1
|
Shang D, Chen Y, Sun D. Umbilical cord-derived mesenchymal stem cells combined with kaempferol synergistically promote repair of damaged endometrium by modulating JAK2/STAT3 signaling pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04056-4. [PMID: 40266305 DOI: 10.1007/s00210-025-04056-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 03/13/2025] [Indexed: 04/24/2025]
Abstract
Intrauterine adhesion (IUA) is a disease caused by endometrial damage without effective treatments. Stem cell therapy has been initiated as a new attempt to repair and regenerate injured tissues. However, the therapeutic efficacy of stem cell therapy is limited. Kaempferol is a natural flavonoid with various beneficial effects. In this study, our goal is to investigate the roles of kaempferol combined with umbilical cord-derived mesenchymal stem cells (UCMSCs) in IUA. UCMSCs were collected from human umbilical cords. The multilineage differentiation potential of human UCMSCs was evaluated by Oil Red O staining, Alizarin Red S staining, and Alcian Blue staining. The phenotype profile of human UCMSCs was assessed by flow cytometry. CCK-8 and Transwell assays were performed to detect cell viability and migration. The IUA rat model was established. Histological changes were examined by hematoxylin-eosin staining and Masson staining. Gene expression was evaluated by western blotting and immunofluorescence. Kaempferol (10 μM) promoted the migration and proliferation of human UCMSCs in vitro. Additionally, kaempferol/UCMSCs combination treatment recovered endometrial injury and inhibited endometrial fibrosis and epithelial-mesenchymal transition occurrence in IUA rat models. Mechanistically, kaempferol/UCMSCs combination treatment inhibited JAK2/STAT3 pathway. Kaempferol/UCMSCs combination treatment can promote the repair of damaged endometrium by decreasing endometrial fibrosis. The inactivation of JAK2/STAT3 pathway is greatly responsible for the protective effect of kaempferol/UCMSCs combination treatment.
Collapse
Affiliation(s)
- Di Shang
- Ultrasound Diagnosis Department, Hubei Maternal and Child Health Hospital, Wuhan, 430070, China
| | - Yuru Chen
- Training Base of Hubei Maternal and Child Health Hospital, Hubei University of Medicine, Wuhan, 442000, China
| | - Dongyan Sun
- Department of Medical Affairs, Hubei Maternal and Child Health Hospital, No.745 Wuluo Road, Hongshan District, Wuhan, 430070, China.
| |
Collapse
|
2
|
Zhang X, Xie Y, Cai Y, Huang H, Liang H, Liao G, Jiang Y, Peng X, Zhan S, Huang X. RNA-seq analysis and in vivo experiments identified the protective effect of kaempferol on idiopathic pulmonary fibrosis by regulating the PPARG/TNC signaling pathway to reduce ECM deposition. Food Funct 2024; 15:12193-12209. [PMID: 39587935 DOI: 10.1039/d4fo01474j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic age-related lung disease with a high mortality rate. Kaempferol (KMP), an active ingredient in common plants and foods with anti-inflammatory, antioxidant and immunomodulatory properties, has been shown to be effective against fibrotic diseases. However, the molecular mechanisms underlying the treatment of IPF with KMP remain unclear. Therefore, IPF mice were established by intratracheal instillation of bleomycin (BLM) to explore the efficacy and underlying mechanism of KMP in the treatment of IPF. We found that KMP improved the body weight changes of BLM-induced IPF mice, alleviated inflammatory infiltration and collagen deposition, and decreased the expression levels of hydroxyproline, α-SMA, Col3a1, Mmp2, Timp1, Vim, Fn, TNF-α, TGF-β1, IL-6 and IL-8, while up-regulating the expression E-cadherin in lung tissues. The transcriptomic results showed that KMP may exert therapeutic effects against IPF by regulating the PPARG/TNC signaling pathway to reduce extracellular matrix (ECM) deposition. Interestingly, ROC curve analysis suggested that TNC and PPARG had good diagnostic performance for IPF, and TF prediction revealed that PPARG is an important upstream gene regulating TNC, and the IF experiment confirmed the co-localization of TNC and PPARG. Molecular docking showed that KMP bound well to PPARG and TNC, and IF results revealed that KMP significantly reduced the interaction between PPARG and TNC. Furthermore, RT-PCR, WB, IHC and IF experiments confirmed that KMP elevated the expression of PPARG and inhibited the expression of TNC, thus inhibiting the ECM-receptor interaction pathway and ultimately serving as a therapeutic treatment for IPF mice. These findings revealed that KMP reduced inflammatory infiltration and collagen deposition in the lungs of IPF mice and that the PPARG/TNC signaling pathway may be an important mechanism for the treatment of IPF with KMP, which provides a new perspective for the development of therapeutic approaches for IPF.
Collapse
Affiliation(s)
- Xinxin Zhang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China.
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Centre of Guangzhou University of Chinese Medicine, Guangzhou, China.
- Guangdong Provincial Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Yizi Xie
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China.
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Centre of Guangzhou University of Chinese Medicine, Guangzhou, China.
