1
|
Liu Y, Du Z, Sheng C, Zhang G, Yan S, Zhang Z, Qin S. The Double-Edge Sword of Natural Phenanthrenes in the Landscape of Tumorigenesis. Molecules 2025; 30:1204. [PMID: 40141980 PMCID: PMC11946065 DOI: 10.3390/molecules30061204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
Phenanthrenes, which are polycyclic aromatic hydrocarbons comprising three benzene rings, exhibit a diverse range of functions. These compounds are utilized in the synthesis of resins, plant growth hormones, reducing dyes, tannins and other products. Notably, phenanthrenes possess significant pharmacological properties, including anti-tumor, anti-inflammatory and antioxidant activities, offering broad prospects for development, particularly in the fields of medicine and health. Interestingly, although aristolochic acid (AA) is a potent carcinogen, its lactam analogs can kill cancer cells and exhibit therapeutic effects against cancer. This provides a promising strategy for the toxicity-effect transformation of phenanthrenes. In this paper, we reviewed 137 articles to systematically review the anti-tumor potential and toxic effects of natural phenanthrenes isolated from the 19th century to the present, thus offering references and laying a foundation for their further research, development and utilization.
Collapse
Affiliation(s)
- Yan Liu
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China; (Y.L.); (Z.D.); (C.S.); (G.Z.); (S.Y.)
- Research Center for Precision Medication of Chinese Medicine, FuRong Laboratory, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Ziwei Du
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China; (Y.L.); (Z.D.); (C.S.); (G.Z.); (S.Y.)
| | - Chen Sheng
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China; (Y.L.); (Z.D.); (C.S.); (G.Z.); (S.Y.)
| | - Guangshuai Zhang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China; (Y.L.); (Z.D.); (C.S.); (G.Z.); (S.Y.)
- Research Center for Precision Medication of Chinese Medicine, FuRong Laboratory, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Si Yan
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China; (Y.L.); (Z.D.); (C.S.); (G.Z.); (S.Y.)
- Research Center for Precision Medication of Chinese Medicine, FuRong Laboratory, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Zhijun Zhang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, China; (Y.L.); (Z.D.); (C.S.); (G.Z.); (S.Y.)
| | - Shuanglin Qin
- Research Center for Precision Medication of Chinese Medicine, FuRong Laboratory, Hunan University of Chinese Medicine, Changsha 410208, China
| |
Collapse
|
2
|
Liu Z, Gao H, Li G, Yu Y, Cui M, Peng H, Guan X, Zhang X, Zhang Z, Shen X, Chen S, Li D, Chen L, Xiao Y, Chen W, Liu L, Wang Q. Genome-wide CRISPR-based screen identifies E2F transcription factor 1 as a regulator and therapeutic target of aristolochic acid-induced nephrotoxicity. ENVIRONMENT INTERNATIONAL 2025; 195:109234. [PMID: 39724681 DOI: 10.1016/j.envint.2024.109234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/06/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024]
Abstract
Aristolochic Acid I (AAI) is widely present in traditional Chinese medicines derived from the Aristolochia genus and is known to cause significant damage to renal tubular epithelial cells. Genome-wide screening has proven to be a powerful tool in identifying critical genes associated with the toxicity of exogenous substances. To identify undiscovered key genes involved in AAI-induced renal toxicity, a genome-wide CRISPR library screen was conducted in the human kidney-2 (HK-2) cell line. Among the altered sgRNAs, a significant enrichment of those targeting the E2F transcription factor 1 (E2F1) gene was observed in surviving HK-2 cells in the AAI-treated group. Interestingly, the role of E2F1 had not been previously explored in studies of AAI nephrotoxicity. Further investigations revealed that E2F1 promotes apoptosis by activating the p53 signaling pathway and upregulating pro-apoptotic genes, such as BAK and BAX. Additionally, using the high-throughput experiment- and reference-guided database of traditional Chinese medicine (HERB), cannabidiol (CBD) was identified as an inhibitor of E2F1 by suppressing the activity of NF-κB pathway. In vitro and in vivo models confirmed that CBD inhibits AAI-induced upregulation of E2F1, thereby suppressing p53-mediated apoptosis. In conclusion, this study highlights the crucial role of E2F1 in AAI-induced renal cell apoptosis and identifies CBD as a novel therapeutic candidate for mitigating AAI nephrotoxicity.
Collapse
Affiliation(s)
- Ziqi Liu
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Huan Gao
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Guoliang Li
- Department of Toxicology, Guangdong Province Hospital for Occupational Disease Prevention and Treatment, Guangzhou, Guangdong, 510300, China
| | - Yongjiang Yu
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Mengxing Cui
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Honghao Peng
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xinchao Guan
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xue Zhang
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhihan Zhang
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiaoyu Shen
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shen Chen
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Daochuan Li
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Liping Chen
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yongmei Xiao
- Department of Occupational and Environmental Health, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Wen Chen
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Lili Liu
- Department of Toxicology, Guangdong Province Hospital for Occupational Disease Prevention and Treatment, Guangzhou, Guangdong, 510300, China.
