separated into five groups (n = 10): Group I, control: rats gavage distilled water orally; Group II, Levofloxacin: rats that administered Levofloxacin (LFX) softened in distilled water at a dosage of 40 mg/kg-1 BW orally every other day; Group III, Zn-RSV: rats administered with Zn-RSV (zinc oxide resveratrol in distilled water at a dose 20 mg/kg-1 BW orally every other day; Group IV, (LFX + Zn-RSV): rats that were administered with Levofloxacin along with Zn-RSV nPs; Group V, Levofloxacin + Zn: rats were administered with Levofloxacin and Zno at a dose of 20 mg/kg-1 BW orally every other day as mentioned before. This study lasted for 2 months. Sera were collected to assess luteinizing hormone (LH), follicle-stimulating hormone (FSH), and testosterone values. Testicular tissues were utilized to evaluate levels of superoxide dismutase (SOD), nitric oxide (NO), malondialdehyde (MDA), and catalase (CAT). Semen samples were utilized to measure their quality (motility, concentration, and vitality). Histopathological and immune histochemical techniques investigated the morphological changes in the testis. Rats treated with Levofloxacin showed significantly lower levels of serum LH, testosterone, FSH, testicular enzymatic NO, catalase, SOD, BAX, and BCL-2 immune reactivity and sperm quality but significantly greater testicular malondialdehyde and caspase-3 immuno-reactivity Compared to both control and zinc oxide resveratrol treatment. Zinc oxide resveratrol nanoparticles ameliorated the harmful side effects of Levofloxacin. Improvements were more pronounced in the co-treatment (LFX + Zn-RSV) Zinc oxide resveratrol group than in the co-treatment (LFX + Zno) Zinc oxide group. Zinc oxide resveratrol nanoparticles could be a possible solution for levofloxacin oxidative stress-induced fertility problems.
Collapse
Affiliation(s)
- Naglaa F Zaki
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Egypt
| | - Sahar H Orabi
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Egypt
| | - Hend Mohamed Abdel-Bar
- Department of Pharmaceutics, Faculty of Pharmacy, University of Sadat City, Sadat City, Egypt
| | - Hamed T Elbaz
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Egypt
| | - Reda M S Korany
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Ayman K Ismail
- Department of Forensic Medicine and Toxicology, College of Veterinary Medicine, Suez Canal University, PO Box 41522, Ismailia, Egypt
| | - Walid M Daoush
- Department of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), P.O. Box 90950, 11623, Riyadh, Saudi Arabia
- Department of Production Technology, Faculty of Technology and Education, Helwan University, Saray-El Qoupa, El Sawah Street, Cairo, 11281, Egypt
| | - Maram H Abduljabbar
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, 21944, Taif, Saudi Arabia
| | - Manal E Alosaimi
- Department of Basic Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, P.O. Box 84428, 11671, Riyadh, Saudi Arabia.
| | - Reem M Alnemari
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Taif University, 21944, Taif, Saudi Arabia
| | - Heba H Mahboub
- Department of Aquatic Animal Medicine, Faculty of Veterinary Medicine, Zagazig University, PO Box 44511, Zagazig, Sharkia, Egypt
| | - Mohamed M Ahmed
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Egypt
| |
Collapse
|
18
|
Demir EA, Mentese A, Yilmaz ZS, Alemdar NT, Demir S, Aliyazicioglu Y. Evaluation of the therapeutic effects of arbutin on cisplatin-induced ovarian toxicity in rats through endoplasmic reticulum stress and Nrf2 pathway. Reprod Biol 2023; 23:100824. [PMID: 37976616 DOI: 10.1016/j.repbio.2023.100824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 11/03/2023] [Accepted: 11/05/2023] [Indexed: 11/19/2023]
Abstract
Arbutin (ARB) is a glycosylated hydroquinone with potent antioxidant effects. Although cisplatin (CP) is widely used in chemotherapy, its toxicity in healthy tissues, including ovotoxicity, is an insurmountable problem. This study aimed to evaluate the therapeutic effect of ARB against CP-related ovototoxicity by including nuclear factor erythroid 2-related factor 2 (Nrf2) pathway in rats for the first time. Rats treated one dose of CP (5 mg/kg) on the first day, followed by ARB (5 and 10 mg/kg) for three days. Serum reproductive hormone levels were determined using ELISA kits. Oxidative stress (OS), inflammation, endoplasmic reticulum stress (ERS) and apoptosis markers in ovarian tissue were also determined colorimetrically. In addition, how CP affects Nrf2 pathway and the effect of ARB on this situation were also addressed. ARB treatment reduced the levels of markers of OS, inflammation, ERS and apoptosis in ovarian tissue of CP-stimulated animals. ARB regenerated the depleted antioxidant system by triggering Nrf2 pathway in the ovarian tissues of animals stimulated by CP. Histological findings also supported the therapeutic efficacy of ARB. The results indicate that ARB may have therapeutic effects against CP-induced reproductive toxicity with its Nrf2 activator potential. ARB should be tested in more extensive studies as a new generation chemopreventive candidate molecule.
Collapse
Affiliation(s)
- Elif Ayazoglu Demir
- Department of Chemistry and Chemical Processing Technologies, Macka Vocational School, Karadeniz Technical University, 61750 Trabzon, Turkiye
| | - Ahmet Mentese
- Department of Medical Biochemistry, Faculty of Medicine, Karadeniz Technical University, 61080 Trabzon, Turkiye
| | - Zeynep Sagnak Yilmaz
- Department of Medical Pathology, Faculty of Medicine, Karadeniz Technical University, 61080 Trabzon, Turkiye
| | - Nihal Turkmen Alemdar
- Department of Medical Biochemistry, Graduate School of Health Sciences, Karadeniz Technical University, 61080 Trabzon, Turkiye; Department of Medical Services and Techniques, Vocational School of Health Services, Recep Tayyip Erdogan University, 53100 Rize, Turkiye
| | - Selim Demir
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Karadeniz Technical University, 61080 Trabzon, Turkiye.
| | - Yuksel Aliyazicioglu
- Department of Medical Biochemistry, Faculty of Medicine, Karadeniz Technical University, 61080 Trabzon, Turkiye
| |
Collapse
|
19
|
Zhao LL, Jayeoye TJ, Ashaolu TJ, Olatunji OJ. Pinostrobin, a dietary bioflavonoid exerts antioxidant, anti-inflammatory, and anti-apoptotic protective effects against methotrexate-induced ovarian toxicity in rats. Tissue Cell 2023; 85:102254. [PMID: 37866152 DOI: 10.1016/j.tice.2023.102254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 10/02/2023] [Accepted: 10/19/2023] [Indexed: 10/24/2023]
Abstract
This study investigated the protective activities of pinostrobin (PIN) against methotrexate (MTX)-induced ovarian toxicity. Female rats were administered with PIN (50 mg/kg) for 4 weeks, while MTX was administered from weeks 2-4 of PIN treatment. Serum hormonal profiles, ovarian oxidative stress, inflammatory and apoptotic biomarkers as well as ovarian histomorphometry were evaluated. MTX administration elicited profound deficit in serum progesterone and estrogen (E2) levels, while luteinizing hormone (LH) and follicle stimulating hormone (FSH) were significantly increased. Additionally, MTX administration was associated with significant increases in ovarian malondialdehyde, nitric oxide, NF-кB, TNF-α, IL-6, IL-1β, iNOS and caspase-3 activity, as well as notable reduction in the activities of glutathione peroxidase, catalase and superoxide dismutase as well as the level of glutathione. Whereas, treatment with PIN significantly decreased serum levels of FSH and LH, as well as ovarian levels of NO, MDA, caspase 3, NF-κB, IL-1β, IL-6, TNF-α and iNOS. PIN also significantly upregulated GSH, GPx, CAT and SOD in the ovarian tissues as well as increased serum E2 and progesterone levels compared to the MTX group. Furthermore, PIN significantly restored altered ovarian histoarchitecture in the treated group. These findings suggests that PIN exerts protective effects against MTX-triggered ovarian damages.