- Guangdong Provincial Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Yan Cai
- Guangdong Provincial Hospital of Chinese Medicine, Zhuhai, China
| | - Huiting Huang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Huiqiu Liang
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, China.
| | - Gang Liao
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, China.
| | - Yong Jiang
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, China.
| | - Xiaoyun Peng
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, China.
| | - Shaofeng Zhan
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Xiufang Huang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China.
- Lingnan Medical Research Centre of Guangzhou University of Chinese Medicine, Guangzhou, China.
- Guangdong Provincial Clinical Research Academy of Chinese Medicine, Guangzhou, China
| |
Collapse
|
3
|
Liang L, Mi Y, Zhou S, Yang A, Wei C, Dai E. Advances in the study of key cells and signaling pathways in renal fibrosis and the interventional role of Chinese medicines. Front Pharmacol 2024; 15:1403227. [PMID: 39687302 PMCID: PMC11647084 DOI: 10.3389/fphar.2024.1403227] [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: 03/19/2024] [Accepted: 10/21/2024] [Indexed: 12/18/2024] Open
Abstract
Renal fibrosis (RF) is a pathological process characterized by the excessive accumulation of extracellular matrix (ECM), which triggers a repair cascade in response to stimuli and pathogenic factors, leading to the activation of molecular signaling pathways involved in fibrosis. This article discusses the key cells, molecules, and signaling pathways implicated in the pathogenesis of RF, with a particular focus on tubular epithelial cells (TECs), cellular senescence, ferroptosis, autophagy, epithelial-mesenchymal transition (EMT), and transforming growth factor-β(TGF-β)/Smad signaling. These factors drive the core and regulatory pathways that significantly influence RF. A comprehensive understanding of their roles is essential. Through a literature review, we explore recent advancements in traditional Chinese medicine (TCM) aimed at reducing RF and inhibiting chronic kidney disease (CKD). We summarize, analyze, and elaborate on the important role of Chinese herbs in RF, aiming to provide new directions for their application in prevention and treatment, as well as scientific guidance for clinical practices.
Collapse
Affiliation(s)
- Lijuan Liang
- Gansu University of Chinese Medicine, Lanzhou, China
- Key Laboratory of Dunhuang Medicine and Translation, Ministry of Education, Lanzhou, China
| | - Youjun Mi
- Institute of pathophysiology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Shihan Zhou
- Gansu University of Chinese Medicine, Lanzhou, China
| | - Aojian Yang
- Gansu University of Chinese Medicine, Lanzhou, China
| | - Chaoyu Wei
- Gansu University of Chinese Medicine, Lanzhou, China
| | - Enlai Dai
- Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
4
|
Alkandahri MY, Sadino A, Pamungkas BT, Oktoba Z, Arfania M, Yuniarsih N, Wahyuningsih ES, Dewi Y, Winarti SA, Dinita ST. Potential Nephroprotective Effect of Kaempferol: Biosynthesis, Mechanisms of Action, and Clinical Prospects. Adv Pharmacol Pharm Sci 2024; 2024:8907717. [PMID: 39377015 PMCID: PMC11458287 DOI: 10.1155/2024/8907717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 08/16/2024] [Accepted: 09/10/2024] [Indexed: 10/09/2024] Open
Abstract
Kidney is an essential organ that is highly susceptible to cellular injury caused by various toxic substances in the blood. Several studies have shown that untreated injuries to this organ can cause glomerulosclerosis, tubulointerstitial fibrosis, and tubular cell apoptosis, leading to kidney failure. Despite significant advancements in modern treatment, there is no fully effective drug for repairing its function, providing complete protection, and assisting in cell regeneration. Furthermore, some available medications have been reported to exacerbate injuries, showing the need to explore alternative treatments. Natural drugs are currently being explored as a new therapeutic strategy for managing kidney diseases. Kaempferol, a polyphenol found in plants, including vegetables, legumes, and fruits, has been extensively studied in various nephrotoxicity protocols. The compound has been reported to have potential as a nephroprotective agent with beneficial effects on various physiological pathways, such as CPL-induced kidney injury, DOX, LPO, ROS, RCC, and diabetic nephropathy. Therefore, this study aims to provide a brief overview of the current nephroprotective effects of kaempferol, as well as its molecular mechanisms of action, biosynthesis pathways, and clinical prospects.
