1
|
Xu M, Xu S, Yi X. A comparative analysis of drug-induced kidney injury adverse reactions between cyclosporine and tacrolimus based on the FAERS database. BMC Immunol 2025; 26:35. [PMID: 40316906 PMCID: PMC12049015 DOI: 10.1186/s12865-025-00714-7] [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: 01/30/2025] [Accepted: 04/14/2025] [Indexed: 05/04/2025] Open
Abstract
BACKGROUND This study utilizes the FDA Adverse Event Reporting System (FAERS) database to compare the adverse reaction signals of cyclosporine and tacrolimus, two widely used immunosuppressants, in relation to drug-induced kidney injury. The findings aim to inform clinical decision-making. METHODS The study retrospectively analyzed data from January 2004 to September 2024, employing both frequency analysis and Bayesian methods. We assessed and compared the mortality rates, hospitalization rates, and the association of cyclosporine and tacrolimus with kidney injury to elucidate the renal toxicity of these two drugs. RESULTS After data processing, we identified a total of 3,449 cyclosporine-related kidney injury reports and 5,538 tacrolimus-related kidney injury reports. The results revealed a stronger association between tacrolimus and kidney injury. Additionally, kidney injuries associated with both cyclosporine and tacrolimus predominantly affected males. Furthermore, the hospitalization rate for cyclosporine-related kidney injury was 34.40%, compared to 44.50% for tacrolimus. The mortality rate associated with cyclosporine-induced kidney injury was higher than that of tacrolimus. CONCLUSION This study utilized the FDA Adverse Event Reporting System (FAERS) database from January 2004 to September 2024 to perform a comprehensive analysis of adverse drug-related kidney injury reactions to cyclosporine and tacrolimus. The results suggest that both cyclosporine and tacrolimus are associated with renal injury, but tacrolimus appears to reduce mortality while increasing hospitalization rates. This serves as a critical warning for planning future treatment regimens, drug monitoring, and reducing adverse effects.
Collapse
Affiliation(s)
- Min Xu
- Department of Respiratory, Clinical Medical College & Affliated Hospital of Chengdu University, Chengdu University, Chengdu, 610036, China
| | - Shanggang Xu
- Department of Emergency, Clinical Medical College & Affliated Hospital of Chengdu University, Chengdu University, Chengdu, 610036, China
| | - Xueliang Yi
- Department of Emergency, Clinical Medical College & Affliated Hospital of Chengdu University, Chengdu University, Chengdu, 610036, China.
| |
Collapse
|
2
|
Kheira HS, Elsayed GR, El-Adl M. Liraglutide and resveratrol alleviated cyclosporin A induced nephrotoxicity in rats through improving antioxidant status, apoptosis and pro-inflammatory markers. Biochem Biophys Res Commun 2024; 730:150337. [PMID: 38986220 DOI: 10.1016/j.bbrc.2024.150337] [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: 05/03/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024]
Abstract
The recent study delves into the role of both liraglutide and/or resveratrol on the nephropathic affection in rats treated with cyclosporine A (CsA). Rats were intoxicated with CsA (25 mg/kg) orally for 21 days and were supplemented with liraglutide (30 μg/kg) s/c daily and 20 mg/kg of resveratrol (20 mg/kg) orally. At the end of the experiment, serum samples and renal tissues were collected to determine renal damage markers, apoptotic markers, proinflammatory markers, and antioxidant status markers. Kidney function tests and antioxidant activity notably improved in the treated rats (CsA + Lir/CsA + Res/CsA + Lir + Res). Moreover, both Lir and/or Res enhanced Bcl-2 levels while down-regulating the Bax levels in rats treated with CsA. Interestingly, the immune-staining for tumor necrosis factor (TNF-α) was tested negative and mild positive in renal tissue of rats given Lir and/or Res while being treated with Cs A which indicated their anti-inflammatory effect that reduced the renal damage. The findings of this investigation revealed the ameliorative anti-inflammatory in addition to the antioxidant role of both liraglutide and resveratrol against the kidney damage caused due to CsA administration.
Collapse
Affiliation(s)
- Hend Samy Kheira
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Gehad Ramadan Elsayed
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed El-Adl
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt.
| |
Collapse
|
3
|
Yen NTH, Tien NTN, Anh NTV, Le QV, Eunsu C, Kim HS, Moon KS, Nguyen HT, Kim DH, Long NP. Cyclosporine A-induced systemic metabolic perturbations in rats: A comprehensive metabolome analysis. Toxicol Lett 2024; 395:50-59. [PMID: 38552811 DOI: 10.1016/j.toxlet.2024.03.009] [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: 11/07/2023] [Revised: 03/12/2024] [Accepted: 03/26/2024] [Indexed: 04/11/2024]
Abstract
A better understanding of cyclosporine A (CsA)-induced nephro- and hepatotoxicity at the molecular level is necessary for safe and effective use. Utilizing a sophisticated study design, this study explored metabolic alterations after long-term CsA treatment in vivo. Rats were exposed to CsA with 4, 10, and 25 mg/kg for 4 weeks and then sacrificed to obtain liver, kidney, urine, and serum for untargeted metabolomics analysis. Differential network analysis was conducted to explore the biological relevance of metabolites significantly altered by toxicity-induced disturbance. Dose-dependent toxicity was observed in all biospecimens. The toxic effects were characterized by alterations of metabolites related to energy metabolism and cellular membrane composition, which could lead to the cholestasis-induced accumulation of bile acids in the tissues. The unfavorable impacts were also demonstrated in the serum and urine. Intriguingly, phenylacetylglycine was increased in the kidney, urine, and serum treated with high doses versus controls. Differential correlation network analysis revealed the strong correlations of deoxycytidine and guanosine with other metabolites in the network, which highlighted the influence of repeated CsA exposure on DNA synthesis. Overall, prolonged CsA administration had system-level dose-dependent effects on the metabolome in treated rats, suggesting the need for careful usage and dose adjustment.
Collapse
Affiliation(s)
- Nguyen Thi Hai Yen
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Nguyen Tran Nam Tien
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Nguyen Thi Van Anh
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Quoc-Viet Le
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam
| | - Cho Eunsu
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Ho-Sook Kim
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Kyoung-Sik Moon
- Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Huy Truong Nguyen
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam
| | - Dong Hyun Kim
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea.
| | - Nguyen Phuoc Long
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea.
| |
Collapse
|
4
|
Abd-Eldayem AM, Makram SM, Messiha BAS, Abd-Elhafeez HH, Abdel-Reheim MA. Cyclosporine-induced kidney damage was halted by sitagliptin and hesperidin via increasing Nrf2 and suppressing TNF-α, NF-κB, and Bax. Sci Rep 2024; 14:7434. [PMID: 38548778 PMCID: PMC10978894 DOI: 10.1038/s41598-024-57300-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 03/16/2024] [Indexed: 04/01/2024] Open
Abstract
Cyclosporine A (CsA) is employed for organ transplantation and autoimmune disorders. Nephrotoxicity is a serious side effect that hampers the therapeutic use of CsA. Hesperidin and sitagliptin were investigated for their antioxidant, anti-inflammatory, and tissue-protective properties. We aimed to investigate and compare the possible nephroprotective effects of hesperidin and sitagliptin. Male Wistar rats were utilized for induction of CsA nephrotoxicity (20 mg/kg/day, intraperitoneally for 7 days). Animals were treated with sitagliptin (10 mg/kg/day, orally for 14 days) or hesperidin (200 mg/kg/day, orally for 14 days). Blood urea, serum creatinine, albumin, cystatin-C (CYS-C), myeloperoxidase (MPO), and glucose were measured. The renal malondialdehyde (MDA), glutathione (GSH), catalase, and SOD were estimated. Renal TNF-α protein expression was evaluated. Histopathological examination and immunostaining study of Bax, Nrf-2, and NF-κB were performed. Sitagliptin or hesperidin attenuated CsA-mediated elevations of blood urea, serum creatinine, CYS-C, glucose, renal MDA, and MPO, and preserved the serum albumin, renal catalase, SOD, and GSH. They reduced the expressions of TNF-α, Bax, NF-κB, and pathological kidney damage. Nrf2 expression in the kidney was raised. Hesperidin or sitagliptin could protect the kidney against CsA through the mitigation of oxidative stress, apoptosis, and inflammation. Sitagliptin proved to be more beneficial than hesperidin.
Collapse
Affiliation(s)
- Ahmed M Abd-Eldayem
- Department of Medical Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt.
- Department of Pharmacology, Faculty of Medicine, Merit University, Sohâg, Egypt.
| | | | | | - Hanan H Abd-Elhafeez
- Department of Cell and Tissue, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Mustafa Ahmed Abdel-Reheim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef, Egypt
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra, Saudi Arabia
| |
Collapse
|
5
|
Jin M, Yi X, Zhu X, Hu W, Wang S, Chen Q, Yang W, Li Y, Li S, Peng Q, Pan M, Gao Y, Xu S, Zhang Y, Zhou S. Schisandrin B promotes hepatic differentiation from human umbilical cord mesenchymal stem cells. iScience 2024; 27:108912. [PMID: 38323006 PMCID: PMC10844828 DOI: 10.1016/j.isci.2024.108912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 10/30/2023] [Accepted: 01/11/2024] [Indexed: 02/08/2024] Open
Abstract
Human umbilical cord mesenchymal stem cells (UC-MSCs)-derived hepatocyte-like cells (HLCs) have shown great promise in the treatment of liver diseases. However, most current induction protocols yield hepatocyte-like cells with limited function as compared with primary hepatocytes. Schisandrin B (Sch B) is one of the main components of Schisandra chinensis, which can prevent fibrosis progression and promote liver cell regeneration. Herein, we investigated the effects of Sch B on hepatic differentiation of UC-MSCs. We found that treatment with 10 μM Sch B from the second stage of the differentiation process increased hepatic marker levels and hepatic function. Additionally, RNA-seq analysis revealed that Sch B promoted hepatic differentiation via activating the JAK2/STAT3 pathway. When transplanted HLCs into mice with CCL4-induced liver fibrosis, Sch B-treated HLCs exhibited significant therapeutic effects. This study provides an optimized hepatic differentiation protocol for UC-MSCs based on Sch B, yielding functioning cells for liver disease treatment.