Collapse
Affiliation(s)
- Ling-Ling Zhao
- Department of Chemistry and Chemical Engineering, Yancheng Institute of Technology, Yancheng, China
| | - Titilope John Jayeoye
- Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | | | | |
Collapse
|
20
|
Moslehi AH, Hoseinpour F, Saber A, Akhavan Taheri M, Hashemian AH. Fertility-enhancing effects of inositol & vitamin C on cisplatin induced ovarian and uterine toxicity in rats via suppressing oxidative stress and apoptosis. Food Chem Toxicol 2023; 179:113995. [PMID: 37619831 DOI: 10.1016/j.fct.2023.113995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 07/22/2023] [Accepted: 08/16/2023] [Indexed: 08/26/2023]
Abstract
Cisplatin can lead to infertility due to its negative impact on the uterus and ovaries. This study aimed to explore the effects of Inositol and vitamin C on cisplatin-induced infertility. Forty-eight adult female Wistar rats were divided into eight groups (N = 6) and orally treated for 21 days. The treatments were as follows: negative control (saline), positive control (saline and cisplatin injected into the abdomen on day 15), T1-T3: rats given vitamin C (150 mg/kg), Inositol (420 mg/kg), and vitamin C + Inositol, respectively, along with cisplatin injected into the abdomen on day 15, T4-T6: rats given only vitamin C, Inositol, and vitamin C + Inositol, respectively. Vitamin C and Inositol enhanced cisplatin-induced histopathological improvements in the uterus and ovaries, raising progesterone and estradiol serum levels. Furthermore, the supplements enhanced ESR1 gene expression in the uterus and ovary, reducing uterine and ovarian apoptosis caused by cisplatin through modulation of caspase 3, 8, and Bcl-2 gene levels. These substances decreased ovarian and uterine malondialdehyde levels, boosted total antioxidant capacity and superoxide dismutase, and alleviated oxidative stress. The findings reveal that vitamin C and Inositol shield against cisplatin-related infertility by reducing oxidative stress and apoptosis in the uterus and ovaries.
Collapse
Affiliation(s)
- Amir Hosein Moslehi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Razi University, Kermanshah, Iran
| | - Fatemeh Hoseinpour
- Department of Basic Sciences, Faculty of Veterinary Medicine, Razi University, Kermanshah, Iran.
| | - Amir Saber
- Department of Nutritional Sciences, School of Nutritional Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Maryam Akhavan Taheri
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran; Gametogenesis Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Amir Hossein Hashemian
- Research Center for Environmental Determinants of Health (RCEDH), Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Biostatistics, School of Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
21
|
Ciplak B, Turmus EG, Kara O, Daglioglu G, Altindag MM, Simsek Y, Daglioglu YK, Kara M. Does resveratrol reduce cisplatin-induced ovarian damage? REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2023; 69:e20230314. [PMID: 37585992 PMCID: PMC10427173 DOI: 10.1590/1806-9282.20230314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 05/07/2023] [Indexed: 08/18/2023]
Abstract
OBJECTIVE The objective of this study was to investigate the protectiveness of resveratrol on cisplatin-induced damage to the ovary using experimental models. METHODS A total of 30 female Wistar-Albino rats constituted the research material. The rats were categorized into three groups: Group 1 was administered one milliliter of 0.9% NaCl solution, Group 2 was administered 7.5 mg/kg cisplatin, and Group 3 was administered 7.5 mg/kg cisplatin and 10 mg/kg resveratrol. Ovaries were extirpated in all groups and subjected to biochemical and histopathological tests. Cisplatin-induced damage to ovarian tissue was graded and scored as the total histopathological findings score. The ovarian function was assessed using immunohistochemical staining for c-kit expression. Rats' malondialdehyde, catalase, and superoxide dismutase levels were determined. RESULTS The histopathological finding score was significantly higher in Group 2 than in other groups (p<0.05). The superoxide dismutase and catalase levels were significantly higher in Group 3 than in Group 2 (p<0.001 for both cases). The malondialdehyde level was significantly higher in Group 2 than in Group 3 (p<0.001). CONCLUSION The study findings demonstrated that resveratrol reduced ovarian injury and enhanced biochemical parameters following cisplatin-induced ovary damage in experimental models.
Collapse
Affiliation(s)
- Baris Ciplak
- Malatya Training and Research Hospital, Clinic of Obstetrics and Gynecology – Malatya, Turkey
| | - Eyup Gokhan Turmus
- Yeşilyurt Hasan Çalık State Hospital, Clinic of Obstetrics and Gynecology – Malatya, Turkey
| | - Ozlem Kara
- Ahi Evran University, Faculty of Medicine, Department of Hystology and Embryology – Kırsehir, Turkey
| | - Gulcin Daglioglu
- Çukurova University, Faculty of Medicine, Department of Biochemistry – Adana, Turkey
| | - Mehmet Murat Altindag
- Malatya Training and Research Hospital, Clinic of Obstetrics and Gynecology – Malatya, Turkey
| | - Yavuz Simsek
- Biruni University, Faculty of Medicine, Department of Obstetrics and Gynecology – Istanbul, Turkey
| | - Yusuf Kenan Daglioglu
- Ahi Evran University, Faculty of Medicine, Department of Medical Microbiology – Kırsehir, Turkey
| | - Mustafa Kara
- Ahi Evran University, Faculty of Medicine, Department of Obstetrics and Gynecology – Kırşehir, Turkey
| |
Collapse
|
22
|
Zhao L, Xiong C, Yang Y, Li Z, He K, Liu Q, He Z, Luo J, Zhang X, Li Z, Yang S. The protective effect of resveratrol on largemouth bass (Micropterus salmoides) during out-of-season spawning. FISH & SHELLFISH IMMUNOLOGY 2023; 136:108688. [PMID: 36935043 DOI: 10.1016/j.fsi.2023.108688] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/05/2023] [Accepted: 03/11/2023] [Indexed: 06/18/2023]
Abstract
In aquaculture production, out-of-season spawning is beneficial to solve the seasonal shortage of fry that are normally produced once annually by species such as largemouth bass (Micropterus salmoides), thereby implementing year-round fry production. Maintaining low temperature over a period of several months can delay largemouth bass ovarian development, but it can cause severe stress to their reproductive function, leading to decreased fertility during out-of-season spawning. Feeding with antioxidants is one of the most effective methods to alleviate the negative effects of low temperature stress. Therefore, the purpose of this study is to: (a) evaluate the changes in oocyte morphology, antioxidant capacity, reproductive hormone-related index, cell apoptosis and autophagy during the out-of-season spawning of largemouth bass, and (b) to investigate the protective effect of the antioxidant resveratrol on this fish during out-of-season spawning from May through August. The study was divided into two groups (three replicates per group, 2000 fish per replicate): control group (Control) (exposure to water temperature of 12-17 °C) and resveratrol supplementation group (Res) (exposure to water temperature of 12-17 °C and fed with 200 mg/kg resveratrol). The results show that: (1) The serum hormones LH and E2 increased first and then remained unchanged, and the ovarian section showed that the ovary remained in stage IV. (2) In the process of off-season reproduction, a large number of follicles experienced follicular atresia, accompanied by endoplasmic reticulum expansion, nuclear chromatin condensation and mitochondrial swelling, which was relieved after feeding resveratrol. (3) Resveratrol decreased the ovarian ROS content and improved the activities of CAT and other antioxidant enzymes in the ovary and liver to some extent. (4) Resveratrol reduced the level of pro-apoptotic (Bax, Caspase3, Caspase8, Caspase9) and autophagy-related components (LC3-B, Beclin-1) while increasing the transcription level of anti-apoptotic (Bcl-2) factors. These findings suggest that resveratrol alleviates some adverse effects of largemouth bass during out-of-season spawning to some extent and provide a model for efficient and high-quality out-of-season spawning.
Collapse
Affiliation(s)
- Liulan Zhao
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Chen Xiong
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Yi Yang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Zhihong Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Kuo He
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Qiao Liu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Zhi He
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Jie Luo
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Xin Zhang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Zhiqiong Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Song Yang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| |
Collapse
|
23
|
Chen M, Li L, Chai Y, Yang Y, Ma S, Pu X, Chen Y. Vitamin D can ameliorate premature ovarian failure by inhibiting neutrophil extracellular traps: A review. Medicine (Baltimore) 2023; 102:e33417. [PMID: 37000081 PMCID: PMC10063315 DOI: 10.1097/md.0000000000033417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 03/07/2023] [Accepted: 03/10/2023] [Indexed: 04/01/2023] Open
Abstract
The etiology of premature ovarian failure (POF) is mainly related to inflammatory diseases, autoimmune diseases, and tumor radiotherapy and chemotherapy; however, its specific pathogenesis has not been clarified. Vitamin D (VD), a fat-soluble vitamin, is an essential steroid hormone in the human body. Neutrophil extracellular traps (NETs) are meshwork structures that are formed when neutrophils are stimulated by inflammation and other factors and are closely associated with autoimmune and inflammatory diseases. Notably, VD inhibits NET formation and intervenes in the development of POF in terms of inflammatory and immune responses, oxidative stress, and tissue fibrosis. Therefore, this study aimed to theorize the relationship between NETs, VD, and POF and provide new ideas and targets for the pathogenesis and clinical treatment of POF.