Collapse
Affiliation(s)
- Maulana Yusuf Alkandahri
- Department of Pharmacology and Clinical PharmacyFaculty of PharmacyUniversitas Buana Perjuangan Karawang, Karawang, West Java, Indonesia
| | - Asman Sadino
- Department of PharmacyFaculty of Mathematics and Natural ScienceUniversitas Garut, Garut, West Java, Indonesia
| | - Barolym Tri Pamungkas
- Department of Pharmaceutical BiologyFaculty of PharmacyUniversitas Mulawarman, Samarinda, East Kalimantan, Indonesia
| | - Zulpakor Oktoba
- Department of PharmacyFaculty of MedicineUniversitas Lampung, Bandar Lampung, Indonesia
| | - Maya Arfania
- Department of Pharmacology and Clinical PharmacyFaculty of PharmacyUniversitas Buana Perjuangan Karawang, Karawang, West Java, Indonesia
| | - Nia Yuniarsih
- Department of Pharmaceutical TechnologyFaculty of PharmacyUniversitas Buana Perjuangan Karawang, Karawang, West Java, Indonesia
| | - Eko Sri Wahyuningsih
- Department of Pharmaceutical BiologyFaculty of PharmacyUniversitas Buana Perjuangan Karawang, Karawang, West Java, Indonesia
| | - Yuliani Dewi
- Faculty of PharmacyUniversitas Buana Perjuangan Karawang, Karawang, West Java, Indonesia
| | - Sri Ayu Winarti
- Faculty of PharmacyUniversitas Buana Perjuangan Karawang, Karawang, West Java, Indonesia
| | - Sri Tantia Dinita
- Faculty of PharmacyUniversitas Buana Perjuangan Karawang, Karawang, West Java, Indonesia
| |
Collapse
|
5
|
Miao J, Wei C, Wang HL, Li YQ, Yu XM, Yang X, Su HW, Li P, Wang L. Mechanism of Chaihuang-Yishen formula to attenuate renal fibrosis in the treatment of chronic kidney disease: Insights from network pharmacology and experimental validation. Heliyon 2024; 10:e35728. [PMID: 39220918 PMCID: PMC11365344 DOI: 10.1016/j.heliyon.2024.e35728] [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: 04/10/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024] Open
Abstract
Renal fibrosis represents a pivotal characteristic of chronic kidney disease (CKD), for which effective interventions are currently lacking. The Src kinase activates the phosphatidylinositol-3 kinases (PI3K)/Akt1 pathway to promote renal fibrosis, casting a promising target for anti-fibrosis treatment. Chaihuang-Yishen formula (CHYS), a traditional Chinese medicinal prescription, has a validated efficacy in the treatment of CKD, however, with the underlying mechanism unresolved. This study aimed to uncover the pharmacological mechanisms mediating the effect of CHYS in treating renal fibrosis using network pharmacology followed by experimental validation. The chemical compounds of CHYS were retrieved from the Traditional Chinese Medicine Systems Pharmacology (TCMSP) database or published literature, followed by the prediction of their targets using SwissTargetPrediction software. Disease (CKD/renal fibrosis)-related targets were retrieved from the Genecards database. Protein-protein interaction (PPI) network was generated using the drug-disease common targets and visualized in Cytoscape software. The drug-disease targets were further subjected to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses by Metascape software. Additionally, the compound-target-pathway network was established in Cytosape to identify key compounds, targets, and pathways. Network pharmacology analysis screened out 96 active compounds and 837 potential targets within the 7 herbal/animal medicines of CHYS, among which 237 drug-disease common targets were identified. GO and KEGG analysis revealed the enrichment of fibrosis-related biological processes and pathways among the 237 common targets. Compound-target-pathway network analysis highlighted protein kinases Src and Akt1 as the top two targets associated with the anti-renal fibrosis effects of CHYS. In UUO mice, treatment with CHYS attenuates renal fibrosis, accompanied by suppressed expression and phosphorylation activation of Src. Unlike Src, CHYS reduced Akt1 phosphorylation without affecting its expression. In summary, network pharmacology and in vivo evidence suggest that CHYS exerts its anti-renal fibrosis effects, at least in part, by inhibiting the Src/Akt1 signaling axis.
Collapse
Affiliation(s)
- Jie Miao
- College of Integrated Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Cong Wei
- The Clinical Laboratory of the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Hong-Lian Wang
- Research Center for Integrative Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu-Qing Li
- College of Integrated Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Xin-Ming Yu
- College of Integrated Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Xiu Yang
- College of Integrated Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Hong-Wei Su
- The Department of Urology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Ping Li
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Li Wang
- Research Center for Integrative Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| |
Collapse
|
6
|