Collapse
Affiliation(s)
- Meixian Jin
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Xiao Yi
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Xiaojuan Zhu
- Department of Anesthesiology, First People’s Hospital of Kashi, Kashi 844000, China
| | - Wei Hu
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Simin Wang
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Qi Chen
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Wanren Yang
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Yang Li
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Shao Li
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Qing Peng
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Mingxin Pan
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Yi Gao
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Shiyuan Xu
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Ying Zhang
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Shuqin Zhou
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
- Anesthesiology Department of The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People’s Hospital of Shenzhen, Shenzhen 518172, China
| |
Collapse
|
6
|
Ramadan MM, Mohamed RS, Hussien AG, Mohawed OA, Mabrouk AM, Mahmoud AE, Ghanem KZ, El-Messery TM. Ameliorative effects of vitamins-loaded flavoured nanophytosomes fortified with star anise volatile oil against CsA-Induced liver and kidney injury in rats: Application in functional ice cream. Heliyon 2024; 10:e23894. [PMID: 38226243 PMCID: PMC10788501 DOI: 10.1016/j.heliyon.2023.e23894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 12/07/2023] [Accepted: 12/14/2023] [Indexed: 01/17/2024] Open
Abstract
This study investigated the effect of flavoured nanophytosomes loaded with vitamins A, E, D, B complex, folic acid, and C, as well as zinc on the immunosuppressive cyclosporin A (CsA)-induced liver and kidney injury in male rats. The vitamins flavoured nanophytosomes (VFnPs) were characterized in terms of particle size, zeta potential, encapsulation efficiency. Ice cream was flavoured with star anise volatile oil to mask the VFnPs' flavour and unacceptable taste. The study found that treatment with CsA alone resulted in increased (P > 0.05) levels of creatinine, urea, and MDA, as well as the activities of AST and ALT, while the levels of SOD, CAT, GST, proteins, CD4, INF-ᵧ, IL-6, IL-1β, and TLR4 decreased (P > 0.05). However, the group that received CsA simultaneously with VFnPs showed a significant (P > 0.05) decrease in the levels of creatinine, urea, and MDA, as well as the activities of AST and ALT, and increased (P > 0.05) levels of SOD, CAT, GST, proteins, CD4, INF-ᵧ, IL-6, IL-1β, and TLR4. The increase in the ratio of VFnPs had little effect on the physiochemical and sensory evaluation of the ice cream. Finally, the study suggests that VFnPs could potentially protect against CsA-induced liver and kidney injury and serve as a promising natural therapy for treating such conditions.
Collapse
Affiliation(s)
- Manal M. Ramadan
- Chemistry of Flavour and Aroma Department, National Research Centre, Cairo, Egypt
| | - Rasha S. Mohamed
- Nutrition and Food Sciences Department, National Research Centre, Cairo, Egypt
| | - Amal G. Hussien
- Biochemistry Department, Biotechnology Research Institute, National Research Centre. Cairo, Egypt
| | - Ola A.M. Mohawed
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre. Cairo, Egypt
| | - Ahmed M. Mabrouk
- Dairy Science Department, National Research Centre, Cairo, Egypt
| | - Abeer E. Mahmoud
- Biochemistry Department, Biotechnology Research Institute, National Research Centre. Cairo, Egypt
| | - Kadry Z. Ghanem
- Nutrition and Food Sciences Department, National Research Centre, Cairo, Egypt
| | - Tamer M. El-Messery
- International Research Centre “Biotechnologies of the Third Millennium”, ITMO University, St. Petersburg, 191002, Russia
| |
Collapse
|
7
|
Song J, Zhang B, Zhang H, Cheng W, Liu P, Kang J. Quantitative Proteomics Combined with Network Pharmacology Analysis Unveils the Biological Basis of Schisandrin B in Treating Diabetic Nephropathy. Comb Chem High Throughput Screen 2024; 27:284-297. [PMID: 37151069 DOI: 10.2174/1386207326666230505111903] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 03/16/2023] [Accepted: 03/22/2023] [Indexed: 05/09/2023]
Abstract
BACKGROUND Diabetic nephropathy (DN) is a major complication of diabetes. Schisandrin B (Sch) is a natural pharmaceutical monomer that was shown to prevent kidney damage caused by diabetes and restore its function. However, there is still a lack of comprehensive and systematic understanding of the mechanism of Sch treatment in DN. OBJECTIVE We aim to provide a systematic overview of the mechanisms of Sch in multiple pathways to treat DN in rats. METHODS Streptozocin was used to build a DN rat model, which was further treated with Sch. The possible mechanism of Sch protective effects against DN was predicted using network pharmacology and was verified by quantitative proteomics analysis. RESULTS High dose Sch treatment significantly downregulated fasting blood glucose, creatinine, blood urea nitrogen, and urinary protein levels and reduced collagen deposition in the glomeruli and tubule-interstitium of DN rats. The activities of superoxide dismutase (SOD) and plasma glutathione peroxidase (GSH-Px) in the kidney of DN rats significantly increased with Sch treatment. In addition, the levels of IL-6, IL-1β, and TNF-α were significantly reduced in DN rats treated with Sch. 11 proteins that target both Sch and DN were enriched in pathways such as MAPK signaling, PI3K-Akt signaling, renal cell carcinoma, gap junction, endocrine resistance, and TNF signaling. Furthermore, quantitative proteomics showed that Xaf1 was downregulated in the model vs. control group and upregulated in the Sch-treated vs. model group. Five proteins, Crb3, Tspan4, Wdr45, Zfp512, and Tmigd1, were found to be upregulated in the model vs. control group and downregulated in the Sch vs. model group. Three intersected proteins between the network pharmacology prediction and proteomics results, Crb3, Xaf1, and Tspan4, were identified. CONCLUSION Sch functions by relieving oxidative stress and the inflammatory response by regulating Crb3, Xaf1, and Tspan4 protein expression levels to treat DN disease.
Collapse
Affiliation(s)
- Jianying Song
- School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin, 300072, People's Republic of China
| | - Bo Zhang
- Institute for TCM-X, MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Huiping Zhang
- Shanghai Applied Protein Technology Co., Ltd., 58 Yuanmei Road, Shanghai, 200233, People's Republic of China
| | - Wenbo Cheng
- Tianjin Key Laboratory of Medical Mass Spectrometry for Accurate Diagnosis, Tianjin, 300399, People's Republic of China
| | - Peiyuan Liu
- School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin, 300072, People's Republic of China
| | - Jun Kang
- School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin, 300072, People's Republic of China
| |
Collapse
|
8
|
Zhou Y, Zhang A, Fang C, Yuan L, Shao A, Xu Y, Zhou D. Oxidative stress in pituitary neuroendocrine tumors: Affecting the tumor microenvironment and becoming a new target for pituitary neuroendocrine tumor therapy. CNS Neurosci Ther 2023; 29:2744-2759. [PMID: 37341156 PMCID: PMC10493678 DOI: 10.1111/cns.14315] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/29/2023] [Accepted: 06/07/2023] [Indexed: 06/22/2023] Open
Abstract
Pituitary adenomas (PAs), or pituitary neuroendocrine tumors (PitNETs), are commonly found in the anterior pituitary gland. Although the majority of PitNETs are benign and stable, several tumors have malignant characteristics. The tumor microenvironment (TME) plays an important role in the process of tumorigenesis and is composed of several types of cells. Various cells in the TME are significantly affected by oxidative stress. It has been reported that immunotherapeutic strategies have good effects in several cancers. However, the clinical potential of immunotherapies in PitNETs has not yet been fully discussed. Oxidative stress can regulate PitNET cells and immune cells in the TME, thus affecting the immune status of the TME of PitNETs. Therefore, modulation of oxidative stress-regulated immune cells using a combination of several agents and the immune system to suppress PitNETs is a promising therapeutic direction. In this review, we systematically analyzed the oxidative stress process within PitNET cells and various immune cells to elucidate the potential value of immunotherapy.
Collapse
Affiliation(s)
- Yuhang Zhou
- The First Clinical Medical CollegeHeilongjiang University of Chinese MedicineHarbinChina
- Health Management CenterTongde Hospital of Zhejiang ProvinceHangzhouChina
| | - Anke Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Chaoyou Fang
- Department of Neurosurgery, Shanghai General Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Ling Yuan
- School of Public Health, School of MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Yuanzhi Xu
- Department of Neurosurgery, Huashan Hospital, School of MedicineFudan UniversityShanghaiChina
| | - Danyang Zhou
- Health Management CenterTongde Hospital of Zhejiang ProvinceHangzhouChina
| |
Collapse
|
9
|
Jia M, Zhou L, Lou Y, Yang X, Zhao H, Ouyang X, Huang Y. An analysis of the nutritional effects of Schisandra chinensis components based on mass spectrometry technology. Front Nutr 2023; 10:1227027. [PMID: 37560060 PMCID: PMC10408133 DOI: 10.3389/fnut.2023.1227027] [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: 05/26/2023] [Accepted: 07/12/2023] [Indexed: 08/11/2023] Open
Abstract
OBJECTIVE Schisandra chinensis (Turcz.) Baill. (S. chinensis) is a Traditional Chinese medicinal herb that can be used both for medicinal purposes and as a food ingredient due to its beneficial properties, and it is enriched with a wide of natural plant nutrients, including flavonoids, phenolic acids, anthocyanins, lignans, triterpenes, organic acids, and sugars. At present, there is lack of comprehensive study or systemic characterization of nutritional and active ingredients of S. chinensis using innovative mass spectrometry techniques. METHODS The comprehensive review was conducted by searching the PubMed databases for relevant literature of various mass spectrometry techniques employed in the analysis of nutritional components in S. chinensis, as well as their main nutritional effects. The literature search covered the past 5 years until March 15, 2023. RESULTS The potential nutritional effects of S. chinensis are discussed, including its ability to enhance immunity, function as an antioxidant, anti-allergen, antidepressant, and anti-anxiety agent, as well as its ability to act as a sedative-hypnotic and improve memory, cognitive function, and metabolic imbalances. Meanwhile, the use of advanced mass spectrometry detection technologies have the potential to enable the discovery of new nutritional components of S. chinensis, and to verify the effects of different extraction methods on these components. The contents of anthocyanins, lignans, organic acids, and polysaccharides, the main nutritional components in S. chinensis, are also closely associated to its quality. CONCLUSION This review will provide guidelines for an in-depth study on the nutritional value of S. chinensis and for the development of healthy food products with effective components.