Collapse
Affiliation(s)
- Menglu Chen
- Guizhou University of Traditional Chinese Medicine, Guiyang City, Guizhou Province, China
| | - Lailai Li
- Guizhou University of Traditional Chinese Medicine, Guiyang City, Guizhou Province, China
| | - Yihui Chai
- Guizhou University of Traditional Chinese Medicine, Guiyang City, Guizhou Province, China
| | - Yuqi Yang
- Guizhou University of Traditional Chinese Medicine, Guiyang City, Guizhou Province, China
| | - Sibu Ma
- Guizhou University of Traditional Chinese Medicine, Guiyang City, Guizhou Province, China
| | - Xiang Pu
- Guizhou University of Traditional Chinese Medicine, Guiyang City, Guizhou Province, China
| | - Yunzhi Chen
- Guizhou University of Traditional Chinese Medicine, Guiyang City, Guizhou Province, China
| |
Collapse
|
24
|
Liu J, Yang Y, He Y, Feng C, Ou H, Yang J, Chen Y, You F, Shao B, Bao J, Guan X, Chen F, Zhao P. Erxian decoction alleviates cisplatin-induced premature ovarian failure in rats by reducing oxidation levels in ovarian granulosa cells. JOURNAL OF ETHNOPHARMACOLOGY 2023; 304:116046. [PMID: 36567042 DOI: 10.1016/j.jep.2022.116046] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/26/2022] [Accepted: 12/08/2022] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANT Erxian Decoction (EXD) has been used empirically for more than 70 years to treat premature ovarian failure (POF), but more research is needed to understand how it works. AIM OF THE RESEARCH The study aims to ascertain both in vivo and in vitro rewards of EXD. MATERIALS AND METHODS EXD is composed of Curculiginis Rhizoma, Epimedii Folium, Morindae Officinalis, Angelicae Sinensis, Anemarrhenae Rhizoma, and Phellodendri Chinensis Cortex. UPLC/MS analysis was used to investigate the components of EXD. Using a POF model created by administering cisplatin to rats intraperitoneally, the pharmacodynamic effects of EXD were investigated. Three dose groups of EXD were garaged into rats: high (15.6 g/kg), medium (7.8 g/kg), and low (3.9 g/kg). By using a vaginal smear, the impact of EXD on the rat estrous cycle was evaluated. An ELISA test was used to measure the anti-Mullerian hormone (AMH), estradiol (E2), follicle-stimulating hormone (FSH), and luteinizing hormone (LH) levels in the serum of rats. By using HE stains, pathological alterations in the ovaries may be seen. MDA and SOD levels in ovarian samples were used to measure the degree of ovarian oxidation. TUNEL labeling of ovarian sections was used to find apoptosis levels. By using ATP, energy production was evaluated. The relative expression of proteins connected to aging and the RAGE pathway was assessed using Western blot. Then, using H2O2, a model of senescent human ovarian granulosa cells (KGN) was created in vitro. The impact of EXD and H2O2 on cellular senescence was discovered using-galactosidase staining. Cell apoptosis levels were found using PI/Hoechest33342. By using DCFH-DA, intracellular ROS was examined. MDA and SOD concentrations were used to measure the degree of cellular oxidation. RAGE-related mRNA and protein expression were evaluated using RT-qPCR and western blotting. RESULTS Using UPLC/MS analysis, 39 chemicals in EXD were found. Rats' estrous cycles were enhanced by EXD, which increased ovarian index and follicle count and reduced the proportion of atretic follicles in the rats. EXD reduced LH and FSH output while restoring AMH and E2 secretion. In ovarian tissues, EXD reduced the amount of apoptosis and MDA while raising SOD activity and ATP levels. The protein levels of p16, p21, p53, and Lamin A/C were among the senescence-related proteins that EXD lowered, along with the levels of RAGE, PI3K, BAX, and CASPASE 3. Anti-apoptotic protein BCL-2 was also raised in the RAGE pathway. Senescence, apoptosis, ROS, and MDA levels in the KGN cells were lowered in vitro by EXD. Additionally, EXD increased the anti-apoptotic potential by changing the expression of CAT, SOD2, and SIRT1. RAGE, BAX, BCL-2, CASPASE 3, and p38 expression levels were altered by EXD, enhancing its anti-apoptotic capability. CONCLUSION EXD boosted the ovary's antioxidant and anti-apoptotic capabilities while enhancing the estrous cycle and hormone output. These findings strongly suggested that EXD may contribute to the alleviation of POF and ovarian granulosa cells senescence.
Collapse
Affiliation(s)
- Jiao Liu
- School of Life Sciences, Beijing University of Chinese Medicine, No.11 East Road, North 3rd Ring Road, Beijing, 100029, China
| | - Yang Yang
- School of Life Sciences, Beijing University of Chinese Medicine, No.11 East Road, North 3rd Ring Road, Beijing, 100029, China
| | - Yueshuang He
- School of Life Sciences, Beijing University of Chinese Medicine, No.11 East Road, North 3rd Ring Road, Beijing, 100029, China
| | - Chenran Feng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, No.11 East Road, North 3rd Ring Road, Beijing, 100029, China
| | - Haosong Ou
- School of Life Sciences, Beijing University of Chinese Medicine, No.11 East Road, North 3rd Ring Road, Beijing, 100029, China
| | - Jiadi Yang
- School of Life Sciences, Beijing University of Chinese Medicine, No.11 East Road, North 3rd Ring Road, Beijing, 100029, China
| | - Yao Chen
- School of Life Sciences, Beijing University of Chinese Medicine, No.11 East Road, North 3rd Ring Road, Beijing, 100029, China
| | - Fengming You
- School of Life Sciences, Beijing University of Chinese Medicine, No.11 East Road, North 3rd Ring Road, Beijing, 100029, China
| | - Binghao Shao
- School of Life Sciences, Beijing University of Chinese Medicine, No.11 East Road, North 3rd Ring Road, Beijing, 100029, China
| | - Jirong Bao
- School of Life Sciences, Beijing University of Chinese Medicine, No.11 East Road, North 3rd Ring Road, Beijing, 100029, China
| | - Xingyu Guan
- School of Life Sciences, Beijing University of Chinese Medicine, No.11 East Road, North 3rd Ring Road, Beijing, 100029, China
| | - Fangfang Chen
- School of Life Sciences, Beijing University of Chinese Medicine, No.11 East Road, North 3rd Ring Road, Beijing, 100029, China
| | - Piwen Zhao
- School of Life Sciences, Beijing University of Chinese Medicine, No.11 East Road, North 3rd Ring Road, Beijing, 100029, China.
| |
Collapse
|
25
|
Dinc K, Ozyurt R, Coban TA, Yazici GN, Suleyman Z, Yavuzer B, Suleyman H. The effect of carvacrol on the proinflammatory cytokines, histology, and fertility outcome of cisplatin-related ovarian change in a rat model. Taiwan J Obstet Gynecol 2023; 62:256-263. [PMID: 36965892 DOI: 10.1016/j.tjog.2022.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2022] [Indexed: 03/27/2023] Open
Abstract
OBJECTIVE In women, agents used in chemotherapy treatment have side effects such as accelerating follicular depletion and early menopause. Thus, cytotoxic treatments may cause various effects ranging from partial damage to the ovary to premature ovarian failure (POI) and infertility. This study aimed to investigate the protective effect of carvacrol on cisplatin (CIS)-induced reproductive toxicity in female rats. MATERIALS AND METHODS The animals were divided to four groups; a healthy group (HG), administered only cisplatin 2.5 mg/kg (CIS); cisplatin 2.5 mg/kg + carvacrol mg/kg (CC-50), and cisplatin 2.5 mg/kg + carvacrol 100 mg/kg (CC-100). In this study, the CC-50 and CC-100 groups were injected with carvacrol at 50 and 100 mg/kg intraperitoneally (IP). The CIS and HG groupswere administered normal saline as a solvent in the same way. One hour afterwardthe CC-50 and CC-100 groups were injected with cisplatin at 2.5 mg/kg IP. This procedure was continued once a day for 14 days. At the end of this period, six rats from each group were euthanized with high-dose anaesthesia. Biochemical (oxidant-antioxidant and proinflammatory cytokines) and histopathological examinations were performed on the right ovarian tissue removed from the dead rats. The remaining (n = 6 in each group) animals were kept in the laboratory with mature male rats for two months for breeding. Rats that didn't give birth within two months were considered infertile. A one-way ANOVA test was used for the biochemical analysis, the a Kruskal Wallis test was used for the histopathological analysis. RESULTS It has been observed that cisplatine causes oxidative stress and inflammatory damage in the ovarian tissue of animals and ultimately causes infertility due to this oxidative stress. While carvacrol significantly suppressed cisplatin-related oxidative stress in ovarian tissue at the 50 and 100 mg/kg doses, it could suppress proinflammatory cytokine increase only at thecytokine increase only at the 100 mg/kg dose. In addition, carvacrol significantly reduced the development of cisplatin-related infertility (from 0 to 83.3%) at a dose of 100 mg/kg. CONCLUSION These findings suggest that carvacrol at high doses can reduce the harmful effects of cisplatin on the ovary and improve ovarian reserve in rats.