Yan L, Jiang MY, Fan XS. Research into the anti-pulmonary fibrosis mechanism of Renshen Pingfei formula based on network pharmacology, metabolomics, and verification of AMPK/PPAR-γ pathway of active ingredients. JOURNAL OF ETHNOPHARMACOLOGY 2023; 317:116773. [PMID: 37308028 DOI: 10.1016/j.jep.2023.116773] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/30/2023] [Accepted: 06/09/2023] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive disease with limited therapy. Renshen Pingfei Formula (RPFF), a classic Chinese medicine derivative formula, has been shown to exert therapeutic effects on IPF. AIM OF THE STUDY The study aimed to explore the anti-pulmonary fibrosis mechanism of RPFF through network pharmacology, clinical plasma metabolomics, and in vitro experiment. METHODS Network pharmacology was used to study the holistic pharmacological mechanism of RPFF in the treatment of IPF. The differential plasma metabolites for RPFF in the treatment of IPF were identified by untargeted metabolomics analysis. By integrated analysis of metabolomics and network pharmacology, the therapeutic target of RPFF for IPF and the corresponding herbal ingredients were identified. In addition, the effects of the main components of the formula, kaempferol and luteolin, which regulate the adenosine monophosphate (AMP)-activated protein kinase (AMPK)/peroxisome proliferator-activated receptor γ (PPAR-γ) pathway were observed in vitro according to the orthogonal design. RESULTS A total of 92 potential targets for RPFF in the treatment of IPF were obtained. The Drug-Ingredients-Disease Target network showed that PTGS2, ESR1, SCN5A, PPAR-γ, and PRSS1 were associated with more herbal ingredients. The protein-protein interaction (PPI) network identified the key targets of RPFF in IPF treatment, including IL6, VEGFA, PTGS2, PPAR-γ, and STAT3. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis acquired the main enriched pathways, and PPAR-γ involved in multiple signaling pathways, including the AMPK signaling pathway. Untargeted clinical metabolomics analysis revealed plasma metabolite variations in patients with IPF versus controls and before versus after RPFF treatment for patients with IPF. Six differential metabolites were explored as differential plasma metabolites for RPFF in IPF treatment. Combined with network pharmacology, a therapeutic target PPAR-γ of RPFF in IPF treatment and the corresponding herbal components were identified. Based on the orthogonal experimental design, the experiments showed that kaempferol and luteolin can decrease the mRNA and protein expression of α-smooth muscle actin (α-SMA), and the combination of lower dose can inhibit α-SMA mRNA and protein expression by promoting the AMPK/PPAR-γ pathway in transforming growth factor beta 1 (TGF-β1)-treated MRC-5 cells. CONCLUSIONS This study revealed that the therapeutic effects of RPFF are due to multiple ingredients and have multiple targets and pathways, and PPAR-γ is one of therapeutic targets for RPPF in IPF and involved in the AMPK signaling pathway. Two ingredients of RPFF, kaempferol and luteolin, can inhibit fibroblast proliferation and the myofibroblast differentiation of TGF-β1, and exert a synergistic effect through AMPK/PPAR-γ pathway activation.
Collapse
Affiliation(s)
- Lu Yan
- School of Traditional Chinese Medicine & Integrated Chinese and Western Medicine, Naning University of Chinese Medicine, Nanjing, 210023, China; Department of Respiratory and Critical Care Medicine, Central Laboratory, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nangjing, 210017, China.
| | - Min-Yue Jiang
- School of Traditional Chinese Medicine & Integrated Chinese and Western Medicine, Naning University of Chinese Medicine, Nanjing, 210023, China.
| | - Xin-Sheng Fan
- School of Traditional Chinese Medicine & Integrated Chinese and Western Medicine, Naning University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
7
|
Nørregaard R, Mutsaers HAM, Frøkiær J, Kwon TH. Obstructive nephropathy and molecular pathophysiology of renal interstitial fibrosis. Physiol Rev 2023; 103:2827-2872. [PMID: 37440209 PMCID: PMC10642920 DOI: 10.1152/physrev.00027.2022] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 07/05/2023] [Accepted: 07/09/2023] [Indexed: 07/14/2023] Open
Abstract
The kidneys play a key role in maintaining total body homeostasis. The complexity of this task is reflected in the unique architecture of the organ. Ureteral obstruction greatly affects renal physiology by altering hemodynamics, changing glomerular filtration and renal metabolism, and inducing architectural malformations of the kidney parenchyma, most importantly renal fibrosis. Persisting pathological changes lead to chronic kidney disease, which currently affects ∼10% of the global population and is one of the major causes of death worldwide. Studies on the consequences of ureteral obstruction date back to the 1800s. Even today, experimental unilateral ureteral obstruction (UUO) remains the standard model for tubulointerstitial fibrosis. However, the model has certain limitations when it comes to studying tubular injury and repair, as well as a limited potential for human translation. Nevertheless, ureteral obstruction has provided the scientific community with a wealth of knowledge on renal (patho)physiology. With the introduction of advanced omics techniques, the classical UUO model has remained relevant to this day and has been instrumental in understanding renal fibrosis at the molecular, genomic, and cellular levels. This review details key concepts and recent advances in the understanding of obstructive nephropathy, highlighting the pathophysiological hallmarks responsible for the functional and architectural changes induced by ureteral obstruction, with a special emphasis on renal fibrosis.