| | - Qing Wang
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
3
|
Chen L, Cheng S, Ying J, Zhang Q, Wang C, Wu H, Wang Y, Zhang H, Wang J, Ye J, Zhang L. Aristolochic acid I promotes renal tubulointerstitial fibrosis by up-regulating expression of indoleamine 2,3-dioxygenase-1 (IDO1). Toxicol Lett 2024; 402:44-55. [PMID: 39547317 DOI: 10.1016/j.toxlet.2024.11.003] [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: 05/08/2024] [Revised: 09/30/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024]
Abstract
Aristolochic acid I (AAI) is strongly nephrotoxic and can cause "Aristolochic acid nephropathy (AAN)". Aristolochic acid nephropathy is characterized by extensive renal interstitial fibrosis. However, the exact mechanism by which it occurs has not been fully elucidated. lt has been reported that indoleamine 2,3-dioxygenase-1 (IDO1) promotes renal fibrosis in renal disorders, but it is unclear how IDO1 functions in AAI-induced kidney fibrosis. In this work, we systematically examined the role of IDO1 in AAI-induced renal tubulointerstitial fibrosis. The results showed that AAI induced upregulation of IDO1 expression in renal tubular epithelial cells and mouse kidney. Inhibition of IDO1 expression reduced the levels of fibrosis-associated markers α-SMA, COL-I and FN and ameliorated renal tubular epithelial cell fibrosis. It also improved renal function, reduced collagen deposition, and ameliorated interstitial fibrosis in mice. Moreover, we discovered that inhibition of IDO1 decreased the expression of the apoptotic protein BAX, raised the expression of BCL-2 protein, and reduced apoptosis. The above studies suggest that IDO1 is a target of action in renal tubulointerstitial fibrosis caused by AAI, and inhibition of IDO1 may be a viable approach for the therapy of AAI-induced renal tubulointerstitial fibrosis.
Collapse
Affiliation(s)
- Langqun Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Siyu Cheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Jiahui Ying
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Qi Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Chen Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Huimin Wu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Ying Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Hong Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Jiahe Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Jing Ye
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China; State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China.
| | - Liang Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China.
| |
Collapse
|
4
|
Wang L, Shao Z, Wang N, Liu W, Zhang L, Wang Y, Tan J, Jiao X, Liu L, Yan L, Chen S, Cao H, Shao F. Receptor-interacting protein kinase 1 confers autophagic promotion of gasdermin E-mediated pyroptosis in aristolochic acid-induced acute kidney injury. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 284:116944. [PMID: 39208575 DOI: 10.1016/j.ecoenv.2024.116944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Aristolochic acid (AA) exposure is a severe public health concern worldwide. AAs damage the kidney with an inevitable acute phase that is similar to acute kidney injury (AKI). Gasdermin E (GSDME) is abundant in the kidney; thus; it-mediated pyroptosis might be essential in connecting cell death and inflammation and promoting AAs-AKI. However, the role and exact mechanism of pyroptosis in AAs-AKI have not been investigated. In this study, aristolochic acid I (AAI) was used to establish AKI models. The expression and translocation results showed GSDME-mediated pyroptosis in AAI-AKI. Knocking down GSDME attenuated AAI-induced cell death and transcription of proinflammatory cytokines. Mechanistic research inhibiting caspase (casp) 3, casp 8, and casp 9 with specific chemical antagonists demonstrated that GSDME was activated by cleaved casp 3. Furthermore, the kinase activity of upstream receptor-interacting protein kinase 1 (RIPK1) was significantly elevated, and inhibiting RIPK1 with specific inhibitors markedly improved AAI-induced cell damage. In addition, the level of autophagy was obviously increased. Pretreatment with a specific autophagic inhibitor (3-methyladenine) or knockdown of autophagic genes (Atg5 or Atg7) evidently reduced the activity of RIPK1 and downstream apoptosis and pyroptosis, thus attenuating AA-induced cell injury, which suggested that RIPK1 was a novel link conferring autophagic promotion of pyroptosis. These findings reveal GSDME-mediated pyroptosis for the first time in AAI-induced AKI, propose its novel role in the transcription of cytokines, and demonstrate that autophagy promotes pyroptosis via the RIPK1-dependent apoptotic pathway. This study promotes the understanding of the toxic effects and exact mechanisms of AAs. This will contribute to evaluating the environmental risk of AA exposure and might provide potential therapeutic targets for AA-AKI.
Collapse
Affiliation(s)
- Limeng Wang
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, 7 Weiwu Road, Jinshui District, Zhengzhou, Henan 450053, China
| | - Zehua Shao
- Children's Heart Center, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450053, China
| | - Ning Wang
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, 7 Weiwu Road, Jinshui District, Zhengzhou, Henan 450053, China
| | - Wenna Liu
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, 7 Weiwu Road, Jinshui District, Zhengzhou, Henan 450053, China; Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan 451464, China
| | - Lina Zhang
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, 7 Weiwu Road, Jinshui District, Zhengzhou, Henan 450053, China
| | - Yanliang Wang
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, 7 Weiwu Road, Jinshui District, Zhengzhou, Henan 450053, China
| | - Jing Tan
- Department of Internal Medicine, Henan Medical College, Longhu Town, Zhengzhou, Henan 451191, China
| | - Xiaojing Jiao
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, 7 Weiwu Road, Jinshui District, Zhengzhou, Henan 450053, China
| | - Lu Liu
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, 7 Weiwu Road, Jinshui District, Zhengzhou, Henan 450053, China
| | - Lei Yan
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, 7 Weiwu Road, Jinshui District, Zhengzhou, Henan 450053, China
| | - Song Chen
- Translational Research Institute of Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450053, China.