Collapse
Affiliation(s)
- Mengzhen Jia
- Department of Pediatrics, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Li Zhou
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yuanyuan Lou
- Department of Pediatrics, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xiaoqing Yang
- Department of Pediatrics, The First Affiliated Hospital of Henan University of CM, Zhengzhou, Henan, China
| | - Hangyu Zhao
- Department of Pediatrics, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xinshou Ouyang
- Department of Internal Medicine, Digestive Disease Section, Yale University, New Haven, CT, United States
| | - Yanjie Huang
- Department of Pediatrics, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- Department of Pediatrics, The First Affiliated Hospital of Henan University of CM, Zhengzhou, Henan, China
| |
Collapse
|
10
|
Natural Reno-Protective Agents against Cyclosporine A-Induced Nephrotoxicity: An Overview. Molecules 2022; 27:molecules27227771. [PMID: 36431872 PMCID: PMC9693876 DOI: 10.3390/molecules27227771] [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: 10/17/2022] [Revised: 11/06/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022] Open
Abstract
CA (cyclosporine A) is a powerful immunosuppressing agent that is commonly utilized for treating various autoimmune illnesses and in transplantation surgery. However, its usage has been significantly restricted because of its unwanted effects, including nephrotoxicity. The pathophysiology of CA-induced kidney injury involves inflammation, apoptosis, tubular injury, oxidative stress, and vascular injury. Despite the fact that exact mechanism accountable for CA's effects is inadequately understood, ROS (reactive oxygen species) involvement has been widely proposed. At present, there are no efficient methods or drugs for treating CA-caused kidney damage. It is noteworthy that diverse natural products have been investigated both in vivo and in-vitro for their possible preventive potential in CA-produced nephrotoxicity. Various extracts and natural metabolites have been found to possess a remarkable potential for restoring CA-produced renal damage and oxidative stress alterations via their anti-apoptosis, anti-inflammatory, and antioxidative potentials. The present article reviews the reported studies that assess the protective capacity of natural products, as well as dietary regimens, in relation to CA-induced nephrotoxicity. Thus, the present study presents novel ideas for designing and developing more efficient prophylactic or remedial strategies versus CA passive influences.
Collapse
|
11
|
Nsengimana B, Okpara ES, Hou W, Yan C, Han S. Involvement of oxidative species in cyclosporine-mediated cholestasis. Front Pharmacol 2022; 13:1004844. [PMID: 36425570 PMCID: PMC9679297 DOI: 10.3389/fphar.2022.1004844] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/24/2022] [Indexed: 04/11/2025] Open
Abstract
Cyclosporine is an established medication for the prevention of transplant rejection. However, adverse consequences such as nephrotoxicity, hepatotoxicity, and cholestasis have been associated with prolonged usage. In cyclosporine-induced obstructive and chronic cholestasis, for example, the overproduction of oxidative stress is significantly increased. Additionally, cyclosporine exerts adverse effects on liver function and redox balance responses in treated rats, as evidenced by its increasing levels of aspartate aminotransferase (AST), alanine aminotransferase (ALT), and bilirubin while also decreasing the levels of glutathione and NADPH. Cyclosporine binds to cyclophilin to produce its therapeutic effects, and the resulting complex inhibits calcineurin, causing calcium to accumulate in the mitochondria. Accumulating calcium with concomitant mitochondrial abnormalities induces oxidative stress, perturbation in ATP balance, and failure of calcium pumps. Also, cyclosporine-induced phagocyte oxidative stress generation via the interaction of phagocytes with Toll-like receptor-4 has been studied. The adverse effect of cyclosporine may be amplified by the release of mitochondrial DNA, mediated by oxidative stress-induced mitochondrial damage. Given the uncertainty surrounding the mechanism of cyclosporine-induced oxidative stress in cholestasis, we aim to illuminate the involvement of oxidative stress in cyclosporine-mediated cholestasis and also explore possible strategic interventions that may be applied in the future.
Collapse
Affiliation(s)
| | | | | | | | - Shuxin Han
- Department of Hepatobiliary Surgery, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
12
|
Feng YL, Yang Y, Chen H. Small molecules as a source for acute kidney injury therapy. Pharmacol Ther 2022; 237:108169. [DOI: 10.1016/j.pharmthera.2022.108169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/28/2022] [Accepted: 03/07/2022] [Indexed: 10/18/2022]
|
13
|
Yao X, Takayama H, Kamoshita K, Oo HK, Tanida R, Kato K, Ishii KA, Takamura T. Cyclosporine A Downregulates Selenoprotein P Expression via a Signal Transducer and Activator of Transcription 3-Forkhead Box Protein O1 Pathway in Hepatocytes In Vitro. J Pharmacol Exp Ther 2022. [PMID: 35906096 DOI: 10.1124/jpet.121.000467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cyclosporine A (CsA) is an immunosuppressant applied worldwide for preventing graft rejection and autoimmune diseases. However, CsA elevates oxidative stress, which can lead to liver injuries. The present study aimed to clarify the mechanisms underlying the CsA-mediated oxidative stress. Among the redox proteins, CsA concentration-dependently downregulated Selenop-encoding selenoprotein P, a major circulating antioxidant protein reducing reactive oxygen species, in hepatocytes cell lines and primary hepatocytes. The luciferase assay identified the CsA-responsive element in the SELENOP promoter containing a putative binding site for forkhead box protein O (FoxO) 1. The CsA-mediated suppression on the SELENOP promoter was independent of the nuclear factor of activated T-cell, a classic target repressed by CsA. A chromatin immunoprecipitation assay showed that CsA suppressed the FoxO1 binding to the SELENOP promoter. Foxo1 knockdown significantly downregulated Selenop expression in H4IIEC3 cells. Furthermore, CsA downregulated FoxO1 by inactivating its upstream signal transducer and activator of transcription 3 (STAT3). Knockdown of Stat3 downregulated Foxo1 and Selenop expression in hepatocytes. These findings revealed a novel mechanism underlying CsA-induced oxidative stress by downregulating the STAT3-FoxO1-Selenop pathway in hepatocytes. SIGNIFICANCE STATEMENT: This study shows that Cyclosporine A (CsA) downregulates Selenop, an antioxidant protein, by suppressing the signal transducer and activator of transcription 3-forkhead box protein O1 pathway in hepatocytes, possibly one of the causations of CsA-induced oxidative stress in hepatocytes. The present study sheds light on the previously unrecognized CsA-redox axis.
Collapse
Affiliation(s)
- Xingyu Yao
- Department of Endocrinology and Metabolism, (X.Y., H.T., Ky.K., H.K.O., R.T., Ka.K., T.T.), Life Sciences Division, Engineering and Technology Department (H.T.), and Department of Integrative Medicine for Longevity (K.-A.I.), Graduate School of Medical Sciences, Kanazawa University, Kanazawa City, Ishikawa Prefecture, Japan
| | - Hiroaki Takayama
- Department of Endocrinology and Metabolism, (X.Y., H.T., Ky.K., H.K.O., R.T., Ka.K., T.T.), Life Sciences Division, Engineering and Technology Department (H.T.), and Department of Integrative Medicine for Longevity (K.-A.I.), Graduate School of Medical Sciences, Kanazawa University, Kanazawa City, Ishikawa Prefecture, Japan
| | - Kyoko Kamoshita
- Department of Endocrinology and Metabolism, (X.Y., H.T., Ky.K., H.K.O., R.T., Ka.K., T.T.), Life Sciences Division, Engineering and Technology Department (H.T.), and Department of Integrative Medicine for Longevity (K.-A.I.), Graduate School of Medical Sciences, Kanazawa University, Kanazawa City, Ishikawa Prefecture, Japan
| | - Hein Ko Oo
- Department of Endocrinology and Metabolism, (X.Y., H.T., Ky.K., H.K.O., R.T., Ka.K., T.T.), Life Sciences Division, Engineering and Technology Department (H.T.), and Department of Integrative Medicine for Longevity (K.-A.I.), Graduate School of Medical Sciences, Kanazawa University, Kanazawa City, Ishikawa Prefecture, Japan
| | - Ryota Tanida
- Department of Endocrinology and Metabolism, (X.Y., H.T., Ky.K., H.K.O., R.T., Ka.K., T.T.), Life Sciences Division, Engineering and Technology Department (H.T.), and Department of Integrative Medicine for Longevity (K.-A.I.), Graduate School of Medical Sciences, Kanazawa University, Kanazawa City, Ishikawa Prefecture, Japan
| | - Kaisei Kato
- Department of Endocrinology and Metabolism, (X.Y., H.T., Ky.K., H.K.O., R.T., Ka.K., T.T.), Life Sciences Division, Engineering and Technology Department (H.T.), and Department of Integrative Medicine for Longevity (K.-A.I.), Graduate School of Medical Sciences, Kanazawa University, Kanazawa City, Ishikawa Prefecture, Japan
| | - Kiyo-Aki Ishii
- Department of Endocrinology and Metabolism, (X.Y., H.T., Ky.K., H.K.O., R.T., Ka.K., T.T.), Life Sciences Division, Engineering and Technology Department (H.T.), and Department of Integrative Medicine for Longevity (K.-A.I.), Graduate School of Medical Sciences, Kanazawa University, Kanazawa City, Ishikawa Prefecture, Japan
| | - Toshinari Takamura
- Department of Endocrinology and Metabolism, (X.Y., H.T., Ky.K., H.K.O., R.T., Ka.K., T.T.), Life Sciences Division, Engineering and Technology Department (H.T.), and Department of Integrative Medicine for Longevity (K.-A.I.), Graduate School of Medical Sciences, Kanazawa University, Kanazawa City, Ishikawa Prefecture, Japan
| |
Collapse
|
14
|
Yao X, Takayama H, Kamoshita K, Oo HK, Tanida R, Kato K, Ishii KA, Takamura T. Cyclosporine A Downregulates Selenoprotein P Expression via a Signal Transducer and Activator of Transcription 3-Forkhead Box Protein O1 Pathway in Hepatocytes In Vitro. J Pharmacol Exp Ther 2022; 382:199-207. [PMID: 35906096 DOI: 10.1124/jpet.121.001175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 11/22/2022] Open
Abstract
Cyclosporine A (CsA) is an immunosuppressant applied worldwide for preventing graft rejection and autoimmune diseases. However, CsA elevates oxidative stress, which can lead to liver injuries. The present study aimed to clarify the mechanisms underlying the CsA-mediated oxidative stress. Among the redox proteins, CsA concentration-dependently downregulated Selenop-encoding selenoprotein P, a major circulating antioxidant protein reducing reactive oxygen species, in hepatocytes cell lines and primary hepatocytes. The luciferase assay identified the CsA-responsive element in the SELENOP promoter containing a putative binding site for forkhead box protein O (FoxO) 1. The CsA-mediated suppression on the SELENOP promoter was independent of the nuclear factor of activated T-cell, a classic target repressed by CsA. A chromatin immunoprecipitation assay showed that CsA suppressed the FoxO1 binding to the SELENOP promoter. Foxo1 knockdown significantly downregulated Selenop expression in H4IIEC3 cells. Furthermore, CsA downregulated FoxO1 by inactivating its upstream signal transducer and activator of transcription 3 (STAT3). Knockdown of Stat3 downregulated Foxo1 and Selenop expression in hepatocytes. These findings revealed a novel mechanism underlying CsA-induced oxidative stress by downregulating the STAT3-FoxO1-Selenop pathway in hepatocytes. SIGNIFICANCE STATEMENT: This study shows that Cyclosporine A (CsA) downregulates Selenop, an antioxidant protein, by suppressing the signal transducer and activator of transcription 3-forkhead box protein O1 pathway in hepatocytes, possibly one of the causations of CsA-induced oxidative stress in hepatocytes. The present study sheds light on the previously unrecognized CsA-redox axis.