Collapse
Affiliation(s)
- Kemal Dinc
- Department of Obstetrics and Gynecology, Faculty of Medicine, Erzincan Binali Yildirim University, Erzincan, Turkey.
| | - Ramazan Ozyurt
- Istanbul Women's Health and IVF Center, Istanbul, Turkey
| | - Taha Abdulkadir Coban
- Department of Medical Biochemistry, Faculty of Medicine, Erzincan Binali Yildirim University, Erzincan, Turkey
| | - Gulce Naz Yazici
- Department of Histology and Embryology, Faculty of Medicine, Erzincan Binali Yildirim University, Erzincan, Turkey
| | - Zeynep Suleyman
- Department of Nursing, Faculty of Health Sciences, Erzincan Binali Yildirim University, Erzincan, Turkey
| | - Bulent Yavuzer
- Department of Pharmacology, Faculty of Medicine, Erzincan Binali Yildirim University, Erzincan, Turkey
| | - Halis Suleyman
- Department of Pharmacology, Faculty of Medicine, Erzincan Binali Yildirim University, Erzincan, Turkey
| |
Collapse
|
26
|
Shahidi M, Abazari O, Dayati P, Haghiralsadat BF, Oroojalian F, Reza JZ, Naghib SM. Ginger's Antiapoptotic and Antioxidant Effects on Ovaries of Cyclophosphamide-therapied Rats. Curr Pharm Des 2023; 29:2264-2275. [PMID: 37817526 DOI: 10.2174/0113816128263943230920093609] [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: 05/24/2023] [Accepted: 07/26/2023] [Indexed: 10/12/2023]
Abstract
BACKGROUND In the recent decade, there has been increasing interest in preventing ovarian toxicity after chemotherapy exposure. It has been documented that ginger (Zingiber officinale) might normalize the hormonal balance and control the menstrual cycle.. OBJECTIVE This study has analyzed whether ginger extract protects against cyclophosphamide (CP)-induced ovarian failure in rats. METHODS Rats were distributed into four groups consisting of vehicle, CP, ginger, and CP + ginger. At the end of the treatment, all rats were killed under anesthesia to obtain ovarian tissues and blood samples for histological, molecular, and biochemical experiments. RESULTS Our results indicated that ginger improves CP-caused histological changes in ovarian tissues and significantly restores serum hormonal abnormalities. Ginger also showed unique antioxidant, anti-inflammatory, and antiapoptotic properties in the ovarian tissues of CP-induced rats. Further, our findings indicated that ginger might activate the Nrf2 and SIRT and inhibit the PI3K/AKT pathway in the ovaries of CP-treated rats. In conclusion, ginger was found to protect against CP-caused ovarian toxicity in rats. CONCLUSION The protective impacts of ginger may mediate, at least partly, by alleviating the oxidant state, inhibiting pro-inflammatory conditions, and exhibiting antiapoptotic activities.
Collapse
Affiliation(s)
- Maryamsadat Shahidi
- Department of Clinical Biochemistry, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Omid Abazari
- Department of Clinical Biochemistry, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Parisa Dayati
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Bibi Fatemeh Haghiralsadat
- Medical Nanotechnology & Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Oroojalian
- Department of Advanced Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnūrd, Iran
| | - Javad Zavar Reza
- Department of Clinical Biochemistry, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology, Tehran, Iran
- Biomaterials and Tissue Engineering Department, Breast Cancer Research Center, Motamed Cancer Institute, IUST, ACECR, Tehran, Iran
| |
Collapse
|
27
|
Mobasher MA, Hassen MT, Ebiya RA, Alturki NA, Alzamami A, Mohamed HK, Awad NS, Khodeer D, Abd El-Motelp BA. Ameliorative Effect of Citrus Lemon Peel Extract and Resveratrol on Premature Ovarian Failure Rat Model: Role of iNOS/Caspase-3 Pathway. Molecules 2022; 28:122. [PMID: 36615313 PMCID: PMC9822383 DOI: 10.3390/molecules28010122] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 12/29/2022] Open
Abstract
Premature ovarian failure (POF) is described as a loss of oocytes and the absence of folliculogenesis and is considered an adverse effect of chemotherapeutic drugs, which leads to infertility. Subsequently, the existing inquiry was achieved by exploring the potential suspicious influences of lemon peel extract (LPE), and resveratrol (RES) on cyclophosphamide (CPA) induced-POF. The results showed that CPA-induced POF significantly decreased serum estradiol (E2) and progesterone levels, along with a considerable rise in serum luteinizing hormone (LH) and follicle-stimulating hormone (FSH) levels. Moreover, CPA administration to rats significantly increased the serum level of Malondialdehyde (MDA) and significantly lowered the levels of reduced glutathione (GSH) and superoxide dismutase (SOD); in addition, it increased nuclear factor kappa B (NF-κB) levels, tumor necrosis factor-α (TNF-α), as well as cyclooxygenase 2 (COX-2) with the spread expression of inducible nitric oxide synthase (iNOS) mRNA levels and caspase-3 (Casp3) levels in ovarian tissues versus the control rats. However, treatment with LPE and RES suppressed the triggering of NF- κB pathways, evidenced by a considerable reduction in Casp3 & iNOS mRNA expression level and significant ameliorative effects in all evaluated parameters, as confirmed by the histological and immunohistochemical investigation when comparing the model group. In overall findings, both lemon peel extract and resveratrol can mitigate the adverse effects of CPA-induced POF. Most crucially, its combination therapy is a promising pharmacological agent for this disease.
Collapse
Affiliation(s)
- Maysa A. Mobasher
- Department of Pathology, Biochemistry Division, College of Medicine, Jouf University, Sakaka 72388, Saudi Arabia
| | - Marwa T. Hassen
- Department of Zoology, Faculty of Women for Arts, Science and Education, Ain Shams University, Cairo 11757, Egypt
| | - Rasha A. Ebiya
- Department of Zoology, Faculty of Women for Arts, Science and Education, Ain Shams University, Cairo 11757, Egypt
| | - Norah A. Alturki
- Clinical Laboratory Science Department, College of Applied Medical Science, King Saud University, Riyadh 11433, Saudi Arabia
| | - Ahmad Alzamami
- Clinical Laboratory Science Department, College of Applied Medical Science, Shaqra University, Al Quwaiiyah 11961, Saudi Arabia
| | - Hanaa K. Mohamed
- Department of Zoology, Faculty of Women for Arts, Science and Education, Ain Shams University, Cairo 11757, Egypt
| | - Nabil S. Awad
- Department of Genetics, Faculty of Agriculture and Natural Resources, Aswan University, Aswan 81528, Egypt
- College of Biotechnology, Misr University for Science and Technology, Giza 12563, Egypt
| | - Dina Khodeer
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Bosy A. Abd El-Motelp
- Department of Zoology, Faculty of Women for Arts, Science and Education, Ain Shams University, Cairo 11757, Egypt
| |
Collapse
|
28
|
Al-Shahat A, Hulail MAE, Soliman NMM, Khamis T, Fericean LM, Arisha AH, Moawad RS. Melatonin Mitigates Cisplatin-Induced Ovarian Dysfunction via Altering Steroidogenesis, Inflammation, Apoptosis, Oxidative Stress, and PTEN/PI3K/Akt/mTOR/AMPK Signaling Pathway in Female Rats. Pharmaceutics 2022; 14:2769. [PMID: 36559263 PMCID: PMC9786155 DOI: 10.3390/pharmaceutics14122769] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
Ovarian damage and fertility impairment are major side effects of chemotherapy in pre-menopausal cancer patients. Cisplatin is a widely used chemotherapeutic drug. The present study was designed to assess the ameliorative effects of melatonin as an adjuvant for fertility preservation. Thirty-two adult female Wistar rats were divided randomly into four equal groups: Control, Melatonin, Cisplatin (CP) treated, and CP + Melatonin treated. The cisplatin-treated group showed decreased body and ovarian weights, decreased serum E2 and AMH, increased serum LH and FSH, reduced ovarian levels of SOD, CAT, GSH, and TAC, and increased ovarian MDA. The histopathological examination of the cisplatin-treated group showed deleterious changes within ovarian tissue in the form of damaged follicles and corpus luteum, hemorrhage, and inflammatory infiltrates with faint PAS reaction in zona pellucida, increased ovarian collagen deposition, and marked expression of caspase-3 immune reaction in granulosa and theca cells, stroma, and oocytes. Alongside, there was a significant downregulation in the mRNA expression of steroidogenic enzymes, IL10, AMPK, PI3K, AKT, mTOR, and PTEN, while TGF-β1, IL1β, IL6, TNF-α, NF-Kβ, P53, p38-MAPK, JNK, and FOXO3 mRNA expressions were upregulated in cisplatin-treated rats' ovarian tissue. Coadministration of cisplatin-treated rats with melatonin reversed these changes significantly. In conclusion, melatonin's antioxidant, anti-inflammatory, and anti-apoptotic activities could modulate ovarian disturbances induced by cisplatin and preserve fertility.
