1
|
Luo H, Yang S, Deng P, Peng Y, Chen Z, Yang C, Wang M, Qin R, Yuan L, Chen X, Wang D, Huang X, Wang J. Network pharmacology combined with transcriptomics reveals that formononetin, a biologically component of Astragalus membranaceus (Fisch.) Bunge, inhibits the PI3K/AKT signaling pathway to improve chronic renal failure. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119041. [PMID: 39510423 DOI: 10.1016/j.jep.2024.119041] [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: 08/16/2024] [Revised: 10/27/2024] [Accepted: 11/05/2024] [Indexed: 11/15/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Formononetin (FMN), one of the main isoflavones isolated from Astragalus membranaceus (Fisch.) Bunge, has multiple pharmacological and renal-protective effects. Our previous study suggested FMN as a candidate compound for the treatment of chronic renal failure (CRF). However, the mechanism underlying the repressive effect of FMN on the development of CRF is still unknown. AIMS OF THE STUDY To investigate the protective effect of FMN on CRF using in vivo and in vitro models and elucidate the potential underlying mechanism. MATERIALS AND METHODS An in vivo model of adenine-induced CRF and an in vitro model of human proximal tubule epithelial cells (HK-2) stimulated with transforming growth factor (TGF)-β1 were used. Serum levels of renal function parameters and inflammatory cytokines were evaluated. Histological analysis was performed to determine the extent of renal injury and fibrosis. Network pharmacology and mRNA sequencing were used to explore the potential mechanism. PPI analysis and molecular docking were used to identify key targets. Polymerase chain reaction and western blotting were used to determine the mechanism underlying the effect of FMN on CRF. RESULTS FMN decreased the levels of renal function biochemical markers, including serum creatinine, blood urea nitrogen, and 24 h urine protein content. Treatment with FMN improved renal tubule injury and extracellular matrix (ECM) components, including collagens I and III. In addition, FMN significantly inhibited epithelial-mesenchymal transition (EMT); decreased the expression of fibronectin, N-cadherin, vimentin, α-SMA, and TGF-β1; and restored the expression of E-cadherin. The effect of FMN on renal interstitial fibrosis contributed to decreasing the expression of PI3K, p-Akt, and interleukin (IL) 4, restoring the expression of nitric oxide synthase 3 (NOS3), and reducing the release of inflammatory cytokines (IL-1β, IL-6, and tumor necrosis factor-alpha), both in vivo and in vitro. FMN treatment improved renal function and deposition of ECM components, reduced protein levels of EMT markers in rat kidneys and HK-2 cells, decreased the release of inflammatory cytokines, and inhibited the PI3K/Akt signaling pathway. CONCLUSIONS FMN treatment significantly reduced the release of inflammatory cytokines and inhibited the effects of the PI3K/Akt signaling pathway on the key targets IL-4 and NOS3. Our results suggest FMN therapy as a novel therapeutic strategy for treating CRF.
Collapse
Affiliation(s)
- Hongyu Luo
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Shuxian Yang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Peng Deng
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Yongbo Peng
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China.