Collapse
Affiliation(s)
- Xingyu Yao
- Department of Endocrinology and Metabolism, (X.Y., H.T., Ky.K., H.K.O., R.T., Ka.K., T.T.), Life Sciences Division, Engineering and Technology Department (H.T.), and Department of Integrative Medicine for Longevity (K.-A.I.), Graduate School of Medical Sciences, Kanazawa University, Kanazawa City, Ishikawa Prefecture, Japan
| | - Hiroaki Takayama
- Department of Endocrinology and Metabolism, (X.Y., H.T., Ky.K., H.K.O., R.T., Ka.K., T.T.), Life Sciences Division, Engineering and Technology Department (H.T.), and Department of Integrative Medicine for Longevity (K.-A.I.), Graduate School of Medical Sciences, Kanazawa University, Kanazawa City, Ishikawa Prefecture, Japan
| | - Kyoko Kamoshita
- Department of Endocrinology and Metabolism, (X.Y., H.T., Ky.K., H.K.O., R.T., Ka.K., T.T.), Life Sciences Division, Engineering and Technology Department (H.T.), and Department of Integrative Medicine for Longevity (K.-A.I.), Graduate School of Medical Sciences, Kanazawa University, Kanazawa City, Ishikawa Prefecture, Japan
| | - Hein Ko Oo
- Department of Endocrinology and Metabolism, (X.Y., H.T., Ky.K., H.K.O., R.T., Ka.K., T.T.), Life Sciences Division, Engineering and Technology Department (H.T.), and Department of Integrative Medicine for Longevity (K.-A.I.), Graduate School of Medical Sciences, Kanazawa University, Kanazawa City, Ishikawa Prefecture, Japan
| | - Ryota Tanida
- Department of Endocrinology and Metabolism, (X.Y., H.T., Ky.K., H.K.O., R.T., Ka.K., T.T.), Life Sciences Division, Engineering and Technology Department (H.T.), and Department of Integrative Medicine for Longevity (K.-A.I.), Graduate School of Medical Sciences, Kanazawa University, Kanazawa City, Ishikawa Prefecture, Japan
| | - Kaisei Kato
- Department of Endocrinology and Metabolism, (X.Y., H.T., Ky.K., H.K.O., R.T., Ka.K., T.T.), Life Sciences Division, Engineering and Technology Department (H.T.), and Department of Integrative Medicine for Longevity (K.-A.I.), Graduate School of Medical Sciences, Kanazawa University, Kanazawa City, Ishikawa Prefecture, Japan
| | - Kiyo-Aki Ishii
- Department of Endocrinology and Metabolism, (X.Y., H.T., Ky.K., H.K.O., R.T., Ka.K., T.T.), Life Sciences Division, Engineering and Technology Department (H.T.), and Department of Integrative Medicine for Longevity (K.-A.I.), Graduate School of Medical Sciences, Kanazawa University, Kanazawa City, Ishikawa Prefecture, Japan
| | - Toshinari Takamura
- Department of Endocrinology and Metabolism, (X.Y., H.T., Ky.K., H.K.O., R.T., Ka.K., T.T.), Life Sciences Division, Engineering and Technology Department (H.T.), and Department of Integrative Medicine for Longevity (K.-A.I.), Graduate School of Medical Sciences, Kanazawa University, Kanazawa City, Ishikawa Prefecture, Japan
| |
Collapse
|
15
|
Osmakov DI, Kalinovskii AP, Belozerova OA, Andreev YA, Kozlov SA. Lignans as Pharmacological Agents in Disorders Related to Oxidative Stress and Inflammation: Chemical Synthesis Approaches and Biological Activities. Int J Mol Sci 2022; 23:6031. [PMID: 35682715 PMCID: PMC9181380 DOI: 10.3390/ijms23116031] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/17/2022] [Accepted: 05/24/2022] [Indexed: 02/06/2023] Open
Abstract
Plant lignans exhibit a wide range of biological activities, which makes them the research objects of potential use as therapeutic agents. They provide diverse naturally-occurring pharmacophores and are available for production by chemical synthesis. A large amount of accumulated data indicates that lignans of different structural groups are apt to demonstrate both anti-inflammatory and antioxidant effects, in many cases, simultaneously. In this review, we summarize the comprehensive knowledge about lignan use as a bioactive agent in disorders associated with oxidative stress and inflammation, pharmacological effects in vitro and in vivo, molecular mechanisms underlying these effects, and chemical synthesis approaches. This article provides an up-to-date overview of the current data in this area, available in PubMed, Scopus, and Web of Science databases, screened from 2000 to 2022.
Collapse
Affiliation(s)
- Dmitry I. Osmakov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (D.I.O.); (A.P.K.); (O.A.B.); (Y.A.A.)
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Aleksandr P. Kalinovskii
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (D.I.O.); (A.P.K.); (O.A.B.); (Y.A.A.)
| | - Olga A. Belozerova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (D.I.O.); (A.P.K.); (O.A.B.); (Y.A.A.)
| | - Yaroslav A. Andreev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (D.I.O.); (A.P.K.); (O.A.B.); (Y.A.A.)
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Sergey A. Kozlov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia; (D.I.O.); (A.P.K.); (O.A.B.); (Y.A.A.)
| |
Collapse
|
16
|
Wu Q, Liu C, Zhang J, Xiao W, Yang F, Yu Y, Li T, Wang Y. Schisandra chinensis polysaccharide protects against cyclosporin A-induced liver injury by promoting hepatocyte proliferation. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
17
|
Schisandrin B Attenuates Hepatic Stellate Cell Activation and Promotes Apoptosis to Protect against Liver Fibrosis. Molecules 2021; 26:molecules26226882. [PMID: 34833975 PMCID: PMC8620732 DOI: 10.3390/molecules26226882] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 12/14/2022] Open
Abstract
The activation of hepatic stellate cells (HSC) plays a key role in the progression of hepatic fibrosis, it is essential to remove activated HSC through apoptosis to reverse hepatic fibrosis. Schisandrin B (Sch B) is the main chemical component of schisandrin lignan, and it has been reported to have good hepatoprotective effects. However, Schisandrin B on HSC apoptosis remains unclear. In our study, we stimulated the HSC-T6 and LX-2 cell lines with TGF-β1 to induce cell activation, and the proliferation and apoptosis of the activated HSC-T6 and LX-2 cells were detected after treatment with different doses of Schisandrin B. Flow cytometry results showed that Sch B significantly reduced the activity of activated HSC-T6 and LX-2 cells and significantly induced apoptosis. In addition, the cleaved-Caspase-3 levels were increased, the Bax activity was increased, and the Bcl-2 expression was decreased in HSC-T6 and LX-2 cells treated with Sch B. Our study showed that Sch B inhibited the TGF-β1-induced activity of hepatic stellate cells by promoting apoptosis.
Collapse
|
18
|
Tian J, Huang Y, Wu T, Huang HD, Ko KM, Zhu BT, Chen J. The Use of Chinese Yang/Qi-Invigorating Tonic Botanical Drugs/Herbal Formulations in Ameliorating Chronic Kidney Disease by Enhancing Mitochondrial Function. Front Pharmacol 2021; 12:622498. [PMID: 34248614 PMCID: PMC8264145 DOI: 10.3389/fphar.2021.622498] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 06/11/2021] [Indexed: 12/27/2022] Open
Abstract
Background: Chronic kidney disease (CKD) is a leading cause of morbidity and mortality. Mitochondrial dysfunction has been implicated as a key factor in the development of CKD. According to traditional Chinese medicine (TCM) theory, many Chinese Yang/Qi-invigorating botanical drugs/herbal formulations have been shown to produce promising outcomes in the clinical management of CKD. Experimental studies have indicated that the health-promoting action of Yang/Qi invigoration in TCM is related to the up-regulation of mitochondrial energy generation and antioxidant status. Objective: In this review, we aim to test whether Chinese Yang/Qi-invigorating tonic botanical drugs/herbal formulations can provide medical benefits in CKD and its complications. And we also explore the possible involvement of mitochondrial-associated signaling pathway underlying the beneficial effects of Yang/Qi invigoration in TCM. Methods: A systematic search of "PubMed", "China National Knowledge Infrastructure (CNKI)" and "Google Scholar" was carried out to collect all the available articles in English or Chinese related to Chinese Yang/Qi-invigorating tonic botanical drugs/herbal formulations and their effects on mitochondrial function and chronic kidney disease. Result and Discussion: The relationship between the progression of CKD and mitochondrial function is discussed. The effects of Chinese Yang/Qi-invigorating tonic botanical drugs/herbal formulations and their active ingredients, including phytosterols/triterpenes, flavonoids, and dibenzocyclooctadiene lignans, on CKD and related alterations in mitochondrial signaling pathways are also presented in this review. In the future, exploration of the possible beneficial effects and clinical studies of more Yang- and Qi-invigorating botanical drugs/herbal formulations in the prevention and/or/treatment of CKD and the molecular mechanisms relating to the enhancement of mitochondrial functions warrants further investigation. Conclusion: Given the critical role of mitochondrial function in safeguarding renal functional integrity, the enhancement of mitochondrial energy metabolism and antioxidant status in kidney tissue is likely involved in renal protection. Future studies on the biochemical and chemical basis underlying the effects of Chinese Yang/Qi-invigorating tonic botanical drugs/herbal formulations from a mitochondrial perspective will hopefully provide novel insights into the rational development of new drugs for the prevention and/or treatment of CKD.
Collapse
Affiliation(s)
- Jiayi Tian
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, China
- School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, China
| | - Yuqi Huang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, China
- School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, China
| | - Tong Wu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, China
- School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, China
| | - Hsien-Da Huang
- School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, China
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, China
| | - Kam Ming Ko
- Division of Life Science, The Hong Kong University of Science & Technology, Hong Kong, China
| | - Bao Ting Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, China
- School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, China
| | - Jihang Chen
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, China
- School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, China
| |
Collapse
|
19
|
Molaei E, Molaei A, Abedi F, Hayes AW, Karimi G. Nephroprotective activity of natural products against chemical toxicants: The role of Nrf2/ARE signaling pathway. Food Sci Nutr 2021; 9:3362-3384. [PMID: 34136201 PMCID: PMC8194945 DOI: 10.1002/fsn3.2320] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 12/18/2022] Open
Abstract
Nephropathy can occur following exposure of the kidneys to oxidative stress. Oxidative stress is the result of reactive oxygen species (ROS) formation due to intracellular catabolism or exogenous toxicant exposure. Many natural products (NPs) with antioxidant properties have been used to demonstrate that oxidative damage-induced nephrotoxicity can be ameliorated or at least reduced through stimulation of the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway. Nrf2 is a basic leucine zipper (bZip) transcription factor that regulates gene expression of the antioxidant response elements (ARE). Nrf2 is involved in the cellular antioxidant-detoxification machinery. Nrf2 activation is a major mechanism of nephroprotective activity for these NPs, which facilitates its entry into the nucleus, primarily by inhibiting Kelch like-ECH-associated protein 1 (Keap1). The purpose of this article was to review the peer-reviewed literature of NPs that have shown mitigating effects on renal disorder by stimulating Nrf2 and thereby suggesting potential new therapeutic or prophylactic strategies against kidney-damaging xenobiotics.