Collapse
Affiliation(s)
- Amal Al-Shahat
- Human Anatomy & Embryology Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Mohey A. E. Hulail
- Human Anatomy & Embryology Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Nada M. M. Soliman
- Human Anatomy & Embryology Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Tarek Khamis
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
- Laboratory of Biotechnology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Liana Mihaela Fericean
- Biology Department, Faculty of Agriculture, University of Life Sciences “King Michael I of Romania” from Timisoara, Aradului St. 119, 300645 Timisoara, Romania
| | - Ahmed Hamed Arisha
- Department of Animal Physiology and Biochemistry, Faculty of Veterinary Medicine, Badr University in Cairo (BUC), Badr City 11829, Egypt
- Department of Physiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Rania S. Moawad
- Human Anatomy & Embryology Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
29
|
Abd-Eltawab Tammam A, A. Khalaf AA, R. Zaki A, Mansour Khalifa M, A. Ibrahim M, M. Mekkawy A, E. Abdelrahman R, Farghali A, A. Noshy P. Hesperidin protects rats’ liver and kidney from oxidative damage and physiological disruption induced by nickel oxide nanoparticles. Front Physiol 2022; 13:912625. [PMID: 36338490 PMCID: PMC9626958 DOI: 10.3389/fphys.2022.912625] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 09/22/2022] [Indexed: 11/25/2022] Open
Abstract
Background: Nickel oxide nanoparticles (NiO-NPs) have recently been utilized in various advanced industrial fields like lithium-ion micro batteries, nanofibers, electrochromic devices, and several biomedical applications. NiO-NPs are classified as extremely toxic substances as they can cause long-term harm to the environment and aquatic life. Moreover, frequent and prolonged exposure can affect human and animal health, causing skin allergies and major toxic consequences, such as hepatorenal toxicity. Hesperidin (HSP) has been proven to possess anti-inflammatory, antioxidant, and free radical scavenging activities. Objective: This study aimed to investigate the underlying protective mechanisms and effects of HSP against NiO-NPs-induced hepatorenal toxicities in rats. Materials and Methods: Forty male Wistar rats were randomly divided into four groups (n = 10 in each). The first group served as a Control group. For 8 weeks, the second group was administered NiO-NPs (100 mg/kg/day), and the third group was given HSP (100 mg/kg/day) via oral gavage for both groups. The fourth group received NiO-NPs and HSP concurrently in the same oral daily doses and duration as the second and third groups. Results: NiO-NPs administration revealed a significant increase in plasma biomarkers of nephrotoxicity (urea, creatinine) and hepatotoxicity (ALT, AST) in NiO-NPs group compared to Control group (p < 0.05). In addition, NiO-NPs administration resulted in a substantial increase in malondialdehyde levels with a significant drop in catalase activity and GSH content in Group II. Also, a significant decreased expression of Nrf-2 and Bcl-2 mRNA levels and upregulation of TNF-α, NF-kβ and BAX in the liver and kidney of NiO-NPs group were also detected. Histologically, the liver and kidney of rats of NiO-NPs group showed significant histopathological disturbances, with a substantial increase in the proliferating cell nuclear antigen (PCNA) positive hepatocytes and renal tubular cells in the NiO-NPs group compared to Control and HSP groups (p < 0.05). In contrast, concomitant administration of HSP with NiO-NPs in group IV showed a significant biochemical, histopathological, and immunohistochemical improvement compared to NiO-NPs group. Conclusion: Co-administration of HSP with NiO-NPs significantly ameliorated most of the NiO-NPs-induced hepatorenal toxicities in male rats.
Collapse
Affiliation(s)
- Ahmed Abd-Eltawab Tammam
- Medical Physiology Department, College of Medicine, Jouf University, Sakaka, Saudi Arabia
- Medical Physiology Department, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
- *Correspondence: Ahmed Abd-Eltawab Tammam,
| | - Abdel Azeim A. Khalaf
- Department of Toxicology and Forensic Medicine, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Amr R. Zaki
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Mohamed Mansour Khalifa
- Department of Human Physiology, Faculty of Medicine, Cairo University, Cairo, Egypt
- Department of Human Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Marwa A. Ibrahim
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Aya M. Mekkawy
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Rehab E. Abdelrahman
- Department of Toxicology and Forensic Medicine, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Ahmed Farghali
- Materials Science and Nanotechnology Department, Faculty of Postgraduate Studies for Advanced Sciences, Beni-Suef University, Beni-Suef, Egypt
| | - Peter A. Noshy
- Department of Toxicology and Forensic Medicine, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
30
|
Ibrahim MA, Khalifa AM, Mohamed AA, Galhom RA, Korayem HE, Abd El-Fadeal NM, Abd-Eltawab Tammam A, Khalifa MM, Elserafy OS, Abdel-Karim RI. Bone-Marrow-Derived Mesenchymal Stem Cells, Their Conditioned Media, and Olive Leaf Extract Protect against Cisplatin-Induced Toxicity by Alleviating Oxidative Stress, Inflammation, and Apoptosis in Rats. TOXICS 2022; 10:toxics10090526. [PMID: 36136492 PMCID: PMC9504158 DOI: 10.3390/toxics10090526] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/26/2022] [Accepted: 09/01/2022] [Indexed: 06/01/2023]
Abstract
BACKGROUND Hepatic and renal damage is a cisplatin (Cis)-induced deleterious effect that is a major limiting factor in clinical chemotherapy. OBJECTIVES The current study was designed to investigate the influence of pretreatment with olive leaf extract (OLE), bone-marrow-derived mesenchymal stem cells (BM-MSC), and their conditioned media (CM-MSC) against genotoxicity, nephrotoxicity, hepatotoxicity, and immunotoxicity induced by cisplatin in rats. METHODS The rats were randomly divided into six groups (six rats each) as follows: Control; OLE group, treated with OLE; Cis group, treated with a single intraperitoneal dose of Cis (7 mg/kg bw); Cis + OLE group, treated with OLE and cisplatin; Cis + CM-MSC group, treated with BM-MSC conditioned media and Cis; and Cis + MSC group, treated with BM-MSC in addition to Cis. RESULTS Cis resulted in a significant deterioration in hepatic and renal functions and histological structures. Furthermore, it increased inflammatory markers (TNF-α, IL-6, and IL-1β) and malondialdehyde (MDA) levels and decreased glutathione (GSH) content, total antioxidant capacity (TAC), catalase (CAT), and superoxide dismutase (SOD) activity in hepatic and renal tissues. Furthermore, apoptosis was evident in rat tissues. A significant increase in serum 8-hydroxy-2-deoxyguanosine (8-OH-dG), nitric oxide (NO) and lactate dehydrogenase (LDH), and a decrease in lysozyme activity were detected in Cis-treated rats. OLE, CM-MSC, and BM-MSC have significantly ameliorated Cis-induced deterioration in hepatic and renal structure and function and improved oxidative stress and inflammatory markers, with preference to BM-MSC. Moreover, apoptosis was significantly inhibited, evident from the decreased expression of Bax and caspase-3 genes and upregulation of Bcl-2 proteins in protective groups as compared to Cis group. CONCLUSIONS These findings indicate that BM-MSC, CM-MSC, and OLE have beneficial effects in ameliorating cisplatin-induced oxidative stress, inflammation, and apoptosis in the hepatotoxicity, nephrotoxicity, immunotoxicity, and genotoxicity in a rat model.