| | - Zhiwei Chen
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Congwen Yang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Meng Wang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Renjie Qin
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Lin Yuan
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Xin Chen
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Dandan Wang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Xuekuan Huang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Jianwei Wang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
2
|
Wang JS, Tsai PH, Tseng KF, Lin CL, Chen FY, Chang CT, Shen MY. Long-Term Pentoxifylline Therapy Is Associated with a Reduced Risk of Atherosclerotic Cardiovascular Disease by Inhibiting Oxidative Stress and Cell Apoptosis in Diabetic Kidney Disease Patients. Antioxidants (Basel) 2024; 13:1471. [PMID: 39765800 PMCID: PMC11673382 DOI: 10.3390/antiox13121471] [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: 09/27/2024] [Revised: 11/20/2024] [Accepted: 11/28/2024] [Indexed: 01/03/2025] Open
Abstract
There is limited understanding of the optimal duration and dosage of pentoxifylline (PTX) therapy required to achieve significant reductions in atherosclerotic cardiovascular disease (ASCVD) risk, particularly in patients with diabetic kidney disease (DKD). This study aimed to evaluate the impact of long-term PTX therapy on the risk of ASCVD in patients with DKD who do not have pre-existing cardiovascular disease, while also exploring potential vascular protective mechanisms. This retrospective cohort study included data from Taiwan's Ministry of Health and Welfare's Health and Welfare Data Science Center. In 2008-2019, we identified and analyzed a specific sample of 129,764 patients with DKD without established cardiovascular disease. Participants were categorized according to their PTX treatment regimen. Short-term PTX users (<763 days) had a greater risk of developing ASCVD than non-PTX users. However, those who used PTX for >763 days (long-term PTX treatment) had a significantly lower risk of ASCVD, with a 47% lower cumulative incidence. A dose-dependent reduction in apoptosis was observed via Klotho treatment in cultured human aortic endothelial cells following PTX treatment. Long-term PTX treatment (24 h) caused a higher reduction in H2O2-induced reactive oxygen species production and cell apoptosis than short-term PTX treatment (2 h). In the DKD mice model experiments, PTX reduced the ASCVD risk by increasing the Klotho levels to inhibit endothelial cell damage. These findings suggest that the cardiovascular and renoprotective benefits of PTX may be extended to primary prevention strategies for people with DKD.
Collapse
Affiliation(s)
- Jie-Sian Wang
- Graduate Institute of Biomedical Sciences, China Medical University, 91, Hsueh-Shih Rd., Taichung 40402, Taiwan; (J.-S.W.); (P.-H.T.); (K.-F.T.); (F.-Y.C.)
- Division of Nephrology, Department of Internal Medicine, China Medical University Hospital, No. 2, Yude Rd., North Dist., Taichung 404327, Taiwan;
| | - Ping-Hsuan Tsai
- Graduate Institute of Biomedical Sciences, China Medical University, 91, Hsueh-Shih Rd., Taichung 40402, Taiwan; (J.-S.W.); (P.-H.T.); (K.-F.T.); (F.-Y.C.)
| | - Kuo-Feng Tseng
- Graduate Institute of Biomedical Sciences, China Medical University, 91, Hsueh-Shih Rd., Taichung 40402, Taiwan; (J.-S.W.); (P.-H.T.); (K.-F.T.); (F.-Y.C.)
| | - Cheng-Li Lin
- Management Office for Health Data, China Medical University Hospital, No. 2, Yude Rd., North Dist., Taichung 404327, Taiwan;
| | - Fang-Yu Chen
- Graduate Institute of Biomedical Sciences, China Medical University, 91, Hsueh-Shih Rd., Taichung 40402, Taiwan; (J.-S.W.); (P.-H.T.); (K.-F.T.); (F.-Y.C.)
| | - Chiz-Tzung Chang
- Division of Nephrology, Department of Internal Medicine, China Medical University Hospital, No. 2, Yude Rd., North Dist., Taichung 404327, Taiwan;
| | - Ming-Yi Shen
- Graduate Institute of Biomedical Sciences, China Medical University, 91, Hsueh-Shih Rd., Taichung 40402, Taiwan; (J.-S.W.); (P.-H.T.); (K.-F.T.); (F.-Y.C.)