Collapse
Affiliation(s)
- Rikke Nørregaard
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | | | - Jørgen Frøkiær
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Tae-Hwan Kwon
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Taegu, Korea
| |
Collapse
|
8
|
Shao YF, Tang BB, Ding YH, Fang CY, Hong L, Shao CX, Yang ZX, Qiu YP, Wang JC, Yang B, Weng QJ, Wang JJ, He QJ. Kaempferide ameliorates cisplatin-induced nephrotoxicity via inhibiting oxidative stress and inducing autophagy. Acta Pharmacol Sin 2023; 44:1442-1454. [PMID: 36658427 PMCID: PMC10310756 DOI: 10.1038/s41401-023-01051-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 01/06/2023] [Indexed: 01/20/2023]
Abstract
Acute kidney injury (AKI) caused by anti-tumor drugs, such as cisplatin, is a severe complication with no effective treatment currently, leading to the reduction or discontinuation of chemotherapy. Natural products or herbal medicines are gradually considered as promising agents against cisplatin-induced AKI with the advantages of multi-targeting, multi-effects, and less resistance. In this study, we investigated the effects of kaempferide, a natural flavonoid extracted from the rhizome of Kaempferia galanga, in experimental AKI models in vitro and in vivo. We first conducted pharmacokinetic study in mice and found a relative stable state of kaempferide with a small amount of conversion into kaempferol. We showed that both kaempferide (10 μM) and kaempferol (10 μM) significantly inhibited cisplatin-caused injuries in immortalized proximal tubule epithelial cell line HK-2. In AKI mice induced by injection of a single dose of cisplatin (15 mg/kg), oral administration of kaempferide (50 mg/kg) either before or after cisplatin injection markedly improved renal function, and ameliorated renal tissue damage. We demonstrated that kaempferide inhibited oxidative stress and induced autophagy in cisplatin-treated mice and HK-2 cells, thus increasing tubular cell viability and decreasing immune responses to attenuate the disease progression. In addition, treatment with kaempferide significantly ameliorated ischemia-reperfusion-induced renal injury in vitro and in vivo. We conclude that kaempferide is a promising natural product for treating various AKI. This study has great implications for promotion of its use in healthcare products, and help to break through the limited use of cisplatin in the clinic.
Collapse
Affiliation(s)
- Yan-Fei Shao
- Center for Drug Safety Evaluation and Research; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Department of Pharmacy, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China
| | - Bing-Bing Tang
- Center for Drug Safety Evaluation and Research; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yu-Hui Ding
- Center for Drug Safety Evaluation and Research; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Chun-Yan Fang
- Center for Drug Safety Evaluation and Research; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ling Hong
- Center for Drug Safety Evaluation and Research; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Department of Pharmacy, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China
| | - Chun-Xiao Shao
- Center for Drug Safety Evaluation and Research; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Zhao-Xu Yang
- Center for Drug Safety Evaluation and Research; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yue-Ping Qiu
- Center for Drug Safety Evaluation and Research; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jin-Cheng Wang
- Center for Drug Safety Evaluation and Research; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Bo Yang
- Center for Drug Safety Evaluation and Research; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Qin-Jie Weng
- Center for Drug Safety Evaluation and Research; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jia-Jia Wang
- Center for Drug Safety Evaluation and Research; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Qiao-Jun He
- Center for Drug Safety Evaluation and Research; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
9
|
Guan Y, Quan D, Chen K, Kang L, Yang D, Wu H, Yan M, Wu S, Lv L, Zhang G. Kaempferol inhibits renal fibrosis by suppression of the sonic hedgehog signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 108:154246. [PMID: 36274411 DOI: 10.1016/j.phymed.2022.154246] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 06/01/2022] [Accepted: 06/05/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Most chronic kidney diseases (CKDs) develop to end-stage renal disease (ESRD), which is characterized by fibrosis and permanent tissue and function loss. As a result, better and more effective remedies are essential. Kaempferol (KAE) is a common flavonoid extracted from plants. It can control the progression of kidney fibrosis and the epithelial-to-mesenchymal transition (EMT) of the renal tubular system. PURPOSE We aim to investigate the effect of KAE therapy on extracellular matrix deposition and stimulation of EMT in vitro and in vivo to elucidate the treatment mechanisms regulating these effects. STUDY DESIGN Chronic hypertension-induced kidney fibrosis was studied in spontaneously hypertensive rats with chronic kidney disease. Biochemical analysis, histological staining, and the expression level of relative proteins were used to assess the effect of KAE on renal function and fibrosis. The direct impact of KAE on proliferation and migration was evaluated using human renal tubular epithelial cells (HK-2) induced by transforming growth factor-β1 (TGF-β1), which can then induce EMT. The molecular mechanism of KAE was verified using co-IP assay and immunofluorescence. RESULTS KAE could reduce blood pressure and decrease the extracellular matrix (ECM) components (including collagen I and collagen Ш), TGF-β1, and α-SMA in the kidneys of hypertension-induced rats with chronic kidney disease. Moreover, in HK-2 cell treated with TGF-β1, KAE administration significantly suppressed proliferation, migration, and EMT via increasing the expression of E-cadherin, while reducing the N-cadherin and α-SMA. Sufu was exceedingly repressed in HK-2 cells treated with TGF-β1. KAE inhibited the activation of Shh and Gli through increasing the expression of Sufu, thereby blocking the nuclear translocation of Gli1 in vitro. CONCLUSION KAE ameliorated kidney fibrosis and EMT by inhibiting the sonic hedgehog signaling pathway, thereby to attenuate the pathological progression of hypertensive kidney fibrosis.