Collapse
Affiliation(s)
- Emad Molaei
- Faculty of PharmacyMashhad University of Medical SciencesMashhadIran
| | - Ali Molaei
- Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Farshad Abedi
- Faculty of PharmacyMashhad University of Medical SciencesMashhadIran
| | | | - Gholamreza Karimi
- Pharmaceutical Research CenterInstitute of Pharmaceutical TechnologyMashhad University of Medical SciencesMashhadIran
- Department of Pharmacodynamics and ToxicologyFaculty of PharmacyMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
20
|
Gao C, Liu C, Chen Y, Wang Q, Hao Z. Protective effects of natural products against drug-induced nephrotoxicity: A review in recent years. Food Chem Toxicol 2021; 153:112255. [PMID: 33989732 DOI: 10.1016/j.fct.2021.112255] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 04/03/2021] [Accepted: 05/05/2021] [Indexed: 12/17/2022]
Abstract
Drug-induced nephrotoxicity (DIN) is a major cause of kidney damage and is associated with high mortality and morbidity, which limits the clinical use of certain therapeutic or diagnostic agents, such as antineoplastic drugs, antibiotics, immunosuppressive agents, non-steroidal anti-inflammatory drugs (NSAIDs), and contrast agents. However, in recent years, a number of studies have shown that many natural products (NPs), including phytochemicals, various plants extracts, herbal formulas, and NPs derived from animals, confer protective effects against DIN through multi-targeting therapeutic mechanisms, such as inhibition of oxidative stress, inflammation, apoptosis, fibrosis, and necroptosis, regulation of autophagy, maintenance of cell polarity, etc., by regulating multiple signaling pathways and novel molecular targets. In this review, we summarize and discuss the protective effects and mechanisms underlying the action of NPs against DIN found in recent years, which will contribute to the development of promising renal protective agents.
Collapse
Affiliation(s)
- Chen Gao
- Innovation Center for Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Chang Liu
- Innovation Center for Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China; College of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Yuwei Chen
- Innovation Center for Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China; College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, 830052, China
| | - Qingtao Wang
- Innovation Center for Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China; College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, 830052, China
| | - Zhihui Hao
- Innovation Center for Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
21
|
Ramazani Y, Knops N, Berlingerio SP, Adebayo OC, Lismont C, Kuypers DJ, Levtchenko E, van den Heuvel LP, Fransen M. Therapeutic concentrations of calcineurin inhibitors do not deregulate glutathione redox balance in human renal proximal tubule cells. PLoS One 2021; 16:e0250996. [PMID: 33930094 PMCID: PMC8087105 DOI: 10.1371/journal.pone.0250996] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 04/18/2021] [Indexed: 12/16/2022] Open
Abstract
The calcineurin inhibitors (CNI) cyclosporine A and tacrolimus comprise the basis of immunosuppressive regimes in all solid organ transplantation. However, long-term or high exposure to CNI leads to histological and functional renal damage (CNI-associated nephrotoxicity). In the kidney, proximal tubule cells are the only cells that metabolize CNI and these cells are believed to play a central role in the origin of the toxicity for this class of drugs, although the underlying mechanisms are not clear. Several studies have reported oxidative stress as an important mediator of CNI-associated nephrotoxicity in response to CNI exposure in different available proximal tubule cell models. However, former models often made use of supra-therapeutic levels of tissue drug exposure. In addition, they were not shown to express the relevant enzymes (e.g., CYP3A5) and transporters (e.g., P-glycoprotein) for the metabolism of CNI in human proximal tubule cells. Moreover, the used methods for detecting ROS were potentially prone to false positive results. In this study, we used a novel proximal tubule cell model established from human allograft biopsies that demonstrated functional expression of relevant enzymes and transporters for the disposition of CNI. We exposed these cells to CNI concentrations as found in tissue of stable solid organ transplant recipients with therapeutic blood concentrations. We measured the glutathione redox balance in this cell model by using organelle-targeted variants of roGFP2, a highly sensitive green fluorescent reporter protein that dynamically equilibrates with the glutathione redox couple through the action of endogenous glutaredoxins. Our findings provide evidence that CNI, at concentrations commonly found in allograft biopsies, do not alter the glutathione redox balance in mitochondria, peroxisomes, and the cytosol. However, at supra-therapeutic concentrations, cyclosporine A but not tacrolimus increases the ratio of oxidized/reduced glutathione in the mitochondria, suggestive of imbalances in the redox environment.
Collapse
Affiliation(s)
- Yasaman Ramazani
- Laboratory of Pediatric Nephrology, Department of Growth and Regeneration, University of Leuven, Leuven, Belgium
| | - Noël Knops
- Laboratory of Pediatric Nephrology, Department of Growth and Regeneration, University of Leuven, Leuven, Belgium
- Department of Pediatric Nephrology and Solid Organ Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Sante Princiero Berlingerio
- Laboratory of Pediatric Nephrology, Department of Growth and Regeneration, University of Leuven, Leuven, Belgium
| | | | - Celien Lismont
- Laboratory of Peroxisome Biology and Intracellular Communication, Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
| | - Dirk J. Kuypers
- Department of Nephrology and Renal Transplantation and Department of Microbiology, Immunology and Transplantation, University of Leuven, Leuven, Belgium
| | - Elena Levtchenko
- Laboratory of Pediatric Nephrology, Department of Growth and Regeneration, University of Leuven, Leuven, Belgium
- Department of Pediatric Nephrology and Solid Organ Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Lambert P. van den Heuvel
- Laboratory of Pediatric Nephrology, Department of Growth and Regeneration, University of Leuven, Leuven, Belgium
- Translational Metabolic Laboratory and Department of Pediatric Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marc Fransen
- Laboratory of Peroxisome Biology and Intracellular Communication, Department of Cellular and Molecular Medicine, University of Leuven, Leuven, Belgium
- * E-mail:
| |
Collapse
|
22
|
Serrya MS, Nader MA, Abdelmageed ME. Hepatoprotective effect of the tyrosine kinase inhibitor nilotinib against cyclosporine-A induced liver injury in rats through blocking the Bax/Cytochrome C/caspase-3 apoptotic signaling pathway. J Biochem Mol Toxicol 2021; 35:1-13. [PMID: 33710703 DOI: 10.1002/jbt.22764] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 12/07/2020] [Accepted: 03/02/2021] [Indexed: 11/09/2022]
Abstract
Cyclosporine-A (CsA) is a powerful immunosuppressive agent and hepatotoxicity results from CsA treatment. This study aimed to elucidate the effectiveness of tyrosine kinase inhibitor nilotinib against CsA-induced hepatotoxicity and the underlying molecular mechanisms. Male Sprague-Dawley rats were allocated into four groups and received drugs for 28 days as follows: Control group: received vehicle, Nilotinib group: received nilotinib (20 mg/kg orally), CsA group: received CsA by subcutaneous injection (20 mg/kg daily), CsA-nilotinib: received nilotinib and CsA. Serum lactate dehydrogenase (LDH), liver function biomarkers, hepatic levels of oxidative stress biomarkers, nuclear factor erythroid-2 like-2 (Nrf2), total antioxidant capacity (TAC), interleukin-2 (IL-2), IL-1β, IL-6, and cytochrome-C were assessed. Additionally, the protein levels and mRNA expression of Bcl2 associated X protein (Bax), caspase-3, nuclear factor-κB (NF-κB), hemoxygenase-1 (HO-1) were measured. Moreover, liver tissues were assessed histopathologically using hematoxylin-eosin and Masson trichrome stain. Nilotinib treatment decreased serum LDH, alanine aminotransferase, aspartate aminotransferase, and γ-glutamyltransferase (γ-GT), hepatic malondialdehyde, and cytochrome-C. It also increased superoxide dismutase, reduced glutathione, glutathione reductase, glutathione peroxidase, glutathione-S-transferase (GST), TAC, and Nrf2 compared to CsA-injected rats. In addition, nilotinib decreased NF-κB, IL-1β, IL-6, Bax, and caspase-3, while elevated IL-2 and immunoexpression of HO-1. Additionally, mRNA expression of Bax and caspase-3 was elevated and that of HO-1 and inhibitory protein κB-α was reduced in the nilotinib-treated group. Moreover, nilotinib significantly attenuated CsA-induced histopathological alterations. Nilotinib may have a promising role as a hepato-protective through its antiapoptotic, antioxidant, and anti-inflammatory effects.
Collapse
Affiliation(s)
- Marwa S Serrya
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Manar A Nader
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Marwa E Abdelmageed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
23
|
Grunenwald A, Roumenina LT, Frimat M. Heme Oxygenase 1: A Defensive Mediator in Kidney Diseases. Int J Mol Sci 2021; 22:2009. [PMID: 33670516 PMCID: PMC7923026 DOI: 10.3390/ijms22042009] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 12/18/2022] Open
Abstract
The incidence of kidney disease is rising, constituting a significant burden on the healthcare system and making identification of new therapeutic targets increasingly urgent. The heme oxygenase (HO) system performs an important function in the regulation of oxidative stress and inflammation and, via these mechanisms, is thought to play a role in the prevention of non-specific injuries following acute renal failure or resulting from chronic kidney disease. The expression of HO-1 is strongly inducible by a wide range of stimuli in the kidney, consequent to the kidney's filtration role which means HO-1 is exposed to a wide range of endogenous and exogenous molecules, and it has been shown to be protective in a variety of nephropathological animal models. Interestingly, the positive effect of HO-1 occurs in both hemolysis- and rhabdomyolysis-dominated diseases, where the kidney is extensively exposed to heme (a major HO-1 inducer), as well as in non-heme-dependent diseases such as hypertension, diabetic nephropathy or progression to end-stage renal disease. This highlights the complexity of HO-1's functions, which is also illustrated by the fact that, despite the abundance of preclinical data, no drug targeting HO-1 has so far been translated into clinical use. The objective of this review is to assess current knowledge relating HO-1's role in the kidney and its potential interest as a nephroprotection agent. The potential therapeutic openings will be presented, in particular through the identification of clinical trials targeting this enzyme or its products.