- Department of Medical Research, China Medical University Hospital, No. 2, Yude Rd., North Dist., Taichung 404327, Taiwan
- Department of Nursing, Asia University, 500, Lioufeng Rd., Wufeng, Taichung 41354, Taiwan
| |
Collapse
|
3
|
Zhao Y, Wang YH, Tu WC, Wang DW, Lu MJ, Shao Y. Costunolide Inhibits Chronic Kidney Disease Development by Attenuating IKKβ/NF-κB Pathway. Drug Des Devel Ther 2024; 18:2693-2712. [PMID: 38974121 PMCID: PMC11227330 DOI: 10.2147/dddt.s466092] [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: 04/16/2024] [Accepted: 06/25/2024] [Indexed: 07/09/2024] Open
Abstract
Background Chronic kidney disease (CKD) is a significant worldwide health concern that leads to high mortality rates. The bioactive substance costunolide (CTD) has demonstrated several pharmacological effects and holds promise as a CKD treatment. This study aims to investigate the impact of CTD on CKD and delve into its mechanisms of action. Methods Unilateral ureteral obstruction (UUO) methods and renal fibrosis mice models were created. Various concentrations of CTD were injected into UUO mice models to investigate the therapeutic effects of CTD on renal fibrosis of mice. Then, renal morphology, pathological changes, and the expression of genes related to fibrosis, inflammation and ferroptosis were analysed. RNA sequencing was utilized to identify the main biological processes and pathways involved in renal injury. Finally, both overexpression and inhibition of IKKβ were studied to examine their respective effects on fibrosis and inflammation in both in vitro and in vivo models. Results CTD treatment was found to significantly alleviate fibrosis, inflammation and ferroptosis in UUO-induced renal fibrosis mice models. The results of RNA sequencing suggested that the IKKβ acted as key regulatory factor in renal injury and the expression of IKKβ was increased in vitro and in vivo renal fibrosis model. Functionally, down-regulated IKKβ expression inhibits ferroptosis, inflammatory cytokine production and collagen deposition. Conversely, IKKβ overexpression exacerbates progressive renal fibrosis. Mechanistically, CTD alleviated renal fibrosis and inflammation by inhibiting the expression of IKKβ and attenuating IKKβ/NF-κB pathway. Conclusion This study demonstrates that CTD could mitigate renal fibrosis, ferroptosis and inflammation in CKD by modulating the IKKβ/NF-κB pathway, which indicates targeting IKKβ has an enormous potential for treating CKD.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Urology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, Shanghai, 201800, People’s Republic of China
| | - Yi-Han Wang
- Department of Urology, Sixth People’s Hospital South Campus Affiliated to Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Wei-Chao Tu
- Department of Urology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, Shanghai, 201800, People’s Republic of China
| | - Da-Wei Wang
- Department of Urology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, Shanghai, 201800, People’s Republic of China
| | - Mu-Jun Lu
- Department of Urology and Andrology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
- Shanghai Institute of Andrology, Shanghai, People’s Republic of China
| | - Yuan Shao
- Department of Urology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, Shanghai, 201800, People’s Republic of China
| |
Collapse
|
4
|
Kumar A, Bajaj P, Singh B, Paul K, Sharma P, Mehra S, Robin, Kaur P, Jasrotia S, Kumar P, Rajat, Singh V, Tuli HS. Sesamol as a potent anticancer compound: from chemistry to cellular interactions. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:4961-4979. [PMID: 38180556 DOI: 10.1007/s00210-023-02919-2] [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: 06/09/2023] [Accepted: 12/20/2023] [Indexed: 01/06/2024]
Abstract
Sesamol (SM), a well-known component isolated from sesame seeds (Sesamum indicum), used in traditional medicines in treating numerous ailments. However, numerous molecular investigations revealed the various mechanisms behind its activity, emphasizing its antiproliferative, anti-inflammatory, and apoptosis-inducing properties, preventing cancer cell spread to distant organs. In several cells derived from various malignant tissues, SM-regulated signal transduction pathways and cellular targets have been identified. This review paper comprehensively describes the anticancer properties of SM and SM-viable anticancer drugs. Additionally, the interactions of this natural substance with standard anticancer drugs are examined, and the benefits of using nanotechnology in SM applications are explored. This makes SM a prime example of how ethnopharmacological knowledge can be applied to the development of contemporary drugs.