Collapse
Affiliation(s)
- Mahrous A. Ibrahim
- Forensic Medicine and Clinical Toxicology, College of Medicine, Jouf University, Sakaka 41412, Saudi Arabia
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Suez Canal University (SCU), Ismailia 41522, Egypt or
| | - Athar M. Khalifa
- Pathology Department, College of Medicine, Jouf University, Sakaka 41412, Saudi Arabia
| | - Alaa A. Mohamed
- Medical Biochemistry Division, Pathology Department, College of Medicine, Jouf University, Sakaka 41412, Saudi Arabia
- Medical Biochemistry Department, Faculty of Medicine, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Rania A. Galhom
- Human Anatomy and Embryology Department, Faculty of Medicine, Suez Canal University (SCU), Ismailia 41522, Egypt
- Center of Excellence in Molecular and Cellular Medicine (CEMCM), Faculty of Medicine, Suez Canal University (SCU), Ismailia 41522, Egypt
- Human Anatomy and Embryology Department, Faculty of Medicine, Badr University in Cairo (BUC), Cairo 11829, Egypt
| | - Horeya E. Korayem
- Histology and Cell Biology Department, Faculty of Medicine, Suez Canal University (SCU), Ismailia 41522, Egypt
| | - Noha M. Abd El-Fadeal
- Center of Excellence in Molecular and Cellular Medicine (CEMCM), Faculty of Medicine, Suez Canal University (SCU), Ismailia 41522, Egypt
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Suez Canal University (SCU), Ismailia 41522, Egypt
- Oncology Diagnostic Unit, Faculty of Medicine, Suez Canal University (SCU), Ismailia 41522, Egypt
| | - Ahmed Abd-Eltawab Tammam
- Physiology Department, College of Medicine, Jouf University, Sakaka 41412, Saudi Arabia
- Physiology Department, Faculty of Medicine, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Mohamed Mansour Khalifa
- Human Physiology Department, Faculty of Medicine, Cairo University, Cairo 11562, Egypt
- Human Physiology Department, College of Medicine, King Saud University, Riyadh 11472, Saudi Arabia
| | - Osama S. Elserafy
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Cairo University, Cairo 11562, Egypt
- Criminal Justice and Forensic Sciences Department, King Fahd Security College, Riyadh 11451, Saudi Arabia
| | - Rehab I. Abdel-Karim
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Suez Canal University (SCU), Ismailia 41522, Egypt or
| |
Collapse
|
31
|
Yifei sanjie Pills Alleviate Chemotherapy-Related Fatigue by Reducing Skeletal Muscle Injury and Inhibiting Tumor Growth in Lung Cancer Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:2357616. [PMID: 36045663 PMCID: PMC9423986 DOI: 10.1155/2022/2357616] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/02/2022] [Indexed: 11/18/2022]
Abstract
Chemotherapy-related fatigue (CRF), one of the most severe adverse effects observed in cancer patients, has been theoretically related to oxidative stress, and antioxidant treatment might be one of the most valuable therapeutic approaches. However, there are still few effective pharmacological therapies. Yifei Sanjie pills (YFSJ), a classical formula used to treat lung cancer as complementary and alternative medicine, have been proved to alleviate CRF of lung cancer patients in clinical practices. However, the underlying mechanisms have not been clarified. In this study, our data showed that YFSJ alleviated CRF presented as reversing the decline of swimming time and locomotor activity induced by cisplatin (DDP). Moreover, YFSJ significantly reduces the accidence of mitophagy and mitochondrial damage and reduces apoptosis in skeletal muscle tissues caused by DDP. It probably works by decreasing the oxidative stress, inhibiting the activation of the AMPK/mTOR pathway, decreasing protein expression levels of Beclin1 and other autophagy-related proteins, and attenuating the activation of Cytochrome c (cyto. C), Cleaved Caspase-9 (c-Casp 9), and other apoptosis-related proteins. Furthermore, YFSJ enhanced DDP sensitivity by specifically promoting oxidative stress and activating apoptosis and autophagy in the tumor tissues of mice. It was also found that YFSJ reduced the loss of body weight caused by DDP, reversed the ascent of serum concentrations of alanine aminotransferase (ALT), aminotransferase (AST), and creatinine (CREA), increased the spleen index, and prolonged the survival time of mice. Taken together, these results revealed that YFSJ could alleviate CRF by reducing mitophagy and apoptosis induced by oxidative stress in skeletal muscle; these results also displayed the effects of YFSJ on enhancing chemotherapy sensitivity, improving quality of life, and prolonging survival time in lung cancer mice received DDP chemotherapy.
Collapse
|
32
|
Wang J, Jia R, Celi P, Zhuo Y, Ding X, Zeng Q, Bai S, Xu S, Yin H, Lv L, Zhang K. Resveratrol Alleviating the Ovarian Function Under Oxidative Stress by Alternating Microbiota Related Tryptophan-Kynurenine Pathway. Front Immunol 2022; 13:911381. [PMID: 35911670 PMCID: PMC9327787 DOI: 10.3389/fimmu.2022.911381] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 06/07/2022] [Indexed: 12/12/2022] Open
Abstract
Oxidative stress (OS) is a key factor regulating the systemic pathophysiological effects and one of the fundamental mechanisms associated with aging and fertility deterioration. Previous studies revealed that resveratrol (RV) exhibits a preventive effect against oxidative stress in the ovary. However, it remains unknown whether gut microbiota respond to resveratrol during an OS challenge. In Exp. 1, layers received intraperitoneal injection of tert-butyl hydroperoxide (tBHP) (0 or 800 μmol/kg BW) or received resveratrol diets (0 or 600 mg/kg) for 28 days. In Exp. 2, the role of intestinal microbiota on the effects of resveratrol on tBHP-induced oxidative stress was assessed through fecal microbiota transplantation (FMT). The OS challenge reduced the egg-laying rate and exhibited lower pre-hierarchical follicles and higher atretic follicles. Oral RV supplementation ameliorated the egg-laying rate reduction and gut microbiota dysbiosis. RV also reversed the tryptphan-kynurenine pathway, upregulated nuclear factor E2-related factor 2 (Nrf2) and silent information regulator 1(SIRT1) levels, and decreased the expression of forkhead box O1 (FoxO1) and P53. These findings indicated that the intestinal microbiota-related tryptophan-kynurenine pathway is involved in the resveratrol-induced amelioration of ovary oxidative stress induced by tBHP in the layer model, while SIRT1-P53/FoxO1 and Nrf2-ARE signaling pathway were involved in this process.
Collapse
Affiliation(s)
- Jianping Wang
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
- *Correspondence: Jianping Wang,
| | - Ru Jia
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Pietro Celi
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Yong Zhuo
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Xuemei Ding
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Qiufeng Zeng
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Shiping Bai
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Shengyu Xu
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Huadong Yin
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Li Lv
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Keying Zhang
- Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
33
|
Resveratrol as a modulatory of apoptosis and autophagy in cancer therapy. Clin Transl Oncol 2022; 24:1219-1230. [PMID: 35038152 DOI: 10.1007/s12094-021-02770-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/24/2021] [Indexed: 12/24/2022]
Abstract
Cancer is one of the leading causes of death, with a heavy socio-economical burden for countries. Despite the great advances that have been made in the treatment of cancer, chemotherapy is still the most common method of treatment. However, many side effects, including hepatotoxicity, renal toxicity, and cardiotoxicity, limit the efficacy of conventional chemotherapy. Over recent years, natural products have attracted attention as therapeutic agents against various diseases, such as cancer. Resveratrol (RES), a natural polyphenol occurring in grapes, nuts, wine, and berries, exhibited potential for preventing and treating various cancer types. RES also ameliorates chemotherapy-induced detrimental effects. Furthermore, RES could modulate apoptosis and autophagy as the main forms of cancer cell deaths by targeting various signaling pathways and up/downregulation of apoptotic and autophagic genes. This review will summarize the anti-cancer effects of RES and focus on the fundamental mechanisms and targets for modulating apoptosis and autophagy by RES.
Collapse
|
34
|
Domingo IK, Latif A, Bhavsar AP. Pro-Inflammatory Signalling PRRopels Cisplatin-Induced Toxicity. Int J Mol Sci 2022; 23:7227. [PMID: 35806229 PMCID: PMC9266867 DOI: 10.3390/ijms23137227] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 02/04/2023] Open
Abstract
Cisplatin is a platinum-based chemotherapeutic that has long since been effective against a variety of solid-cancers, substantially improving the five-year survival rates for cancer patients. Its use has also historically been limited by its adverse drug reactions, or cisplatin-induced toxicities (CITs). Of these reactions, cisplatin-induced nephrotoxicity (CIN), cisplatin-induced peripheral neuropathy (CIPN), and cisplatin-induced ototoxicity (CIO) are the three most common of several CITs recognised thus far. While the anti-cancer activity of cisplatin is well understood, the mechanisms driving its toxicities have only begun to be defined. Most of the literature pertains to damage caused by oxidative stress that occurs downstream of cisplatin treatment, but recent evidence suggests that the instigator of CIT development is inflammation. Cisplatin has been shown to induce pro-inflammatory signalling in CIN, CIPN, and CIO, all of which are associated with persisting markers of inflammation, particularly from the innate immune system. This review covered the hallmarks of inflammation common and distinct between different CITs, the role of innate immune components in development of CITs, as well as current treatments targeting pro-inflammatory signalling pathways to conserve the use of cisplatin in chemotherapy and improve long-term health outcomes of cancer patients.