Collapse
Affiliation(s)
- Anne Grunenwald
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France; (A.G.); (L.T.R.)
| | - Lubka T. Roumenina
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, F-75006 Paris, France; (A.G.); (L.T.R.)
| | - Marie Frimat
- U1167-RID-AGE, Institut Pasteur de Lille, Inserm, Univ. Lille, F-59000 Lille, France
- Nephrology Department, CHU Lille, Univ. Lille, F-59000 Lille, France
| |
Collapse
|
24
|
Nath M, Agarwal A. New insights into the role of heme oxygenase-1 in acute kidney injury. Kidney Res Clin Pract 2020; 39:387-401. [PMID: 33184238 PMCID: PMC7770992 DOI: 10.23876/j.krcp.20.091] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 12/17/2022] Open
Abstract
Acute kidney injury (AKI) is attended by injury-related biomarkers appearing in the urine and serum, decreased urine output, and impaired glomerular filtration rate. AKI causes increased morbidity and mortality and can progress to chronic kidney disease and end-stage kidney failure. AKI is without specific therapies and is managed by supported care. Heme oxygenase-1 (HO-1) is a cytoprotective, inducible enzyme that degrades toxic free heme released from destabilized heme proteins and, during this process, releases beneficial by-products such as carbon monoxide and biliverdin/bilirubin and promotes ferritin synthesis. HO-1 induction protects against assorted renal insults as demonstrated by in vitro and preclinical models. This review summarizes the advances in understanding of the protection conferred by HO-1 in AKI, how HO-1 can be induced including via its transcription factor Nrf2, and HO-1 induction as a therapeutic strategy.
Collapse
Affiliation(s)
- Meryl Nath
- Deparment of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anupam Agarwal
- Deparment of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.,Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, AL, USA.,Department of Veterans Affairs, Birmingham Veterans Administration Medical Center, Birmingham, AL, USA
| |
Collapse
|
25
|
Zhou Y, Men L, Sun Y, Wei M, Fan X. Pharmacodynamic effects and molecular mechanisms of lignans from Schisandra chinensis Turcz. (Baill.), a current review. Eur J Pharmacol 2020; 892:173796. [PMID: 33345853 DOI: 10.1016/j.ejphar.2020.173796] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/08/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023]
Abstract
Fruit of Schisandra chinensis Turcz. (Baill.) (S. chinensis) is a traditional herbal medicine widely used in China, Korea, and many other east Asian countries. At present, S. chinensis commonly forms Chinese medicinal formulae with other herbal medicines to treat liver disease and neurological disease in clinical. Modern researches indicated that lignans were the main active ingredients of S. chinensis with high content and novel dibenzocyclooctadiene skeletal structure, exhibited considerable antioxidant, anti-inflammatory, and neuroprotective properties. Additionally, some of these lignans also showed certain potentials in anti-cancer, anti-fibrosis, and other effects. In the current review, we summarize literature reported lignans from S. chinensis in the past five years, and highlight the molecular mechanisms of lignans in exerting their biological functions. Also, we point out some deficiencies of existing researches and discuss the future direction of lignans study.
Collapse
Affiliation(s)
- Yuan Zhou
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Lihui Men
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Yunxia Sun
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Mengying Wei
- Natural Medicine Institute of Zhejiang YangShengTang Co., Hangzhou, 310000, China
| | - Xiang Fan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
26
|
Wang Z, Wang L, Luo J, Zhang J. Protection against acute renal injury by naturally occurring medicines which act through Nrf2 signaling pathway. J Food Biochem 2020; 45:e13556. [PMID: 33152804 DOI: 10.1111/jfbc.13556] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/26/2020] [Accepted: 10/20/2020] [Indexed: 12/14/2022]
Abstract
The cellular defense pathway plays a key role in maintaining the homeostasis, tissues and organisms. Nuclear factor E2-related factor 2 (Nrf2), as a key cell signaling pathway, plays an important role in encoding detoxification enzymes and other stress response mediators. Recent studies have shown that it is closely related to the prevention and treatment of acute kidney injury (AKI). Therefore, this article reviews the protective effects of Nrf2-related signaling pathways on acute kidney injury, and summarizes the strategies of natural pharmaceutical ingredients such as flavonoids, alkaloids, terpenes, phenylpropionic acid, polyphenols, and polysaccharides to prevent and treat acute kidney injury. It is of great significance to further study the relationship between Nrf2 regulated signal pathway and kidney disease and the development of new medicines for acute kidney injury treatment. It can also provide new ideas and treatment strategies for clinical treatment of acute kidney injury. PRACTICAL APPLICATIONS: This article reviewed the mechanisms by which the active ingredients of natural medicines slow down acute kidney injury through the Nrf2 pathway. It will help us to understand the regulatory role of the Nrf2 pathway in AKI more comprehensively, and provide a theoretical basis for further exploring the mechanism of more natural drugs to reduce acute kidney injury.
Collapse
Affiliation(s)
- Zhenyi Wang
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, China
| | - Lulu Wang
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, China.,Changchun Institute of Technology School of Medicine, Changchun, China
| | - Jiacheng Luo
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, China
| | - Jing Zhang
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, China.,Changchun Institute of Technology School of Medicine, Changchun, China
| |
Collapse
|
27
|
Teng Q, Ma L, Ma Y, Zhang Y, Kang N, Hu Y, Zhang S. The challenge of managing comorbidities: a case report of primary Sjogren's syndrome in a patient with acute intermittent porphyria. Intractable Rare Dis Res 2020; 9:137-140. [PMID: 32844069 PMCID: PMC7441034 DOI: 10.5582/irdr.2020.03064] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Acute intermittent porphyria (AIP) is a rare inherited metabolic disease associated with heme metabolism. Primary Sjogren's syndrome (PSS) is a common autoimmune disease. The combined presence of AIP and PSS complicates treatment. A rare case of concomitant AIP and PSS is reported in this paper. A 30-year-old woman with AIP had recurrent acute abdominal pain, nausea and vomiting, constipation, persistent chest, back, and waist pain, red urine, positivity for porphobilinogen (PBG) in urine and a pathogenic mutation of the HMBS gene. Two and a half years after she was diagnosed with AIP, she was diagnosed with PSS based on dryness of the eyes and mouth, the elevation of immunoglobulins (IgG and IgA) and positive results on an anti-SS-A antibody test, an anti-SS-B antibody test, Schirmer's test and a labial gland biopsy. A mutation in the HMBS gene was detected in the patient and her cousin, but the patient had more severe AIP and more severe symptoms (such as epilepsy and a limp), which may be related to the co-morbidity of PSS. According to her PSS activity score, the patient had an ESSDAI score of 9 and required systemic treatment. However, potential medications were limited by AIP, so mycophenolate mofetil was eventually added to delay the progression of the primary disease.
Collapse
Affiliation(s)
- Qing Teng
- Department of Endocrinology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Liyan Ma
- Department of Rheumatology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yuelin Ma
- Department of Endocrinology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yiran Zhang
- School of First Clinical Medical College, Southern Medical University, Guangzhou, Guangdong, China
| | - Ninglin Kang
- Hebei Provincial Hospital of Traditional Chinese Medicine, Shijiazhuang, Hebei, China
| | - Yuanxiang Hu
- Geriatrics, First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| | - Songyun Zhang
- Department of Endocrinology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Address correspondence to:Songyun Zhang, Department of Endocrinology, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei, China. E-mail:
| |
Collapse
|
28
|
Lu Y, Li CF, Ping NN, Sun YY, Wang Z, Zhao GX, Yuan SH, Zibrila AI, Soong L, Liu JJ. Hydrogen-rich water alleviates cyclosporine A-induced nephrotoxicity via the Keap1/Nrf2 signaling pathway. J Biochem Mol Toxicol 2020; 34:e22467. [PMID: 32040235 DOI: 10.1002/jbt.22467] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/08/2020] [Accepted: 01/31/2020] [Indexed: 12/12/2022]
Abstract
Oxidative stress induced by long-term cyclosporine A (CsA) administration is a major cause of chronic nephrotoxicity, which is characterized by tubular atrophy, tubular cell apoptosis, and interstitial fibrosis in the progression of organ transplantation. Although hydrogen-rich water (HRW) has been used to prevent various oxidative stress-related diseases, its underlying mechanisms remain unclear. This study investigated the effects of HRW on CsA-induced nephrotoxicity and its potential mechanisms. After administration of CsA (25 mg/kg/day), rats were treated with or without HRW (12 mL/kg) for 4 weeks. Renal function and vascular activity were investigated. Histological changes in kidney tissues were analyzed using Masson's trichrome and terminal deoxynucleotidyl transferase dUTP nick-end labeling stains. Oxidative stress markers and the activation of the Kelch-like ECH-associated protein 1 (Keap1)/nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway were also measured. We found that CsA increased the levels of reactive oxygen species (ROS) and malonaldehyde (MDA), but it reduced glutathione (GSH) and superoxide dismutase (SOD) levels. Such alterations induced vascular dysfunction, tubular atrophy, interstitial fibrosis, and tubular apoptosis. This was evident secondary to an increase in urinary protein, serum creatinine, and blood urea nitrogen, ultimately leading to renal dysfunction. Conversely, HRW decreased levels of ROS and MDA while increasing the activity of GSH and SOD. This was accompanied by an improvement in vascular and renal function. Moreover, HRW significantly decreased the level of Keap1 and increased the expression of Nrf2, NADPH dehydrogenase quinone 1, and heme oxygenase 1. In conclusion, HRW restored the balance of redox status, suppressed oxidative stress damage, and improved kidney function induced by CsA via activation of the Keap1/Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Yi Lu
- Key Laboratory of Shannxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Clinical Research Center of Shannxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Department of Pharmacy, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Chun-Fang Li
- Department of Obstetrics and Gynaecology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Na-Na Ping
- Department of Blood Component, Shaanxi Blood Center, Xi'an, Shaanxi, China
| | - Yu-Yao Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xian Jiaotong University, Xi'an, Shaanxi, China
| | - Zheng Wang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Gong-Xiao Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xian Jiaotong University, Xi'an, Shaanxi, China
| | - Shi-Hui Yuan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xian Jiaotong University, Xi'an, Shaanxi, China
| | - Abdoulaye Issotina Zibrila
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xian Jiaotong University, Xi'an, Shaanxi, China
| | - Lynn Soong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Jin-Jun Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xian Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
29
|
Schizandrin Protects against OGD/R-Induced Neuronal Injury by Suppressing Autophagy: Involvement of the AMPK/mTOR Pathway. Molecules 2019; 24:molecules24193624. [PMID: 31597329 PMCID: PMC6804185 DOI: 10.3390/molecules24193624] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 09/25/2019] [Accepted: 10/06/2019] [Indexed: 01/11/2023] Open
Abstract
The neuroprotective role of schizandrin (SA) in cerebral ischemia-reperfusion (I/R) was recently highlighted. However, whether SA plays a regulatory role on autophagy in cerebral I/R injury is still unclear. This study aimed to explore whether the neuroprotective mechanisms of SA were linked to its regulation of AMP-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR)/autophagy pathway in vivo and in vitro. The present study confirmed that SA significantly improved oxygen-glucose deprivation/re-oxygenation (OGD/R)-induced PC12 cells injury. The results of immunoblotting and confocal microscope showed that SA decreased autophagy in OGD/R-injured PC12 cells, which was reflected by the decreased Beclin-1 and LC3-II expression, autophagy flux level, and LC3 puncta formation. In addition, the autophagy inducer rapamycin partially prevented the effects of SA on cell viability and autophagy after OGD/R, whereas the autophagy inhibitor 3-methyladenine (3-MA) exerted the opposite effect. The results of Western blotting showed that SA markedly decreased the phosphorylation of AMPK (p-AMPK), whereas the phosphor-mTOR (p-mTOR) levels increased in the presence of OGD/R insult. Furthermore, pretreatment with the AMPK inducer AICAR partially reversed the protective effects and autophagy inhibition of SA. However, AMPK inhibitor Compound C pretreatment further promoted the inhibition of SA on autophagy induction and cell damage induced by OGD/R. Taken together, these findings demonstrate that SA protects against OGD/R insult by inhibiting autophagy through the regulation of the AMPK-mTOR pathway and that SA may have therapeutic value for protecting neurons from cerebral ischemia.