Collapse
Affiliation(s)
- Ajay Kumar
- University Center for Research & Development (UCRD), Biotechnology Engineering & Food Technology, Chandigarh University, Gharuan, Mohali, 140413, Punjab, India.
| | - Payal Bajaj
- Advanced Eye Center, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Brahmjot Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, 143005, Punjab, India
| | - Kapil Paul
- Kanya Maha Vidyalaya, Jalandhar, 144004, Punjab, India
| | - Pooja Sharma
- Department of Chemistry, Guru Nanak Dev University, Amritsar, 143005, Punjab, India
| | - Sukanya Mehra
- P.G. Department of Science, Khalsa College For Women, Amritsar, 143001, Punjab, India
| | - Robin
- Regional Water Testing Laboratory, Department of Water Supply and Sanitation, Agilent Technologies India Pvt. Ltd., Amritsar, Punjab, India
| | - Pardeep Kaur
- Post Graduate Department of Botany, Khalsa College, Amritsar, Punjab, India
| | - Shivam Jasrotia
- Department of Biosciences, University Institute of Biotechnology, Chandigarh University, Mohali, Punjab, 140413, India
| | - Parveen Kumar
- Department of Chemistry, Chandigarh University, Gharuan, Mohali, 140413, Punjab, India
| | - Rajat
- Punjab Biotechnology Incubator (PBTI), Phase VIII, Mohali, 160071, India
| | - Vipourpreet Singh
- Coast Mountain College, Prince Rupert, British Columbia, V8J3S8, Canada
| | - Hardeep Singh Tuli
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, 133207, India
| |
Collapse
|
5
|
Peng D, Huang J, Gao X, Zhou Y, Zhou M. Renoprotective effects of Schoenoplectus tabernaemontani rhizomes aqueous extracts against Adriamycin-induced nephropathy in rats. Nat Prod Res 2024:1-6. [PMID: 38808595 DOI: 10.1080/14786419.2024.2357664] [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: 01/21/2024] [Accepted: 05/14/2024] [Indexed: 05/30/2024]
Abstract
In recent years, chronic kidney disease (CKD) has emerged as an increasingly significant issue due to the growing prevalence and high treatment costs. While recorded the positive diuretic effect of Schoenoplectus tabernaemontani, there is a lack of reports on its efficacy in treating CKD. The pharmacological effects and mechanisms of Schoenoplectus tabernaemontani rhizomes aqueous extracts (STE) in CKD were investigated by inducing a rodent model of CKD via injection of Adriamycin (ADR; 7.5 mg/kg) into the tail vein of Wistar rats. In summary, our findings suggest that STE has a beneficial effect on anti-renal fibrosis and can reverse ADR-induced renal injury by suppressing oxidative stress and inflammation. Therefore, STE holds promising potential as a treatment option for CKD.
Collapse
Affiliation(s)
- Dian Peng
- Changsha Health Vocational College, Changsha, China
| | - Juan Huang
- Department of Cardiology, Hunan Children's Hospital, Changsha, China
| | - Xiaohui Gao
- Changsha Health Vocational College, Changsha, China
| | - Yingjun Zhou
- Xiangya School of Pharmaceutical Science, Central South University, Changsha, China
| | | |
Collapse
|
6
|
Mostashari P, Mousavi Khaneghah A. Sesame Seeds: A Nutrient-Rich Superfood. Foods 2024; 13:1153. [PMID: 38672826 PMCID: PMC11049391 DOI: 10.3390/foods13081153] [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: 12/24/2023] [Revised: 03/23/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Sesame seeds (Sesamum indicum L.) have been cultivated for thousands of years and have long been celebrated for their culinary versatility. Beyond their delightful nutty flavor and crunchy texture, sesame seeds have also gained recognition for their remarkable health benefits. This article provides an in-depth exploration of the numerous ways in which sesame seeds contribute to overall well-being. Sesame seeds are a powerhouse of phytochemicals, including lignans derivatives, tocopherol isomers, phytosterols, and phytates, which have been associated with various health benefits, including the preservation of cardiovascular health and the prevention of cancer, neurodegenerative disorders, and brain dysfunction. These compounds have also been substantiated for their efficacy in cholesterol management. Their potential as a natural source of beneficial plant compounds is presented in detail. The article further explores the positive impact of sesame seeds on reducing the risk of chronic diseases thanks to their rich polyunsaturated fatty acids content. Nevertheless, it is crucial to remember the significance of maintaining a well-rounded diet to achieve the proper balance of n-3 and n-6 polyunsaturated fatty acids, a balance lacking in sesame seed oil. The significance of bioactive polypeptides derived from sesame seeds is also discussed, shedding light on their applications as nutritional supplements, nutraceuticals, and functional ingredients. Recognizing the pivotal role of processing methods on sesame seeds, this review discusses how these methods can influence bioactive compounds. While roasting the seeds enhances the antioxidant properties of the oil extract, certain processing techniques may reduce phenolic compounds.