| | - Huixia Cao
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, 7 Weiwu Road, Jinshui District, Zhengzhou, Henan 450053, China.
| | - Fengmin Shao
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial Clinical Research Center for Kidney Disease, People's Hospital of Zhengzhou University and Henan Provincial People's Hospital, 7 Weiwu Road, Jinshui District, Zhengzhou, Henan 450053, China.
| |
Collapse
|
5
|
Zhang L, Sang X, Han Y, Abulitibu A, Elken M, Mao Z, Kang S, Yang W, Lu C. The expression of apoptosis related genes in HK-2 cells overexpressing PPM1K was determined by RNA-seq analysis. Front Genet 2022; 13:1004610. [PMID: 36386814 PMCID: PMC9663473 DOI: 10.3389/fgene.2022.1004610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/18/2022] [Indexed: 01/24/2023] Open
Abstract
Chronic kidney disease (CKD) is a serious disease that endangers human health. It is reported that inhibiting renal cell apoptosis can delay the progress of CKD. Our previous study found that the mice with protein phosphatase Mg2+/Mn2+ dependent 1K (PPM1K) gene deletion had obvious symptoms of glomerular vascular and interstitial vascular dilatation, congestion and hemorrhage, glomerular hemorrhage and necrosis, interstitial fibrous tissue proliferation, decreased urinary creatinine clearance, and increased urinary protein level. In addition, studies have found that PPM1K is essential for cell survival, apoptosis and metabolism. However, no study has confirmed that PPM1K can inhibit renal cell apoptosis. In this study, PPM1K was overexpressed in human kidney-2 cells (HK-2), and the biological process of differentially expressed genes and its effect on apoptosis were comprehensively screened by RNA sequencing (RNA-seq). Through sequencing analysis, we found that there were 796 differentially expressed genes in human renal tubular epithelial cells transfected with PPM1K gene, of which 553 were down-regulated and 243 were up-regulated. Enrichment analysis found that differentially expressed genes may play an important role in amino acid metabolism and biosynthesis. In the GO analysis functional pathway list, we also found that multiple genes can be enriched in apoptosis related pathways, such as G0S2, GADD45A, TRIB3, VEGFA, NUPR1 and other up-regulated genes, and IL-6, MAGED1, CCL2, TP53INP1 and other down-regulated genes. Then we verified these differentially expressed genes by RT-PCR, and found that only the RT-PCR results of G0S2, VEGFA and NUPR1 were consistent with the transcriptome sequencing results. We believe that G0S2, VEGFA, NUPR1 and other genes may participate in the apoptosis process of HK-2 cells induced by PPM1K.In conclusion, these findings provide some data support for the study of HK-2 cell apoptosis mechanism, and also provide a scientific theoretical basis for further study of the effect of PPM1K on kidney disease.
Collapse
Affiliation(s)
- Li Zhang
- Nephrology Center of the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China,Xinjiang Clinical Research Center of Renal Replacement Therapy, Urumqi, China,Xinjiang Branch of National Clinical Research Center for Kidney Disease, Urumqi, China,Xinjiang Blood Purification Medical Quality Control Center, Urumqi, China,Institute of Nephrology of Xinjiang, Urumqi, China
| | - Xiaohong Sang
- Nephrology Center of the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China,Xinjiang Clinical Research Center of Renal Replacement Therapy, Urumqi, China,Xinjiang Branch of National Clinical Research Center for Kidney Disease, Urumqi, China,Xinjiang Blood Purification Medical Quality Control Center, Urumqi, China,Institute of Nephrology of Xinjiang, Urumqi, China
| | - Yuanyuan Han
- Nephrology Center of the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China,Xinjiang Clinical Research Center of Renal Replacement Therapy, Urumqi, China,Xinjiang Branch of National Clinical Research Center for Kidney Disease, Urumqi, China,Xinjiang Blood Purification Medical Quality Control Center, Urumqi, China,Institute of Nephrology of Xinjiang, Urumqi, China
| | - Alpati Abulitibu
- Nephrology Center of the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China,Xinjiang Clinical Research Center of Renal Replacement Therapy, Urumqi, China,Xinjiang Branch of National Clinical Research Center for Kidney Disease, Urumqi, China,Xinjiang Blood Purification Medical Quality Control Center, Urumqi, China,Institute of Nephrology of Xinjiang, Urumqi, China
| | - Mufunayi Elken
- Nephrology Center of the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China,Xinjiang Clinical Research Center of Renal Replacement Therapy, Urumqi, China,Xinjiang Branch of National Clinical Research Center for Kidney Disease, Urumqi, China,Xinjiang Blood Purification Medical Quality Control Center, Urumqi, China,Institute of Nephrology of Xinjiang, Urumqi, China
| | - Zhijie Mao
- Nephrology Center of the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China,Xinjiang Clinical Research Center of Renal Replacement Therapy, Urumqi, China,Xinjiang Branch of National Clinical Research Center for Kidney Disease, Urumqi, China,Xinjiang Blood Purification Medical Quality Control Center, Urumqi, China,Institute of Nephrology of Xinjiang, Urumqi, China
| | - Shaotao Kang
- Nephrology Center of the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China,Xinjiang Clinical Research Center of Renal Replacement Therapy, Urumqi, China,Xinjiang Branch of National Clinical Research Center for Kidney Disease, Urumqi, China,Xinjiang Blood Purification Medical Quality Control Center, Urumqi, China,Institute of Nephrology of Xinjiang, Urumqi, China