Collapse
|
30
|
Molecular Interactions Between Reactive Oxygen Species and Autophagy in Kidney Disease. Int J Mol Sci 2019; 20:ijms20153791. [PMID: 31382550 PMCID: PMC6696055 DOI: 10.3390/ijms20153791] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 12/11/2022] Open
Abstract
Reactive oxygen species (ROS) are highly reactive signaling molecules that maintain redox homeostasis in mammalian cells. Dysregulation of redox homeostasis under pathological conditions results in excessive generation of ROS, culminating in oxidative stress and the associated oxidative damage of cellular components. ROS and oxidative stress play a vital role in the pathogenesis of acute kidney injury and chronic kidney disease, and it is well documented that increased oxidative stress in patients enhances the progression of renal diseases. Oxidative stress activates autophagy, which facilitates cellular adaptation and diminishes oxidative damage by degrading and recycling intracellular oxidized and damaged macromolecules and dysfunctional organelles. In this review, we report the current understanding of the molecular regulation of autophagy in response to oxidative stress in general and in the pathogenesis of kidney diseases. We summarize how the molecular interactions between ROS and autophagy involve ROS-mediated activation of autophagy and autophagy-mediated reduction of oxidative stress. In particular, we describe how ROS impact various signaling pathways of autophagy, including mTORC1-ULK1, AMPK-mTORC1-ULK1, and Keap1-Nrf2-p62, as well as selective autophagy including mitophagy and pexophagy. Precise elucidation of the molecular mechanisms of interactions between ROS and autophagy in the pathogenesis of renal diseases may identify novel targets for development of drugs for preventing renal injury.
Collapse
|
31
|
El-Sheikh AAK, Morsy MA, Abdel-Latif RG. Modulation of eNOS/iNOS by nebivolol protects against cyclosporine A-mediated nephrotoxicity through targeting inflammatory and apoptotic pathways. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2019; 69:26-35. [PMID: 30927701 DOI: 10.1016/j.etap.2019.03.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 03/18/2019] [Accepted: 03/19/2019] [Indexed: 06/09/2023]
Abstract
The aim of this study was to investigate effect of nitric oxide (NO) modulation on possible nephroprotective mechanisms of nebivolol (NEB) in cyclosporine A (CsA)-induced nephrotoxicity. Rats were treated with 20 mg/kg/day s.c. of CsA for 21 days, with NEB alone (10 mg/kg/day orally) or together with a NOS inhibitor, L-NAME (10 mg/kg/day i.p.). NEB conferred nephroprotection against CsA-induced toxicity, significantly decreasing serum kidney function tests and improving renal histopathology. NEB showed antioxidant effects, by significantly decreasing renal malondialdehyde levels, while increasing reduced glutathione levels and catalase activity. NEB showed anti-inflammatory and anti-apoptotic effects; reducing renal expression NF-κB and fas ligand. NEB also reversed CsA-induced effects on NO system; increasing renal NO level, with up-regulation of eNOS and down-regulation of iNOS expression. Administering L-NAME with NEB reversed all beneficial effects of NEB. Thus, NEB's modulation of NO system in CsA-induced nephrotoxicity might be the triggering mechanism controlling NEB's nephroprotective effect.
Collapse
Affiliation(s)
- Azza A K El-Sheikh
- Basic Health Sciences Department, Faculty of Medicine, Princess Nourah bint Abdulrahman University, 11671 Riyadh, Saudi Arabia; Department of Pharmacology, Faculty of Medicine, Minia University, 61511 El-Minia, Egypt.
| | - Mohamed A Morsy
- Department of Pharmacology, Faculty of Medicine, Minia University, 61511 El-Minia, Egypt; Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, 31982 Al-Ahsa, Saudi Arabia
| | - Rania G Abdel-Latif
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, 61511 El-Minia, Egypt
| |
Collapse
|
32
|
Wu Q, Kuca K. Metabolic Pathway of Cyclosporine A and Its Correlation with Nephrotoxicity. Curr Drug Metab 2019; 20:84-90. [DOI: 10.2174/1389200219666181031113505] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 10/16/2018] [Accepted: 10/22/2018] [Indexed: 12/15/2022]
Abstract
Background:Cyclosporine A (CsA) is widely used for organ transplantation and autoimmune disorders. However, CsA nephrotoxicity is a serious side effect that limits the clinical use of CsA. The metabolism of CsA has a close relationship with this disease in renal-transplant patients. However, the metabolic pathways of CsA and its metabolizing enzymes have rarely been comprehensively reviewed. In this review, we have summarized the specific metabolic profiles of CsA in humans, especially renal-transplant patients. Moreover, the specific metabolizing enzymes and the potential roles that CsA metabolism plays in CsA nephrotoxicity were summarized and discussed.Methods:Electronic databases including PubMed, Web of Science, and Scifinder were searched with the keywords "Cyclosporine A and metabolism", and "Cyclosporine A and nephrotoxicity", "Cyclosporine A metabolism and nephrotoxicity". All these studies published until 2018 were included in this review.Results:The major metabolic pathways of CsA in humans are hydroxylation and N-demethylation. Normally, these metabolites are relatively less toxic than CsA. However, the metabolism of CsA in the kidneys is much weaker than that in the liver, which explains why CsA is so toxic to the kidneys. CYP3A families, especially CYP3A4 and CYP3A5, play an important role in the biotransformation of CsA. Moreover, increased lines of evidence show that some metabolites (including AM19) associate directly with nephrotoxicity in CsA-treated organ-transplant patients.Conclusion:The findings of this review help to further understand the metabolic activities of CsA in renal-transplant patients and cast some light on the mechanisms of CsA nephrotoxicity.
Collapse
Affiliation(s)
- Qinghua Wu
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
33
|
El-Bassossy HM, Hassanien MA, Bima A, Ghoneim FM, Elsamanoudy AZ. Renal Oxidative Stress and Inflammatory Response in Perinatal Cyclosporine-A Exposed Rat Progeny and its Relation to Gender. J Microsc Ultrastruct 2019; 7:44-49. [PMID: 31008055 PMCID: PMC6442325 DOI: 10.4103/jmau.jmau_52_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Background and Aim of the Work: The current study postulated that cyclosporine A (CSA) could induce gender-specific renal damage. Hence, the current study aims to investigate the nephrotoxic effect of perinatal exposure of male and female rat progeny to CSA. Moreover, it aims to evaluate the oxidative stress and inflammation as a possible pathophysiologic mechanism. Materials and Methods: Female rats were randomly allocated to two groups of four and assigned to undergo either CSA (15 mg/kg/day; the 6th day after conception and continuing until the progeny were weaned) or vehicle treatment as control groups. At the age of 6 weeks, the progeny were divided into the following four groups: male progeny of control-group mothers (M-vehicle, 7); male progeny of CSA-treated mothers (M-CSA, 9); female progeny of control-group mothers (F-vehicle, 7); and female progeny of CSA-treated mothers (F-CSA, 6). Serum adiponectin, tumor necrosis factor-α (TNF-α) and creatinine, creatinine clearance, and urinary 8-isoprostane were measured. Histopathological examination by hematoxylin and eosin stain of Kidney was carried out. Results: Proteinuria and decreased creatinine clearance are significant in M-CSA than M-vehicle and F-CSA. 8-isoprostane is lower in F-CSA than F-vehicle. Increased TNF-α and decreased adiponectin levels in M-CSA than M-vehicle were observed. No significant differences were found in female rat groups. Conclusion: From the current study, it could be concluded that CSA could induce renal inflammation as well as oxidative stress that may explain the impaired renal function. The sex difference was a prominent finding in their vulnerability to CSA effects.
Collapse
Affiliation(s)
- Hany M El-Bassossy
- Department of Pharmacology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Pharmacology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Mohammed A Hassanien
- Assessment Centre and Medical Education Department, Fakeeh College for Medical Sciences, Jeddah, Saudi Arabia.,Department of Medical Biochemistry, College of Medicine, Tanta University, Tanta, Egypt
| | - Abdulhadi Bima
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Fatma M Ghoneim
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ayman Zaky Elsamanoudy
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
34
|
Kortesoja M, Karhu E, Olafsdottir ES, Freysdottir J, Hanski L. Impact of dibenzocyclooctadiene lignans from Schisandra chinensis on the redox status and activation of human innate immune system cells. Free Radic Biol Med 2019; 131:309-317. [PMID: 30578916 DOI: 10.1016/j.freeradbiomed.2018.12.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/05/2018] [Accepted: 12/17/2018] [Indexed: 12/15/2022]
Abstract
Redox signaling has been established as an essential component of inflammatory responses, and redox active compounds are of interest as potential immunomodulatory agents. Dibenzocyclooctadiene lignans isolated from Schisandra chinensis, a medicinal plant with widespread use in oriental medicine, have been implicated to possess immunomodulatory properties but their effects on the human innate immune system cells have not been described. In this contribution, data are presented on the impact of schisandrin, schisandrin B and schisandrin C on human monocytic cell redox status, as well as their impact on dendritic cell maturation and T cell activation capacity and cytokine production. In THP-1 cells, levels of intracellular reactive oxygen species (ROS) were elevated after 1 h exposure to schisandrin. Schisandrin B and schisandrin C decreased cellular glutathione pools, which is a phenotype previously reported to promote anti-inflammatory functions. Treatment of human primary monocytes with the lignans during their maturation to dendritic cells did not have any effect on the appearance of surface markers HLA-DR and CD86 but schisandrin B and schisandrin C suppressed the secretion of cytokines interleukin (IL)-6, IL-10 and IL-12 by the mature dendritic cells. Dendritic cells maturated in presence of schisandrin C were further cocultured with naïve CD4+ T cells, resulting in reduced IL-12 production. In THP-1 cells, schisandrin B and schisandrin C reduced the IL-6 and IL-12 production triggered by E. coli lipopolysaccharide and IL-12 production induced by an infection with Chlamydia pneumoniae. In conclusion, the studied lignans act as immunomodulatory agents by altering the cytokine secretion, but do not interfere with dendritic cell maturation. And the observed effects may be associated with the ability of the lignans to alter cellular redox status.