Collapse
Affiliation(s)
- Parisa Mostashari
- Department of Food Science and Technology, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran 1981619573, Iran;
- Department of Food Science and Technology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran 1941933111, Iran
| | - Amin Mousavi Khaneghah
- Faculty of Biotechnologies (BioTech), ITMO University, 9 Lomonosova Street, Saint Petersburg 191002, Russia
| |
Collapse
|
7
|
Deol PK, Kaur IP, Dhiman R, Kaur H, Sharma G, Rishi P, Ghosh D. Investigating wound healing potential of sesamol loaded solid lipid nanoparticles: Ex-vivo, in vitro and in-vivo proof of concept. Int J Pharm 2024; 654:123974. [PMID: 38447777 DOI: 10.1016/j.ijpharm.2024.123974] [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: 12/25/2023] [Revised: 02/07/2024] [Accepted: 03/03/2024] [Indexed: 03/08/2024]
Abstract
Sesamol, a lignan, obtained from sesame seeds (Sesamum indicum Linn., Pedaliaciae) has a promising antioxidant, and anti-inflammatory profile. When applied topically, free sesamol rapidly crosses skin layers and gets absorbed in systemic circulation. Its encapsulation into solid lipid nanoparticles not only improved its localised delivery to skin but also resulted in better skin retention, as found in ex-vivo skin retention studies. Free and encapsulated sesamol was compared for antimicrobial and antibiofilm activity against some common skin pathogens and it was found that encapsulation improved the antimicrobial profile by 200%. In vivo evaluation in diabetic open excision wound model suggested that encapsulation of sesamol in SLNs substantially enhanced its wound healing potential when investigated for biophysical, biochemical and histological parameters. It was envisaged that this was achieved via inhibiting bacterial growth and clearing the bacterial biofilm at the wound site, and by regulating oxidative stress in skin tissue.
Collapse
Affiliation(s)
- Parneet Kaur Deol
- G.H.G. Khalsa College of Pharmacy Gurusar Sadhar, Ludhiana, Punjab, India.