| | - Wenjun Yang
- Nephrology Center of the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China,Xinjiang Clinical Research Center of Renal Replacement Therapy, Urumqi, China,Xinjiang Branch of National Clinical Research Center for Kidney Disease, Urumqi, China,Xinjiang Blood Purification Medical Quality Control Center, Urumqi, China,Institute of Nephrology of Xinjiang, Urumqi, China,*Correspondence: Wenjun Yang, ; Chen Lu,
| | - Chen Lu
- Nephrology Center of the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China,Xinjiang Clinical Research Center of Renal Replacement Therapy, Urumqi, China,Xinjiang Branch of National Clinical Research Center for Kidney Disease, Urumqi, China,Xinjiang Blood Purification Medical Quality Control Center, Urumqi, China,Institute of Nephrology of Xinjiang, Urumqi, China,*Correspondence: Wenjun Yang, ; Chen Lu,
| |
Collapse
|
6
|
Dong LY, Cao TY, Guo YH, Chen R, Zhao YS, Zhao Y, Kong H, Qu HH. Aristolochic Acid Nephropathy: A Novel Suppression Strategy of Carbon Dots Derived from Astragali Radix Carbonisata. J Biomed Nanotechnol 2022. [DOI: 10.1166/jbn.2022.3403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Despite strict restrictions on the use of aristolochic acids (AAs)-containing merchandise or drugs in many countries, a substantial amounts of occurrences aristolochic acid nephropathy (AAN) had been accounted worldwide. Clinically, there is no effective incurable therapy regimen to
reverse the progression of AAN. Although carbon dots have shown surprising bioactivity, research on the acute kidney injury caused by AAs is lacking. Here, a novel biomass-carbon dots from Astragali Radix (AR) as precursors was synthesized through one-step pyrolysis treatment. The ARC-carbon
dots (ARC-CDs) was demonstrated in detail for its inhibitory effect on aristolochic acid nephropathy in a mice model. The indexes of inflammatory cytokines as well as oxidative stress were significantly reduced by the ARC-CDs in kidney tissue cells. Additionally, the ARC-CDs administration
resulted in a large decrease in positive apoptotic cells according to TUNEL labeling and western blotting, which may be connected to the ARC-CDs’ modulation of the protein in the Akt/Mdm2/p53 signaling pathway. These findings show that ARC-CDs have remarkable anti-inflammatory, antioxidant,
and anti-apoptotic capabilities against acute kidney injury spurred by aristolochic acids via the AKT/Mdm2/p53 signaling pathway.
Collapse
Affiliation(s)
- Li-Yang Dong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People’s Republic of China
| | - Tian-You Cao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, People’s Republic of China
| | - Ying-Hui Guo
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People’s Republic of China
| | - Rui Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People’s Republic of China
| | - Yu-Sheng Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People’s Republic of China
| | - Yan Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People’s Republic of China
| | - Hui Kong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People’s Republic of China
| | - Hui-Hua Qu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People’s Republic of China
| |
Collapse
|
7
|
Ye B, Ma J, Li Z, Li Y, Han X. Ononin Shows Anticancer Activity Against Laryngeal Cancer via the Inhibition of ERK/JNK/p38 Signaling Pathway. Front Oncol 2022; 12:939646. [PMID: 35912256 PMCID: PMC9334013 DOI: 10.3389/fonc.2022.939646] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/13/2022] [Indexed: 12/24/2022] Open
Abstract
BackgroundLaryngeal cancer is a type of head and neck tumor with a poor prognosis and survival rate. The new cases of laryngeal cancer increased rapidly with a higher mortality rate around the world.ObjectiveThe current research work was focused to unveil the in vitro antitumor effects of ononin against the laryngeal cancer Hep-2 cells.MethodologyThe cytotoxic effects of ononin against the laryngeal cancer Hep-2 cells and normal HuLa-PC laryngeal cells were studied using an 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay. The intracellular Reactive Oxygen Species (ROS) generation, apoptotic cell death, Mitochondrial Membrane Potential (MMP), and cell adhesion on the 25 and 50 µM ononin-treated Hep-2 cells were detected using respective staining assays. The levels of TBARS and antioxidants were assayed using specific kits. The expressions of c-Jun N-terminal kinase 1/2 (JNK1/2), Extracellular Signal-regulated Kinase 1/2 (ERK1/2), p38, Phosphatidylinositol-3 Kinase 1/2 (PI3K1/2), and protein kinase-B (Akt) in the ononin-treated Hep-2 cells were investigated using Reverse Transcription-Polymerase Chain Reaction (RT-PCR) assay.ResultsThe ononin treatment effectively inhibited the Hep-2 cell viability but did not affect the viability of HuLa-PC cells. Furthermore, the ononin treatment effectively improved the intracellular ROS accumulation, depleted the MMP, and triggered apoptosis in Hep-2 cells. The Thiobarbituric acid reactive substances (TBARS) were improved, and Glutathione (GSH) levels and Superoxide dismutase (SOD) were depleted in the ononin-administered Hep-2 cells. The ononin treatment substantially inhibited the JNK/ERK/p38 axis in the Hep-2 cells.ConclusionTogether, the outcomes of this exploration proved that the ononin has remarkable antitumor activity against laryngeal cancer Hep-2 cells.