Collapse
Affiliation(s)
- Maarit Kortesoja
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 University of Helsinki, Finland
| | - Elina Karhu
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 University of Helsinki, Finland
| | - Elin Soffia Olafsdottir
- Faculty of Pharmaceutical Sciences, University of Iceland, Reykjavik, Hofsvallagata 53, 107 Reykjavik, Iceland
| | - Jona Freysdottir
- Department of Immunology and Center for Rheumatology Research, Landspitali-The National University Hospital of Iceland and Faculty of Medicine, University of Iceland, Eiriksgata, 101 Reykjavik, Iceland
| | - Leena Hanski
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 University of Helsinki, Finland.
| |
Collapse
|
35
|
Gao Y, Wu S, Cong R, Xiao J, Ma F. Characterization of lignans in Schisandra chinensis oil with a single analysis process by UPLC-Q/TOF-MS. Chem Phys Lipids 2019; 218:158-167. [PMID: 30610837 DOI: 10.1016/j.chemphyslip.2018.12.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/04/2018] [Accepted: 12/27/2018] [Indexed: 10/27/2022]
Abstract
Schisandra chinensis is a medicinal and edible plant that contains various bioactive compounds. Among these, lignans are the major functional compounds. Nevertheless, detailed information about lignans in Schisandra chinensis oil remains scarce. A powerful UPLC-Q/TOF-MS method was established for the rapid identification of the lignan constituents of Schisandra chinensis oils. The results showed that 21 lignans have been unambiguously identified, and four lignans have been tentatively identified in the Schisandra chinensis oils. In addition, semi-quantitative analysis indicated that the total lignan content in the Schisandra chinensis oils was distributed from 67.73 ± 0.06 to 87.61 ± 1.83 mg/g. Schisandrin and schisandrin B were the most abundant lignans in the Schisandra chinensis oils, their content ranging from 15.85 ± 0.09 to 20.57 ± 0.38 mg/g. Additionally, this study provided a systematic characterization of lignans in Schisandra chinensis oil and indicated that the oil might be used as lignan-related functional foods.
Collapse
Affiliation(s)
- Yuan Gao
- Department of Food Science and Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China; Key Laboratory of Urban Agriculture (South), Ministry of Agriculture, 800 Dongchuan Road, Shanghai, 200240, China
| | - Shimin Wu
- Department of Food Science and Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China; Key Laboratory of Urban Agriculture (South), Ministry of Agriculture, 800 Dongchuan Road, Shanghai, 200240, China.
| | - Renhuai Cong
- Research & Development Centre, Infinitus (China) Company Ltd., 19 Sicheng Road, Guangzhou, 510663, China
| | - Junyong Xiao
- Research & Development Centre, Infinitus (China) Company Ltd., 19 Sicheng Road, Guangzhou, 510663, China
| | - Fangli Ma
- Research & Development Centre, Infinitus (China) Company Ltd., 19 Sicheng Road, Guangzhou, 510663, China.
| |
Collapse
|
36
|
Gu X, Jiang Y, Chen J, Zhang Y, Guan M, Li X, Zhou Q, Lu Q, Qiu J, Yin X. Synthesis and biological evaluation of bifendate derivatives bearing acrylamide moiety as novel antioxidant agents. Eur J Med Chem 2019; 162:59-69. [DOI: 10.1016/j.ejmech.2018.11.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 11/01/2018] [Indexed: 12/29/2022]
|
37
|
PIG11 over-expression predicts good prognosis and induces HepG2 cell apoptosis via reactive oxygen species-dependent mitochondrial pathway. Biomed Pharmacother 2018; 108:435-442. [DOI: 10.1016/j.biopha.2018.09.062] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 09/11/2018] [Accepted: 09/11/2018] [Indexed: 12/20/2022] Open
|
38
|
Ding M, Shu P, Gao S, Wang F, Gao Y, Chen Y, Deng W, He G, Hu Z, Li T. Schisandrin B protects human keratinocyte-derived HaCaT cells from tert-butyl hydroperoxide-induced oxidative damage through activating the Nrf2 signaling pathway. Int J Mol Med 2018; 42:3571-3581. [PMID: 30272282 DOI: 10.3892/ijmm.2018.3901] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 09/06/2018] [Indexed: 11/05/2022] Open
Abstract
Schisandrin B (Sch B), an active extract of Schisandra chinensis, has demonstrated antioxidant activity in a number of in vitro and in vivo models. In the present study, the capacity of Sch B to protect against oxidative injury in keratinocytes using the human keratinocyte‑derived HaCaT cell line was investigated. To induce oxidative injury, tert‑Butyl hydroperoxide (tBHP) was employed. The results indicate that Sch B efficiently reduced tBHP‑induced cell death, reactive oxygen species (ROS) generation, protein oxidation, lipid peroxidation and DNA damage. Sch B also effectively attenuated the loss of mitochondrial membrane potential (MMP), and restored adenosine triphosphate (ATP) levels in tBHP‑injured HaCaT cells. Furthermore, Sch B enhanced the expression of key antioxidant enzymes, including catalase, heme oxygenase‑1, glutathione peroxidase, and superoxide dismutase, and further engaged the nuclear factor‑erythroid 2‑related factor 2 (Nrf2) signaling pathway by modulating its phosphorylation through activating multiple upstream kinases, including protein kinase B, adenosine monophosphate‑activated protein kinase and mitogen‑activated protein kinases (MAPKs). The present study suggests that Sch B provides a protective effect in keratinocytes in response to oxidative injury via reinforcing the endogenous antioxidant defense system. Therefore, it may be applied as an adjuvant therapy or in health foods to delay the skin aging process and the onset of skin diseases caused by oxidative stress.
Collapse
Affiliation(s)
- Ming Ding
- School of Chemistry and Pharmaceutical Engineering, Qilu University of Technology, Jinan, Shandong 250353, P.R. China
| | - Peng Shu
- Infinitus (China) Company, Ltd., Guangzhou, Guangdong 510663, P.R. China
| | - Shuang Gao
- Institute of Life Science, Wenzhou University, Wenzhou, Zhejiang 325000, P.R. China
| | - Fenglou Wang
- LB Cosmeceutical Technology Co., Ltd., Shanghai 200233, P.R. China
| | - Yitian Gao
- Institute of Life Science, Wenzhou University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yu Chen
- Institute of Life Science, Wenzhou University, Wenzhou, Zhejiang 325000, P.R. China
| | - Wenjuan Deng
- Infinitus (China) Company, Ltd., Guangzhou, Guangdong 510663, P.R. China
| | - Gaiying He
- LB Cosmeceutical Technology Co., Ltd., Shanghai 200233, P.R. China
| | - Zhenlin Hu
- Institute of Life Science, Wenzhou University, Wenzhou, Zhejiang 325000, P.R. China
| | - Tianduo Li
- School of Chemistry and Pharmaceutical Engineering, Qilu University of Technology, Jinan, Shandong 250353, P.R. China
| |
Collapse
|
39
|
Eberhardt W, Nasrullah U, Pfeilschifter J. Activation of renal profibrotic TGFβ controlled signaling cascades by calcineurin and mTOR inhibitors. Cell Signal 2018; 52:1-11. [PMID: 30145216 DOI: 10.1016/j.cellsig.2018.08.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 08/21/2018] [Accepted: 08/22/2018] [Indexed: 12/11/2022]
Abstract
The calcineurin inhibitors (CNI) cyclosporine A (CsA) and tacrolimus represent potent immunosuppressive agents frequently used for solid organ transplantation and treatment of autoimmune disorders. Despite of their immense therapeutic benefits, residual fibrosis mainly in the kidney represents a common side effect of long-term therapy with CNI. Regardless of the immunosuppressive action, an increasing body of evidence implicates that a drug-induced increase in TGFβ and subsequent activation of TGFβ-initiated signaling pathways is closely associated with the development and progression of CNI-induced nephropathy. Mechanistically, an increase in reactive oxygen species (ROS) generation due to drug-induced changes in the intracellular redox homeostasis functions as an important trigger of the profibrotic signaling cascades activated under therapy with CNI. Although, inhibitors of the mechanistic target of rapamycin (mTOR) kinase have firmly been established as alternative compounds with a lower nephrotoxic potential, an activation of fibrogenic signaling cascades has been reported for these drugs as well. This review will comprehensively summarize recent advances in the understanding of profibrotic signaling events modulated by these widely used compounds with a specific focus put on mechanisms occurring independent of their respective immunosuppressive action. Herein, the impact of redox modulation, the activation of canonical TGFβ and non-Smad pathways and modulation of autophagy by both classes of immunosuppressive drugs will be highlighted and discussed in a broader perspective. The comprehensive knowledge of profibrotic signaling events specifically accompanying the immunomodulatory activity of these widely used drugs is needed for a reliable benefit-risk assessment under therapeutic regimens.
Collapse
Affiliation(s)
- Wolfgang Eberhardt
- Pharmazentrum frankfurt/ZAFES, Universitätsklinikum und Goethe-Universität, Frankfurt am Main, Germany.
| | - Usman Nasrullah
- Pharmazentrum frankfurt/ZAFES, Universitätsklinikum und Goethe-Universität, Frankfurt am Main, Germany
| | - Josef Pfeilschifter
- Pharmazentrum frankfurt/ZAFES, Universitätsklinikum und Goethe-Universität, Frankfurt am Main, Germany
| |
Collapse
|
40
|
Mechanism of cyclosporine A nephrotoxicity: Oxidative stress, autophagy, and signalings. Food Chem Toxicol 2018; 118:889-907. [DOI: 10.1016/j.fct.2018.06.054] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 06/21/2018] [Accepted: 06/22/2018] [Indexed: 12/16/2022]
|