| | - Indu Pal Kaur
- University Institute of Pharmaceutical Sciences, Punjab University Chandigarh, India
| | - Ravi Dhiman
- G.H.G. Khalsa College of Pharmacy Gurusar Sadhar, Ludhiana, Punjab, India
| | - Harmanjot Kaur
- G.H.G. Khalsa College of Pharmacy Gurusar Sadhar, Ludhiana, Punjab, India
| | - Garima Sharma
- University Institute of Pharmaceutical Sciences, Punjab University Chandigarh, India
| | - Parveen Rishi
- Department of Microbiology, Punjab University Chandigarh, India
| | - Deepa Ghosh
- Institute of Nano Science and Technology, Mohali, Punjab, India
| |
Collapse
|
8
|
Xue L, Jiang S, Wan XY. Protective Effects of Sesamol on Renal Ischemia-Reperfusion Injury Via Regulation of Nuclear Factor Erythroid 2-Related Factor 2 Pathway. Transplant Proc 2024; 56:290-296. [PMID: 38350822 DOI: 10.1016/j.transproceed.2023.12.009] [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: 08/18/2023] [Accepted: 12/28/2023] [Indexed: 02/15/2024]
Abstract
BACKGROUND Sesamol is a natural antioxidant known for its potent antioxidant and free radical scavenging properties. This study aimed to explore the therapeutic effects and underlying mechanisms of sesamol in the development of renal ischemia-reperfusion injury (IRI) in mice. METHODS C57BL/6J wild-type mice were divided into 3 groups: IR group, treated with normal saline after undergoing the IRI procedure; Sesamol + IR group, treated with 30 mg/kg/d of sesamol after the IRI procedure; and Sham group, treated with normal saline but not subjected to the IRI process. Renal IRI was induced by performing a right kidney nephrectomy and subjecting the left kidney to 30-minute ischemia, followed by 24-hour reperfusion. Kidney tissues and serum were collected 24 hours post-IRI to assess the impact of sesamol on renal function after IRI. Serum creatinine and blood urea nitrogen levels were assessed, and renal cell apoptosis was detected through terminal deoxynucleotidyl transferase dUTP nick-end labeling staining. The levels of interleukin 1β and interleukin 18 in kidney tissues, as well as indicators of oxidative stress, were also measured. Furthermore, Nrf2-deficient mice were used to examine the protective function of the nuclear factor erythroid 2-related factor 2 (Nrf2)/hemeoxygenase-1 (HO-1) and NAD(P)H dehydrogenase quinone 1 (NQO1) signaling pathways induced by sesamol, as determined by western blot assay. RESULTS Sesamol demonstrated significant improvement in renal function, along with reductions in renal tubular injury, cell necrosis, and apoptosis in mice. It also effectively lowered key inflammatory mediator levels. Sesamol exhibited antioxidant properties by reducing malondialdehyde levels and enhancing superoxide dismutase activities 24 hours after IRI. Western blot assay revealed increased Nrf2, HO-1, and NQO-1 protein levels with sesamol treatment. Notably, Nrf2-deficient mice did not exhibit the beneficial effects of sesamol. CONCLUSIONS This study demonstrates that sesamol effectively alleviates renal IRI by enhancing antioxidant defenses and reducing inflammation potentially through the Nrf2/HO-1 and NQO1 signaling pathways.
Collapse
Affiliation(s)
- Lu Xue
- Department of Critical Care Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, People's Republic of China
| | - Su Jiang
- Department of Rehabilitation Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, People's Republic of China
| | - Xian-Yao Wan
- Department of Critical Care Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, People's Republic of China.
| |
Collapse
|
9
|
Majdalawieh AF, Ahari SH, Yousef SM, Nasrallah GK. Sesamol: A lignan in sesame seeds with potent anti-inflammatory and immunomodulatory properties. Eur J Pharmacol 2023; 960:176163. [PMID: 37925135 DOI: 10.1016/j.ejphar.2023.176163] [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: 07/05/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 11/06/2023]
Abstract
Inflammation is associated with the development and progression of a plethora of diseases including joint, metabolic, neurological, hepatic, and renal disorders. Sesamol, derived from the seeds of Sesamum indicum L., has received considerable attention due to its well-documented multipotent phytotherapeutic effects, including its anti-inflammatory and immunomodulatory properties. However, to date, no comprehensive review has been established to highlight or summarize the anti-inflammatory and immunomodulatory properties of sesamol. Herein, we aim to address this gap in the literature by presenting a thorough review encapsulating evidence surrounding the range of inflammatory mediators and cytokines shown to be targeted by sesamol in modulating its anti-inflammatory actions against a range of inflammatory disorders. Additionally, evidence highlighting the role that sesamol has in modulating components of adaptive immunity including cellular immune responses and Th1/Th2 balance is underscored. Moreover, the molecular mechanisms and the signaling pathways underlying such effects are also highlighted. Findings indicate that this seemingly potent lignan mediates its anti-inflammatory actions, at least in part, via suppression of various pro-inflammatory cytokines like IL-1β and TNFα, and downregulation of a multitude of signaling pathways including NF-κB and MAPK. In conclusion, we anticipate that sesamol may be employed in future therapeutic regimens to aid in more effective drug development to alleviate immune-related and inflammatory conditions.