Collapse
Affiliation(s)
- Ben Ye
- Department of Ear, Nose, and Throat (ENT), Shandong Provincial Hospital Affiliated to Shandong First Medical University, Ji’nan, China
| | - Jianhua Ma
- Department of Cardiology, Shandong Rongjun General Hospital, Ji’nan, China
| | - Zhaoxia Li
- Department of Ear, Nose, and Throat (ENT), Shandong Provincial Hospital Affiliated to Shandong First Medical University, Ji’nan, China
| | - Yang Li
- Department of Plastic Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiaopan Han
- Department of ENT, Central Hospital Affiliated to Shandong First Medical University, Ji’nan, China
- *Correspondence: Xiaopan Han,
| |
Collapse
|
8
|
Wang A, Li Z, Zhuo S, Gao F, Zhang H, Zhang Z, Ren G, Ma X. Mechanisms of Cardiorenal Protection With SGLT2 Inhibitors in Patients With T2DM Based on Network Pharmacology. Front Cardiovasc Med 2022; 9:857952. [PMID: 35677689 PMCID: PMC9169967 DOI: 10.3389/fcvm.2022.857952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 05/04/2022] [Indexed: 11/24/2022] Open
Abstract
Purpose Sodium-glucose cotransporter 2 (SGLT2) inhibitors have cardiorenal protective effects regardless of whether they are combined with type 2 diabetes mellitus, but their specific pharmacological mechanisms remain undetermined. Materials and Methods We used databases to obtain information on the disease targets of “Chronic Kidney Disease,” “Heart Failure,” and “Type 2 Diabetes Mellitus” as well as the targets of SGLT2 inhibitors. After screening the common targets, we used Cytoscape 3.8.2 software to construct SGLT2 inhibitors' regulatory network and protein-protein interaction network. The clusterProfiler R package was used to perform gene ontology functional analysis and Kyoto encyclopedia of genes and genomes pathway enrichment analyses on the target genes. Molecular docking was utilized to verify the relationship between SGLT2 inhibitors and core targets. Results Seven different SGLT2 inhibitors were found to have cardiorenal protective effects on 146 targets. The main mechanisms of action may be associated with lipid and atherosclerosis, MAPK signaling pathway, Rap1 signaling pathway, endocrine resistance, fluid shear stress, atherosclerosis, TNF signaling pathway, relaxin signaling pathway, neurotrophin signaling pathway, and AGEs-RAGE signaling pathway in diabetic complications were related. Docking of SGLT2 inhibitors with key targets such as GAPDH, MAPK3, MMP9, MAPK1, and NRAS revealed that these compounds bind to proteins spontaneously. Conclusion Based on pharmacological networks, this study elucidates the potential mechanisms of action of SGLT2 inhibitors from a systemic and holistic perspective. These key targets and pathways will provide new ideas for future studies on the pharmacological mechanisms of cardiorenal protection by SGLT2 inhibitors.