Collapse
Affiliation(s)
- Yiqing Guan
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510000, China
| | - Dongling Quan
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510000, China
| | - Kai Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510000, China
| | - Liangqi Kang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510000, China
| | - Danni Yang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510000, China
| | - Huanxian Wu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510000, China
| | - Mengqiu Yan
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510000, China
| | - Shaoyu Wu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510000, China
| | - Lin Lv
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510000, China.
| | - Guohua Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510000, China.
| |
Collapse
|
10
|
Zhao Y, Li X, Wang F, Huang S, Du H, Li S, Chen J. Network Pharmacology and Experimental Verification Strategies to Illustrate the Mechanism of Jian-Pi-Yi-Shen Formula in Suppressing Epithelial-Mesenchymal Transition. Front Pharmacol 2022; 13:873023. [PMID: 35656312 PMCID: PMC9152215 DOI: 10.3389/fphar.2022.873023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/24/2022] [Indexed: 01/18/2023] Open
Abstract
Jian-Pi-Yi-Shen formula (JPYSF), a traditional Chinese medicine, has been recommended to treat renal fibrosis for decades. Previous studies had shown that JPYSF could inhibit epithelial–mesenchymal transition (EMT), an important regulatory role in renal fibrosis. However, the mechanism of JPYSF action is largely unknown. In this study, network pharmacology and experimental verification were combined to elucidate and identify the potential mechanism of JPYSF against renal fibrosis by suppressing EMT at molecular and pathway levels. Network pharmacology was first performed to explore the mechanism of JPYSF against renal fibrosis targeting EMT, and then a 5/6 nephrectomy (5/6 Nx)-induced rat model of renal fibrosis was selected to verify the predictive results by Masson’s trichrome stains and western blot analysis. Two hundred and thirty-two compounds in JPYSF were selected for the network approach analysis, which identified 137 candidate targets of JPYSF and 4,796 known therapeutic targets of EMT. The results of the Gene Ontology (GO) function enrichment analysis included 2098, 88, and 133 GO terms for biological processes (BPs), molecular functions (MFs), and cell component entries, respectively. The top 10 enrichment items of BP annotations included a response to a steroid hormone, a metal ion, oxygen levels, and so on. Cellular composition (CC) is mainly enriched in membrane raft, membrane microdomain, membrane region, etc. The MF of JPYSF analysis on EMT was predominately involved in proximal promoter sequence-specific DNA binding, protein heterodimerization activity, RNA polymerase II proximal promoter sequence-specific DNA binding, and so on. The involvement signaling pathway of JPYSF in the treatment of renal fibrosis targeting EMT was associated with anti-fibrosis, anti-inflammation, podocyte protection, and metabolism regulation. Furthermore, the in vivo experiments confirmed that JPYSF effectively ameliorated interstitial fibrosis and inhibited the overexpression of α-SMA, Wnt3a, and β-catenin, and increased the expression of E-cadherin by wnt3a/β-catenin signaling pathway in 5/6 Nx-induced renal fibrosis rats. Using an integrative network pharmacology-based approach and experimental verification, the study showed that JPYSF had therapeutic effects on EMT by regulating multi-pathway, among which one proven pathway was the Wnt3a/β-catenin signaling pathway. These findings provide insights into the renoprotective effects of JPYSF against EMT, which could suggest directions for further research of JPYSF in attenuating renal fibrosis by suppressing EMT.
Collapse
Affiliation(s)
- Yuan Zhao
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Xiangbin Li
- Shenzhen Hospital of Beijing University of Chinese Medicine (Longgang), Shenzhen, China
| | - Fochang Wang
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Shiying Huang
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Hanqian Du
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shunmin Li
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Jianping Chen
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| |
Collapse
|
11
|
Alagal RI, AlFaris NA, Alshammari GM, ALTamimi JZ, AlMousa LA, Yahya MA. Kaempferol attenuates doxorubicin-mediated nephropathy in rats by activating SIRT1 signaling. J Funct Foods 2022. [DOI: 10.1016/j.jff.2021.104918] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
12
|
Hu L, Yang K, Mai X, Wei J, Ma C. Depleted HDAC3 attenuates hyperuricemia-induced renal interstitial fibrosis via miR-19b-3p/SF3B3 axis. Cell Cycle 2022; 21:450-461. [PMID: 35025700 PMCID: PMC8942505 DOI: 10.1080/15384101.2021.1989899] [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] [Indexed: 11/03/2022] Open
Abstract
Dysfunctional histone deacetylases (HDACs) elicit unrestrained fibrosis and damage to organs. With regard to the link between HDACs and fibrosis, this research is practiced to decipher the concrete mechanism of HDAC3 in hyperuricemia (HN)-induced renal interstitial fibrosis (RIF) from microRNA-19b-3p/splicing factor 3b subunit 3 (miR-19b-3p/SF3B3) axis.The HN model was established on rats to induce RIF by oral administration of adenine and potassium oxalate. HN rats were injected with miR-19b-3p- or HDAC3-related vectors to figure out their effects on RIF through detecting 24-h urine protein, uric acid (UA), blood urea nitrogen (BUN) and serum creatinine (Scr) contents and α-smooth muscle actin (α-SMA), transforming growth factor β1 (TGF-β1) and fibronectin (FN) contents in renal tissues and observing pathological damages and RIF index of renal tissues. HDAC3, miR-19b-3p and SF3B3 expression in renal tissues were tested, along with their interactions.Elevated HDAC3 and SF3B3 and reduced miR-19b-3p were displayed in renal tissues of HN rats. Suppressed HDAC3 or promoted miR-19b-3p relieved HN-induced RIF, as reflected by their inhibitory effects on 24 h urine protein, UA, BUN, Scr, α-SMA, TGF-β1, and FN contents and RIF index and their ameliorated effects on pathological damages of renal tissues. HDAC3 bound to the promoter of miR-19b-3p to regulate SF3B3. MiR-19b-3p depletion abrogated down-regulated HDAC3-induced effects on HN-induced RIF.It is delineated that depressed HDAC3 relives HN-induced RIF through restoring miR-19b-3p and knocking down SF3B3, replenishing the references for RIF curing.