Collapse
Affiliation(s)
- Amin F Majdalawieh
- Department of Biology, Chemistry, and Environmental Sciences, College of Arts and Sciences, American University of Sharjah, Sharjah, P.O. Box 26666, United Arab Emirates.
| | - Sogand H Ahari
- Department of Biology, Chemistry, and Environmental Sciences, College of Arts and Sciences, American University of Sharjah, Sharjah, P.O. Box 26666, United Arab Emirates
| | - Sarah M Yousef
- Department of Psychology, College of Arts and Sciences, American University of Sharjah, Sharjah, P.O. Box 26666, United Arab Emirates
| | - Gheyath K Nasrallah
- Department of Biomedical Science, College of Health Sciences, Member of QU Health, Qatar University, Doha, Qatar; Biomedical Research Center, Qatar University, Doha, Qatar
| |
Collapse
|
10
|
Lin X, Zhou Q, Zhou L, Sun Y, Han X, Cheng X, Wu M, Lv W, Wang J, Zhao W. Quinoa ( Chenopodium quinoa Willd) Bran Saponins Alleviate Hyperuricemia and Inhibit Renal Injury by Regulating the PI3K/AKT/NFκB Signaling Pathway and Uric Acid Transport. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:6635-6649. [PMID: 37083411 DOI: 10.1021/acs.jafc.3c00088] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Triterpenoids derived from natural products can exert antihyperuricemic effects. Here, we investigated the antihyperuricemic activity and mechanism of quinoa bran saponins (QBSs) in hyperuricemic mouse and cell models. The QBS4 fraction, with the highest saponin content, was used. Fourier-transform infrared, high-performance liquid chromatography, and ultrahigh-performance liquid chromatography-mass spectrometry identified 11 individual saponins in QBS4, of which the main components were hederagenin and oleanolic acid. The QBS4 effects on hyperuricemic mice (induced by adenine and potassium oxonate) were then studied. QBS4 reduced the levels of uric acid (UA), serum urea nitrogen, creatinine, and lipids in mice with hyperuricemia (HUA) and decreased renal inflammation and renal damage. Molecular analysis revealed that QBS4 may alleviate HUA by regulating the expression of key genes involved in the transport of UA and by inhibiting the activation of the PI3K/AKT/NFκB inflammatory signaling pathway. In conclusion, QBS4 has promise for using as a natural dietary supplement to treat and prevent HUA.
Collapse
Affiliation(s)
- Xuan Lin
- Department of Nutrition and Food Safety, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, P. R. China
| | - Qian Zhou
- Department of Nutrition and Food Safety, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, P. R. China
| | - Liangfu Zhou
- Department of Nutrition and Food Safety, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, P. R. China
| | - Yasai Sun
- Department of Nutrition and Food Safety, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, P. R. China
| | - Xue Han
- Department of Nutrition and Food Safety, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, P. R. China
| | - Xinlong Cheng
- Department of Nutrition and Food Safety, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, P. R. China
| | - Mengying Wu
- Department of Nutrition and Food Safety, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, P. R. China
| | - Wei Lv
- National Engineering Research Center for Semi-arid Agriculture, Shijiazhuang 050000, Hebei Province, China
| | - Jie Wang
- Department of Nutrition and Food Safety, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, P. R. China
| | - Wen Zhao
- Department of Nutrition and Food Safety, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, P. R. China
| |
Collapse
|