Collapse
Affiliation(s)
- Anzhu Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhendong Li
- Qingdao West Coast New Area People's Hospital, Qingdao, China
| | - Sun Zhuo
- Qingdao West Coast New Area People's Hospital, Qingdao, China
| | - Feng Gao
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongwei Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhibo Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Gaocan Ren
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaochang Ma
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
- *Correspondence: Xiaochang Ma
| |
Collapse
|
9
|
Yang Y, Ge FL, Zhan XY, Mu WQ, Li ZY, Lin L, Wei ZY, Bai ZF, Sun Q, Xiao XH. Schisandra chinensis Oil Attenuates Aristolochic Acid I-Induced Nephrotoxicity in vivo and in vitro. Chin J Integr Med 2022; 28:603-611. [PMID: 35391592 DOI: 10.1007/s11655-022-3574-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2022] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To investigate the protective effects of Schisandra chinensis oil (SCEO) against aristolochic acid I (AA I)-induced nephrotoxicity in vivo and in vitro and elucidate the underlying mechanism. METHODS C57BL/6 mice were randomly divided into 5 groups according to a random number table, including control group, AA I group, and AA I +SCEO (0.25, 0.5 and 1 g/kg) groups (n=5 per group). Pretreatment with SCEO was done for 2 days by oral administration, while the control and AA I groups were treated with sodium carboxymethyl cellulose. Mice of all groups except for the control group were injected intraperitoneally with AA I (5 mg/kg) from day 3 until day 7. Histopathological examination and apoptosis of kidney tissue were observed by hematoxylin and eosin and TdT-mediated dUTP nick-end labeling (TUNEL) staining, respectively. The levels of serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), blood urea nitrogen (BUN), and serum creatinine (SCr), as well as renal malondialdehyde (MDA), glutathione, r-glutamyl cysteingl+glycine (GSH), and superoxide dismutase (SOD) were analyzed using enzyme-linked immunosorbent assay (ELISA). Expressions of hepatic cytochrome P450 1A1 (CYP1A1), CYP1A2, and nad(p)hquinonedehydrogenase1 (NQO1) were analyzed using ELISA, quantitative real-time polymerase chain reaction (qPCR) and Western blot, respectively. In vitro, SCEO (40 µ g/mL) was added 12 h before treatment with AA I (40 µ mol/mL for 48 h) in human renal proximal tubule cell line (HK-2), then apoptosis and reactive oxygen species (ROS) were analyzed by flow cytometry. RESULTS SCEO 0.5 and 1 g/kg ameliorated histopathological changes and TUNEL+ staining in the kidney tissues of mice with AA I-induced nephrotoxicity, and reduced serum levels of ALT, AST, BUN and SCr (P<0.01 or P<0.05). SCEO 0.5 and 1 g/kg alleviated the ROS generation in kidney, containing MDA, GSH and SOD (P<0.01 or P<0.05). SCEO 1 g/kg increased the expressions of CYP1A1 and CYP1A2 and decreased NQO1 level in the liver tissues (P<0.01 or P<0.05). Besides, in vitro studies also demonstrated that SCEO 40 µ g/mL inhibited apoptosis and ROS generation (P<0.05 or P<0.01). CONCLUSIONS SCEO can alleviate AA I-induced kidney damage both in vivo and in vitro. The protective mechanism may be closely related to the regulation of metabolic enzymes, thereby inhibiting apoptosis and ROS production.
Collapse
Affiliation(s)
- Yan Yang
- School of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, Sichuan Province, 646000, China.,Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China.,China Military Institute of Chinese Materia, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Fei-Lin Ge
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China.,China Military Institute of Chinese Materia, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Xiao-Yan Zhan
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China.,China Military Institute of Chinese Materia, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Wen-Qing Mu
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Zhi-Yong Li
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Li Lin
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Zi-Ying Wei
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Zhao-Fang Bai
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China.,China Military Institute of Chinese Materia, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| | - Qin Sun
- School of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, Sichuan Province, 646000, China.
| | - Xiao-He Xiao
- Senior Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China.,China Military Institute of Chinese Materia, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
| |
Collapse
|
10
|
Samuel CS, Bennett RG. Relaxin as an anti-fibrotic treatment: Perspectives, challenges and future directions. Biochem Pharmacol 2021; 197:114884. [PMID: 34968489 DOI: 10.1016/j.bcp.2021.114884] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 02/07/2023]
Abstract
Fibrosis refers to the scarring and hardening of tissues, which results from a failed immune system-coordinated wound healing response to chronic organ injury and which manifests from the aberrant accumulation of various extracellular matrix components (ECM), primarily collagen. Despite being a hallmark of prolonged tissue damage and related dysfunction, and commonly associated with high morbidity and mortality, there are currently no effective cures for its regression. An emerging therapy that meets several criteria of an effective anti-fibrotic treatment, is the recombinant drug-based form of the human hormone, relaxin (also referred to as serelaxin, which is bioactive in several other species). This review outlines the broad anti-fibrotic and related organ-protective roles of relaxin, mainly from studies conducted in preclinical models of ageing and fibrotic disease, including its ability to ameliorate several aspects of fibrosis progression and maturation, from immune cell infiltration, pro-inflammatory and pro-fibrotic cytokine secretion, oxidative stress, organ hypertrophy, cell apoptosis, myofibroblast differentiation and ECM production, to its ability to facilitate established ECM degradation. Studies that have compared and/or combined these therapeutic effects of relaxin with current standard of care medication have also been discussed, along with the main challenges that have hindered the translation of the anti-fibrotic efficacy of relaxin to the clinic. The review then outlines the future directions as to where scientists and several pharmaceutical companies that have recognized the therapeutic potential of relaxin are working towards, to progress its development as a treatment for human patients suffering from various fibrotic diseases.