Collapse
Affiliation(s)
| | | | - Amit P. Bhavsar
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2E1, Canada; (I.K.D.); (A.L.)
| |
Collapse
|
35
|
Barberino RS, Silva RLS, Palheta Junior RC, Smitz JEJ, Matos MHT. Protective Effects of Antioxidants on Cyclophosphamide-Induced Ovarian Toxicity. Biopreserv Biobank 2022; 21:121-141. [PMID: 35696235 DOI: 10.1089/bio.2021.0159] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The most common limitation of anticancer chemotherapy is the injury to normal cells. Cyclophosphamide, which is one of the most widely used alkylating agents, can cause premature ovarian insufficiency and infertility since the ovarian follicles are extremely sensitive to their effects. Although little information is available about the pathogenic mechanism of cyclophosphamide-induced ovarian damage, its toxicity is attributed to oxidative stress, inflammation, and apoptosis. The use of compounds with antioxidant and cytoprotective properties to protect ovarian function from deleterious effects during chemotherapy would be a significant advantage. Thus, this article reviews the mechanism by which cyclophosphamide exerts its toxic effects on the different cellular components of the ovary, and describes 24 cytoprotective compounds used to ameliorate cyclophosphamide-induced ovarian injury and their possible mechanisms of action. Understanding these mechanisms is essential for the development of efficient and targeted pharmacological complementary therapies that could protect and prolong female fertility.
Collapse
Affiliation(s)
- Ricássio S Barberino
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Department of Veterinary Medicine, Federal University of São Francisco Valley-UNIVASF, Petrolina, Brazil
| | - Regina Lucia S Silva
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Department of Veterinary Medicine, Federal University of São Francisco Valley-UNIVASF, Petrolina, Brazil
| | - Raimundo C Palheta Junior
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Federal University of São Francisco Valley-UNIVASF, Petrolina, Brazil
| | - Johan E J Smitz
- Follicle Biology Laboratory, Center for Reproductive Medicine, Free University Brussels-VUB, Brussels, Belgium
| | - Maria Helena T Matos
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Department of Veterinary Medicine, Federal University of São Francisco Valley-UNIVASF, Petrolina, Brazil
| |
Collapse
|
36
|
Jiao JH, Gao L, Yong WL, Kou ZY, Ren ZQ, Cai R, Chu GY, Pang WJ. Resveratrol improves estrus disorder induced by bisphenol A through attenuating oxidative stress, autophagy, and apoptosis. J Biochem Mol Toxicol 2022; 36:e23120. [PMID: 35670589 DOI: 10.1002/jbt.23120] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 04/26/2022] [Accepted: 05/28/2022] [Indexed: 11/08/2022]
Abstract
Bisphenol A (BPA), as a widely used plasticizer, is easily absorbed by animals and humans. It has certain toxic effects on various tissues, including liver, heart, kidney, testis, and ovary. The toxic effects of BPA on animal reproduction have aroused widespread concern, but its regulatory mechanism and antidote in female animals estrus cycle remain unclear. In this study, the results displayed that BPA destroyed the normal estrus cycle of mice through decreasing the levels of progesterone and estradiol. Furthermore, BPA significantly increased the levels of oxidative stress, autophagy, and apoptosis in ovaries and granulosa cells. Interestingly, we found that the natural antioxidant resveratrol rescued estrus disorder and impaired estradiol secretion, reduced the abnormal reactive oxygen species accumulation, autophagy, and apoptosis in BPA exposed ovarian tissues. Moreover, transmission electron microscopy showed that resveratrol reduced BPA-induced autophagic vesicles formation and flow cytometry showed that resveratrol inhibited the increase of apoptotic cells induced by BPA on granulosa cells. Therefore, the supplement of resveratrol could restore BPA-induced estrus disorder by protecting ovarian granulosa cells. Overall, resveratrol is a potential drug to alleviate BPA-induced estrous cycle disorders and ovarian damage.
Collapse
Affiliation(s)
- Jun-Heng Jiao
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling Shaanxi, China
| | - Lei Gao
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling Shaanxi, China
| | - Wen-Long Yong
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling Shaanxi, China
| | - Zhong-Yun Kou
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling Shaanxi, China
| | - Zhi-Qiang Ren
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling Shaanxi, China
| | - Rui Cai
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling Shaanxi, China
| | - Gui-Yan Chu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling Shaanxi, China
| | - Wei-Jun Pang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling Shaanxi, China
| |
Collapse
|
37
|
Meng T, Deng J, Xiao D, Arowolo MA, Liu C, Chen L, Deng W, He S, He J. Protective Effects and Potential Mechanisms of Dietary Resveratrol Supplementation on the Spleen of Broilers Under Heat Stress. Front Nutr 2022; 9:821272. [PMID: 35651504 PMCID: PMC9150503 DOI: 10.3389/fnut.2022.821272] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Resveratrol (RSV) is a natural polyphenolic compound with potent antioxidant and anti-inflammatory properties. This study aimed to investigate the protective effects of RSV supplementation on the inflammatory responses of broilers during heat stress. A total of 432 28-d-old white-feathered broilers (817 crossbred chicken) with an average weight of 549 ± 4 g were randomly allotted to 4 equal groups (Half male and half female, 6 replicates/group, 18 chickens/replicate), including normal temperature (NT) group (24 ± 2°C for 24 h/d, basal diet), NT+RSV group (24 ± 2°C for 24 h/d, basal diet + RSV), heat stress (HT) group (37 ± 2°C for 8 h/d, basal diet), and HT+RSV group (37 ± 2°C for 8 h/d, basal diet + RSV). Serum samples were collected on d 7 and 14 of heat stress, and thymus, spleen, jejunum, and bursa of Fabricius samples were collected and analyzed on d14. RSV treatment decreased the feed conversion ratio, partially reversed the negative alternations in body weight, average daily gain, and average daily feed intake caused by heat stress. RSV treatment also decreased the elevated levels of corticosterone on d 14, adrenocorticotropic hormone, and triiodothyronine in serum on d 7 caused by heat stress, and significantly increased the villus height to crypt depth ratio in the jejunum on d 14. Dietary RSV also reduced heat stress-induced splenic pro-inflammatory cytokine concentrations. TUNEL assay showed that RSV significantly reduced heat stress-induced the number of apoptotic cells. Remarkably, RSV down-regulated some splenic related genes for apoptosis genes, including BCL-2, Apaf-1, and MDM2 mRNA levels induced by heat stress. According to GO and KEGG enrichment analyses, the differential genes between HT and HT + RSV groups were mainly associated with immune system process, hematopoietic or lymphoid organ development, and toll-like receptor signaling pathway. The relative mRNA expression of NF-κB, heat shock protein 70 (HSP70), and p38 MAPK were markedly decreased by the combination of RSV and heat stress. These findings showed that RSV might reduce the splenic inflammatory response in heat-stressed white-feather broilers by inhibiting heat stress-induced activation of NF-B, MAPK, and HSP70, as well as inhibiting the activation of mitochondrial apoptotic pathways.
Collapse
Affiliation(s)
- Tiantian Meng
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Juying Deng
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Dingfu Xiao
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | | | - Chunming Liu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Liang Chen
- Institute of Animal Husbandry and Aquaculture, Huaihua Academy of Agricultural Sciences, Huaihua, China
| | - Wei Deng
- Institute of Animal Husbandry and Aquaculture, Huaihua Academy of Agricultural Sciences, Huaihua, China
| | - Shaoping He
- College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jianhua He
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China.,Institute of Animal Husbandry and Aquaculture, Huaihua Academy of Agricultural Sciences, Huaihua, China
| |
Collapse
|
38
|
Li Q, Zhao T, He H, Robert N, Ding T, Hu X, Zhang T, Pan Y, Cui Y, Yu S. Ascorbic acid protects the toxic effects of aflatoxin B 1 on yak oocyte maturation. Anim Sci J 2022; 93:e13702. [PMID: 35257449 DOI: 10.1111/asj.13702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 01/26/2022] [Accepted: 02/03/2022] [Indexed: 11/29/2022]
Abstract
High-quality oocytes are a prerequisite for successful fertilization. Mammals feeding on aflatoxin-contaminated feed can cause reproductive toxicity, including follicular atresia, poor oocyte development and maturation, and aberrant epigenetic modifications of oocytes. In addition, the important role of ascorbic acid (AA) in reproductive biology has been confirmed, and AA is widely used as an antioxidant in cell culture. However, the toxic effects of aflatoxin B1 (AFB1 ) on yak oocytes and whether AA has protective effects remain unknown. In this study, we found that exposure to AFB1 impedes meiotic maturation of oocytes, promotes apoptosis by triggering high levels of reactive oxygen species (ROS), and disrupts mitochondrial distribution and actin integrity, resulting in a decrease in the fertilization ability and parthenogenetic development ability of oocytes. In addition, these injuries changed the DNA methylation transferase transcription level of mature oocytes. After adding 50 μg/ml AA, the indices recovered to levels close to those of the control group. The results showed that AA could protect yak oocytes from the toxic effects of AFB1 and improve the quality of oocytes.