Collapse
Affiliation(s)
- Langtao Hu
- Department of Nephrology, Hainan General Hospital, Haikou, China.,Department of Nephrology, Hainan Affiliated Hospital of Hainan Medical College, Haikou, China
| | - Kai Yang
- Department of Nephrology, Hainan General Hospital, Haikou, China.,Department of Nephrology, Hainan Affiliated Hospital of Hainan Medical College, Haikou, China
| | - Xing Mai
- Department of Nephrology, Hainan General Hospital, Haikou, China.,Department of Nephrology, Hainan Affiliated Hospital of Hainan Medical College, Haikou, China
| | - Jiali Wei
- Department of Nephrology, Hainan General Hospital, Haikou, China.,Department of Nephrology, Hainan Affiliated Hospital of Hainan Medical College, Haikou, China
| | - Chunyang Ma
- Department of Neurosurgery, First Affiliated Hospital of Hainan Medical College, Haikou, China
| |
Collapse
|
13
|
Xu H, Wu T, Huang L. Therapeutic and delivery strategies of phytoconstituents for renal fibrosis. Adv Drug Deliv Rev 2021; 177:113911. [PMID: 34358538 DOI: 10.1016/j.addr.2021.113911] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/07/2021] [Accepted: 07/29/2021] [Indexed: 12/11/2022]
Abstract
Chronic kidney disease (CKD) is one of the most common diseases endangering human health and life. By 2030, 14 per 100,000 people may die from CKD. Renal fibrosis (RF) is an important intermediate link and the final pathological change during CKD progression to the terminal stage. Therefore, identifying safe and effective treatment methods for RF has become an important goal. In 2018, the World Health Organization introduced traditional Chinese medicine into its effective global medical program. Various phytoconstituents that affect the RF process have been extracted from different plants. Here, we review the potential therapeutic capabilities of active phytoconstituents in RF treatment and discuss how phytoconstituents can be structurally modified or combined with other ingredients to enhance efficiency and reduce toxicity. We also summarize phytoconstituent delivery strategies to overcome renal barriers and improve bioavailability and targeting.
Collapse
Affiliation(s)
- Huan Xu
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, People's Republic of China.
| | - Tianyi Wu
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, People's Republic of China
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| |
Collapse
|
14
|
Shen WX, Luo RC, Wang JQ, Chen ZS. Features of Cytokine Storm Identified by Distinguishing Clinical Manifestations in COVID-19. Front Public Health 2021; 9:671788. [PMID: 34109148 PMCID: PMC8180556 DOI: 10.3389/fpubh.2021.671788] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/27/2021] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is caused by a new coronavirus, namely severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and is currently spreading all over the world. In this paper, we developed a practical model for identifying the features of cytokine storm, which is common in acute infectious diseases and harmful manifestation of COVID-19, by distinguishing major and minor clinical events. This model is particularly suitable for identifying febrile and infectious diseases like COVID-19. Based on this model, features of cytokine storm and pathogenesis of COVID-19 have been proposed to be a consequence of the disequilibrated cytokine network resulting from increased biological activity of transforming growth factor-β (TGF-β), which induces certain clinical manifestations such as fatigue, fever, dry cough, pneumonia, abatement and losing of olfactory, and taste senses in some patients. Research and clarification of the pathogenesis of COVID-19 will contribute to precision treatment. Various anti-TGF-β therapies may be explored as potential COVID-19 treatment. This novel model will be helpful in reducing the widespread mortality of COVID-19.