Collapse
Affiliation(s)
- Chrishan S Samuel
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia; Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3052, Australia.
| | - Robert G Bennett
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; Department of Internal Medicine, Division of Diabetes, Endocrinology & Metabolism, University of Nebraska Medical Center, Omaha, NE 68198-4130, USA.
| |
Collapse
|
11
|
Jin C, Miao X, Zhong Y, Han J, Liu Q, Zhu J, Xia X, Peng X. The renoprotective effect of diosgenin on aristolochic acid I-induced renal injury in rats: impact on apoptosis, mitochondrial dynamics and autophagy. Food Funct 2021; 11:7456-7467. [PMID: 32789347 DOI: 10.1039/d0fo00401d] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Aristolochic acid I (AA-I) remains a leading cause of aristolochic acid nephropathy (AAN), however few prevention and treatment strategies exist. In this work, the nephroprotective effect of diosgenin, a steroidal saponin distributed abundantly in several plants, on AA-I-induced renal injury and its underlying mechanism were investigated. Sprague-Dawley rats were intragastrically administered with 30 mg kg-1 d-1 diosgenin two hours before exposure to 10 mg kg-1 d-1 AA-I for consecutive four weeks, and the histological change, the renal and liver function, apoptosis, autophagy and the involved pathways were investigated. The results showed that diosgenin relieved AA-I-induced renal histological damage, including mild edematous disorder of renal tubular arrangement and widening of renal tubular lumen. No obvious changes in the hepatic tissue structure were observed in all treatment groups. Moreover, diosgenin up-regulated the expression of Bcl-2 and down-regulated Bax, and subsequently inhibited AIF expression and the cleaved form of Caspase-3, thereby alleviating apoptosis triggered by AA-I. Diosgenin also mitigated AA-I-induced renal mitochondrial dynamics disorder by increasing the expression of mitochondrial dynamics-related proteins including DRP1 and MFN2. Diosgenin inhibited AA-I-evoked autophagy via ULK1-mediated inhibition of the mTOR pathway. Overall, these results suggest that diosgenin has a protective effect against AA-I-induced renal damage and it may be a potential agent for preventing AA-I-induced AAN.
Collapse
Affiliation(s)
- Chengni Jin
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Xin Miao
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Yujie Zhong
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Jiahui Han
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Qi Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Jiachang Zhu
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Xiaodong Xia
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Xiaoli Peng
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China. and Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University (BTBU), Beijing, 100048, China
| |
Collapse
|
12
|
Deng HF, Yue LX, Wang NN, Zhou YQ, Zhou W, Liu X, Ni YH, Huang CS, Qiu LZ, Liu H, Tan HL, Tang XL, Wang YG, Ma ZC, Gao Y. Mitochondrial Iron Overload-Mediated Inhibition of Nrf2-HO-1/GPX4 Assisted ALI-Induced Nephrotoxicity. Front Pharmacol 2021; 11:624529. [PMID: 33584308 PMCID: PMC7873870 DOI: 10.3389/fphar.2020.624529] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/17/2020] [Indexed: 01/31/2023] Open
Abstract
Aristolactam I (ALI) is an active component derived from some Traditional Chinese medicines (TCMs), and also the important metabolite of aristolochic acid. Long-term administration of medicine-containing ALI was reported to be related to aristolochic acid nephropathy (AAN), which was attributed to ALI-induced nephrotoxicity. However, the toxic mechanism of action involved is still unclear. Recently, pathogenic ferroptosis mediated lipid peroxidation was demonstrated to cause kidney injury. Therefore, this study explored the role of ferroptosis induced by mitochondrial iron overload in ALI-induced nephrotoxicity, aiming to identify the possible toxic mechanism of ALI-induced chronic nephropathy. Our results showed that ALI inhibited HK-2 cell activity in a dose-dependent manner and significantly suppressed glutathione (GSH) levels, accompanying by significant increases in intracellular 4-hydroxynonenal (4-HNE) and intracellular iron ions. Moreover, the ALI-mediated cytotoxicity could be reversed by deferoxamine mesylate (DFO). Compared with other inhibitors, Ferrostatin-1 (Fer-1), a ferroptosis inhibitor, obviously alleviated ALI-induced cytotoxicity. Furthermore, we have shown that ALI could remarkably increase the levels of superoxide anion and ferrous ions in mitochondria, and induce mitochondrial damage and condensed mitochondrial membrane density, the morphological characteristics of ferroptosis, all of which could be reversed by DFO. Interestingly, ALI dose-dependently inhibited these protein contents of nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), and glutathione peroxidase 4 (GPX4), which could be partly rescued by Tin-protoporphyrin IX (SnPP) and mitoTEMPO co-treatment. In conclusion, our results demonstrated that mitochondrial iron overload-mediated antioxidant system inhibition would assist ALI-induced ferroptosis in renal tubular epithelial cells, and Nrf2-HO-1/GPX4 antioxidative system could be an important intervention target to prevent medicine containing ALI-induced nephropathy.