Collapse
Affiliation(s)
- Qin Li
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Tian Zhao
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Honghong He
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Niayale Robert
- Laboratory of Animal Anatomy & Tissue Embryology, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Tianyi Ding
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Xuequan Hu
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Tongxiang Zhang
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yangyang Pan
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yan Cui
- Laboratory of Animal Anatomy & Tissue Embryology, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Sijiu Yu
- Gansu Province Livestock Embryo Engineering Research Center, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| |
Collapse
|
39
|
Luo X, Xu J, Zhao R, Qin J, Wang X, Yan Y, Wang LJ, Wang G, Yang X. The Role of Inactivated NF-κB in Premature Ovarian Failure. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:468-483. [PMID: 34971586 DOI: 10.1016/j.ajpath.2021.12.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/25/2021] [Accepted: 12/02/2021] [Indexed: 06/14/2023]
Abstract
Premature ovarian failure (POF) is defined as deployment of amenorrhea due to the cessation of ovarian function in a woman younger than 40 years old. The pathologic mechanism of POF is not yet well understood, although genetic aberrations, autoimmune damage, and environmental factors have been identified. The current study demonstrated that NF-κB inactivation is closely associated with the development of POF based on the data from literature and cyclophosphamide (Cytoxan)-induced POF mouse model. In the successfully established NF-κB-inactivated mouse model, the results showed the reduced expression of nuclear p65 and the increased expression of IκBα in ovarian granulosa cells; the reduced numbers of antral follicles; the reduction of Ki-67/proliferating cell nuclear antigen-labeled cell proliferation and enhanced Fas/FasL-dependent apoptosis in granulosa cells; the reduced level of E2 and anti-Müllerian hormone; the decreased expression of follicle-stimulating hormone receptor and cytochrome P450 family 19 subfamily A member 1 (CYP19A1) in granulosa cells, which was reversed in the context of blocking NF-κB signaling with BAY 11-7082; and the decreased expressions of glucose-regulated protein 78 (GRP78), activating transcription factor 6, protein kinase R-like endoplasmic reticulum kinase, and inositol-requiring enzyme 1 in granulosa cells. Dual-luciferase reporter assay demonstrated that p50 stimulated the transcription of GRP78, and NF-κB affected the expression of follicle-stimulating hormone receptor and promoted granulosa cell proliferation through GRP78-mediated endoplasmic reticulum stress. Taken together, these data indicate, for the first time, that the inactivation of NF-κB signaling plays an important role in POF.
Collapse
Affiliation(s)
- Xin Luo
- International Joint Laboratory for Embryonic Development and Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, China
| | - Junjie Xu
- International Joint Laboratory for Embryonic Development and Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, China
| | - Ran Zhao
- International Joint Laboratory for Embryonic Development and Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, China
| | - Jiajia Qin
- Gynecology, Chinese Medicine College, Jinan University, Guangzhou, China
| | - Xiaoyu Wang
- International Joint Laboratory for Embryonic Development and Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, China
| | - Yu Yan
- International Joint Laboratory for Embryonic Development and Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, China
| | - Li-Jing Wang
- Institute of Vascular Biological Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Guang Wang
- International Joint Laboratory for Embryonic Development and Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, China; Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, China; Guangdong-Hong Kong Metabolism and Reproduction Joint Laboratory, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, China.
| | - Xuesong Yang
- International Joint Laboratory for Embryonic Development and Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, China; Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, China; Guangdong-Hong Kong Metabolism and Reproduction Joint Laboratory, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, China.
| |
Collapse
|
40
|
Interactions of Analgesics with Cisplatin: Modulation of Anticancer Efficacy and Potential Organ Toxicity. MEDICINA (KAUNAS, LITHUANIA) 2021; 58:medicina58010046. [PMID: 35056355 PMCID: PMC8781901 DOI: 10.3390/medicina58010046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/19/2021] [Accepted: 12/20/2021] [Indexed: 12/17/2022]
Abstract
Cisplatin (CDDP), one of the most eminent cancer chemotherapeutic agents, has been successfully used to treat more than half of all known cancers worldwide. Despite its effectiveness, CDDP might cause severe toxic adverse effects on multiple body organs during cancer chemotherapy, including the kidneys, heart, liver, gastrointestinal tract, and auditory system, as well as peripheral nerves causing severely painful neuropathy. The latter, among other pains patients feel during chemotherapy, is an indication for the use of analgesics during treatment with CDDP. Different types of analgesics, such as acetaminophen, non-steroidal anti-inflammatory drugs (NSAIDS), and narcotic analgesics, could be used according to the severity of pain. Administered analgesics might modulate CDDP’s efficacy as an anticancer drug. NSAIDS, on one hand, might have cytotoxic effects on their own and few of them can potentiate CDDP’s anticancer effects via inhibiting the CDDP-induced cyclooxygenase (COX) enzyme, or through COX-independent mechanisms. On the other hand, some narcotic analgesics might ameliorate CDDP’s anti-neoplastic effects, causing chemotherapy to fail. Concerning safety, some analgesics share the same adverse effects on normal tissues as CDDP, augmenting its potentially hazardous effects on organ impairment. This article offers an overview of the reported literature on the interactions between analgesics and CDDP, paying special attention to possible mechanisms that modulate CDDP’s cytotoxic efficacy and potential adverse reactions.
Collapse
|
41
|
Koh YC, Ho CT, Pan MH. Recent Advances in Health Benefits of Stilbenoids. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:10036-10057. [PMID: 34460268 DOI: 10.1021/acs.jafc.1c03699] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Biological targeting or molecular targeting is the main strategy in drug development and disease prevention. However, the problem of "off-targets" cannot be neglected. Naturally derived drugs are preferred over synthetic compounds in pharmaceutical markets, and the main goals are high effectiveness, lower cost, and fewer side effects. Single-target drug binding may be the major cause of failure, as the pathogenesis of diseases is predominantly multifactorial. Naturally derived drugs are advantageous because they are expected to have multitarget effects, but not off-targets, in disease prevention or therapeutic actions. The capability of phytochemicals to modulate molecular signals in numerous diseases has been widely discussed. Among them, stilbenoids, especially resveratrol, have been well-studied, along with their potential molecular targets, including AMPK, Sirt1, NF-κB, PKC, Nrf2, and PPARs. The analogues of resveratrol, pterostilbene, and hydroxylated-pterostilbene may have similar, if not more, potential biological targeting effects compared with their original counterpart. Furthermore, new targets that have been discussed in recent studies are reviewed in this paper.
Collapse
Affiliation(s)
- Yen-Chun Koh
- Institute of Food Sciences and Technology, National Taiwan University, Taipei 10617, Taiwan
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, New Jersey 08901, United States
| | - Min-Hsiung Pan
- Institute of Food Sciences and Technology, National Taiwan University, Taipei 10617, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung City, Taiwan 404
- Department of Health and Nutrition Biotechnology, Asia University, Taichung City, Taiwan 413
| |
Collapse
|
42
|
Xia T, Li J, Ren X, Liu C, Sun C. Research progress of phenolic compounds regulating IL-6 to exert antitumor effects. Phytother Res 2021; 35:6720-6734. [PMID: 34427003 DOI: 10.1002/ptr.7258] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 08/08/2021] [Accepted: 08/08/2021] [Indexed: 02/06/2023]
Abstract
Cytokine therapy, which activates the host immune system, has become an important and novel therapeutic approach to treat various cancers. Recent studies have shown that IL-6 is an important cytokine that regulates the homeostasis in vivo. However, excessive IL-6 plays a pathological role in a variety of acute and chronic inflammatory diseases, especially in cancer. IL-6 can transmit signals through JAK/STAT, RAS /MAPK, PI3K/ Akt, NF-κB, and other pathways to promote cancer progression. Phenolic compounds can effectively regulate the level of IL-6 in tumor cells and improve the tumor microenvironment. This article focuses on the phenolic compounds through the regulation of IL-6, participate in the prevention of cancer, inhibit the proliferation of cancer cells, reduce angiogenesis, improve therapeutic efficacy, and reduce side effects and other aspects. This will help to further advance research on cytokine therapy to reduce the burden of cancer and improve patient prognosis. However, current studies are mostly limited to animal and cellular experiments, and high-quality clinical studies are needed to further determine their antitumor efficacy in humans.
Collapse
Affiliation(s)
- Tingting Xia
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jie Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xin Ren
- Clinical Medical Colleges, Weifang Medical University, Weifang, China
| | - Cun Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Changgang Sun
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China.,Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, China
| |
Collapse
|