Collapse
Affiliation(s)
- Wei-Xi Shen
- Shenzhen Hospital, Southern Medical University, Shenzhen, China
- Shenzhen Tianyou Medical Institute, Shenzhen, China
| | - Rong-Cheng Luo
- Shenzhen Hospital, Southern Medical University, Shenzhen, China
- Shenzhen Tianyou Medical Institute, Shenzhen, China
| | - Jing-Quan Wang
- College of Pharmacy and Health Science, St. John's University, New York, NY, United States
| | - Zhe-Sheng Chen
- College of Pharmacy and Health Science, St. John's University, New York, NY, United States
| |
Collapse
|
15
|
Zhang S, Chang Q, Li P, Tong X, Feng Y, Hao X, Zhang X, Yuan Z, Tan J. Concentrated small extracellular vesicles from menstrual blood-derived stromal cells improve intrauterine adhesion, a pre-clinical study in a rat model. NANOSCALE 2021; 13:7334-7347. [PMID: 33889891 DOI: 10.1039/d0nr08942g] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
We previously reported that transplantation of menstrual blood-derived stromal cells (MenSCs) significantly improved fertility restoration in intrauterine adhesion (IUA). However, it is difficult to obtain menstrual blood samples in some severe IUA patients who have amenorrhea or oligomenorrhea. Thus, a safe and effective stem cell replacement therapy is necessary to promote endometrial regeneration. Recent studies demonstrated that the effects of MenSCs are partly mediated in a paracrine manner via small extracellular vesicles (sEVs). To explore this possibility, we performed a pre-clinical study to investigate whether concentrated MenSC-derived sEVs (MenSCs-sEVs) are sufficient to repair IUA and the mechanisms underlying their action. Rat IUA models were established by mechanical injury, followed by the administration of MenSCs or MenSCs-sEVs through intrauterine transplantation. Consistent with the efficacy of MenSCs, MenSCs-sEVs effectively recovered the morphology, promoted regeneration of the glands and angiogenesis, and reversed endometrial fibrosis in the IUA uterus. The endometrial receptivity and pregnancy outcome significantly improved after repeated MenSCs-sEVs transplantations. In addition, all rats in the MenSCs-sEVs group had no hematological or biochemical abnormalities. Three-dimensional fluorescence imaging suggested that MenSCs tended to migrate through the bloodstream, whereas MenSCs-sEVs had a better localized therapeutic effect. Moreover, MenSCs and MenSCs-sEVs inhibited the TGFβ1/SMAD3 pathway in the IUA endometrium, while promoting the phosphorylation of SMAD1/5/8 and ERK 1/2 and upregulating the expression of BMP7. Thus, MenSCs-sEVs safely and effectively enhanced endometrial restoration, suggesting a promising non-cellular therapy for endometrial regeneration and a key role in MenSC-mediated IUA treatment.
Collapse
Affiliation(s)
- Siwen Zhang
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110022, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Liang T, Wei C, Lu S, Qin M, Qin G, Zhang Y, Zhong X, Zou X, Yang Y. Ginaton injection alleviates cisplatin-induced renal interstitial fibrosis in rats via inhibition of apoptosis through regulation of the p38MAPK/TGF-β1 and p38MAPK/HIF-1α pathways. Biomed Rep 2021; 14:38. [PMID: 33692901 PMCID: PMC7938297 DOI: 10.3892/br.2021.1414] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 02/04/2021] [Indexed: 11/06/2022] Open
Abstract
Ginaton injection (Ginkgo biloba extract; GBE) has been reported to protect against cisplatin-induced acute renal failure in rats. In the present study, the effects and molecular mechanisms of GBE on cisplatin-induced renal interstitial fibrosis were evaluated using a rat model. The rats were intraperitoneally injected with cisplatin once on the first day and a subset of rats were treated with GBE or SB203580 (SB; a specific p38 MAPK inhibitor) daily from days 22 to 40. The levels of N-acetyl-β-D-Glucosaminidase (NAG) in the urine, and of urea nitrogen (BUN) and creatinine (Scr) in the blood were assessed. The damage and fibrosis of renal tissues were evaluated using hematoxylin and eosin staining, as well as Masson's trichrome staining, respectively. Apoptosis in renal tissues was detected using a TUNEL assay. The protein expression levels of α-smooth muscle actin (SMA), collagen 1 (Col I), Bax, Bcl-2, caspase-3/cleaved caspase-3, hypoxia-inducible factor-1α (HIF-1α), TGF-β1 and p38MAPK, as well as the mRNA levels of p38MAPK in renal tissues were investigated. The results showed that GBE markedly reduced the levels of urinary NAG, Scr and BUN, and renal expression of α-SMA and Col I levels were also reduced. Furthermore, GBE significantly reduced renal tissue injury and the relative area of renal interstitial fibrosis induced by cisplatin. GBE effectively reduced the apoptotic rate of renal tissues, the protein expression levels of Bax, cleaved caspase-3, phospho-p38MAPK, TGF-β1 and HIF-1α, as well as the mRNA expression levels of p38MAPK in renal tissues induced by cisplatin, whereas GBE significantly increased Bcl-2 protein expression. SB exhibited similar effects to GBE, although it was not as effective. In summary, the present study is the first to show that GBE significantly alleviated renal interstitial fibrosis following cisplatin-induced acute renal injury. The mechanisms by which GBE exhibited its effects were associated with the inhibition of apoptosis via downregulation of the p38MAPK/TGF-β1 and p38MAPK/HIF-1α signaling pathways.
Collapse
Affiliation(s)
- Taolin Liang
- Postgraduate Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Chongying Wei
- Postgraduate Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Sisi Lu
- Postgraduate Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Mengyuan Qin
- Postgraduate Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Guiming Qin
- Postgraduate Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yansong Zhang
- Postgraduate Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xiaobin Zhong
- Regenerative Medicine Research Center of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xiaoqin Zou
- Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yufang Yang
- Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|