Collapse
Affiliation(s)
- Hui-Fang Deng
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Lan-Xin Yue
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Ning-Ning Wang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China.,Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yong-Qiang Zhou
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Wei Zhou
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Xian Liu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Yu-Hao Ni
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Cong-Shu Huang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China.,School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Li-Zhen Qiu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China.,Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hong Liu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China.,School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Hong-Ling Tan
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Xiang-Lin Tang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Yu-Guang Wang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Zeng-Chun Ma
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Yue Gao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China.,Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
13
|
Li Y, Xu H, Cai D, Zhu S, Liu X, Zhao Y, Zhang Z, Bian Y, Xue M, Zhang L. Integration of transcriptomic, proteomic and metabolomic data to reveal the biological mechanisms of AAI injury in renal epithelial cells. Toxicol In Vitro 2020; 70:105054. [PMID: 33212167 DOI: 10.1016/j.tiv.2020.105054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 11/06/2020] [Accepted: 11/14/2020] [Indexed: 10/23/2022]
Abstract
Overexposure to aristolochic acid I (AAI) can induce aristolochic acid nephropathy (AAN). However, the comprehensive mechanisms of AAI-induced nephrotoxicity have not been entirely explicated. To investigate the toxicological mechanisms by which AAI induces renal injury, human kidney cells (HK-2 cells) were subjected to comprehensive transcriptomic, proteomic and metabolomic analyses. The transcriptomic analysis identified a total of 7749 differentially expressed genes (DEGs) after AAI treatment, while the proteomic analysis found 598 differentially expressed proteins (DEPs) after AAI treatment. The metabolomic analysis revealed 49 and 42 differentially expressed metabolites (DEMs) in the positive and negative ion modes, respectively. Gene Ontology (GO) functional annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were performed on these DEGs, DEPs and DEMs. The results of the comprehensive analyses of transcripts, proteins, and metabolites indicated that the DEGs, DEPs, and DEMs were jointly regulated in three ways. These genes, proteins and metabolites and their related dysregulated pathways may be promising targets for research on the mechanisms of AAI injury in human renal epithelial cells. This study provides large-scale omics data that can be used to formulate new strategies for the prevention, rapid diagnosis, and treatment of AAI injury.
Collapse
Affiliation(s)
- Yu Li
- School of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, PR China
| | - Houxi Xu
- Key Laboratory of Acupuncture and Medicine Research of the Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210046, China
| | - Danhong Cai
- School of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, PR China
| | - Sirui Zhu
- School of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, PR China
| | - Xiaoli Liu
- School of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, PR China
| | - Ye Zhao
- School of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, PR China
| | - Zhaofeng Zhang
- School of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, PR China
| | - Yaoyao Bian
- School of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, PR China
| | - Mei Xue
- College of Basic Medical Sciences, Institute of TCM-related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing 210023, PR China.
| | - Liang Zhang
- School of Pharmacy, Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, PR China.
| |
Collapse
|
14
|
Gan QX, Wang J, Hu J, Lou GH, Xiong HJ, Peng CY, Huang QW. Modulation of Apoptosis by Plant Polysaccharides for Exerting Anti-Cancer Effects: A Review. Front Pharmacol 2020; 11:792. [PMID: 32536869 PMCID: PMC7267062 DOI: 10.3389/fphar.2020.00792] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 05/13/2020] [Indexed: 12/17/2022] Open
Abstract
Cancer has become a significant public health problem with high disease burden and mortality. At present, radiotherapy and chemotherapy are the main means of treating cancer, but they have shown serious safety problems. The severity of this problem has caused further attention and research on effective and safe cancer treatment methods. Polysaccharides are natural products with anti-cancer activity that are widely present in a lot of plants, and many studies have found that inducing apoptosis of cancer cells is one of their important mechanisms. Therefore, this article reviews the various ways in which plant polysaccharides promote apoptosis of cancer cells. The major apoptotic pathways involved include the mitochondrial pathway, the death receptor pathway, and their upstream signal transduction such as MAPK pathway, PI3K/AKT pathway, and NF-κB pathway. Moreover, the paper has also been focused on the absorption and toxicity of plant polysaccharides with reference to extant literature, making the research more scientific and comprehensive. It is hoped that this review could provide some directions for the future development of plant polysaccharides as anticancer drugs in pharmacological experiments and clinical researches.
Collapse
Affiliation(s)
- Qing-Xia Gan
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jin Wang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ju Hu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Guan-Hua Lou
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hai-Jun Xiong
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng-Yi Peng
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qin-Wan Huang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
15
|
Anger EE, Yu F, Li J. Aristolochic Acid-Induced Nephrotoxicity: Molecular Mechanisms and Potential Protective Approaches. Int J Mol Sci 2020; 21:E1157. [PMID: 32050524 PMCID: PMC7043226 DOI: 10.3390/ijms21031157] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 01/21/2020] [Accepted: 02/05/2020] [Indexed: 12/13/2022] Open
Abstract
Aristolochic acid (AA) is a generic term that describes a group of structurally related compounds found in the Aristolochiaceae plants family. These plants have been used for decades to treat various diseases. However, the consumption of products derived from plants containing AA has been associated with the development of nephropathy and carcinoma, mainly the upper urothelial carcinoma (UUC). AA has been identified as the causative agent of these pathologies. Several studies on mechanisms of action of AA nephrotoxicity have been conducted, but the comprehensive mechanisms of AA-induced nephrotoxicity and carcinogenesis have not yet fully been elucidated, and therapeutic measures are therefore limited. This review aimed to summarize the molecular mechanisms underlying AA-induced nephrotoxicity with an emphasis on its enzymatic bioactivation, and to discuss some agents and their modes of action to reduce AA nephrotoxicity. By addressing these two aspects, including mechanisms of action of AA nephrotoxicity and protective approaches against the latter, and especially by covering the whole range of these protective agents, this review provides an overview on AA nephrotoxicity. It also reports new knowledge on mechanisms of AA-mediated nephrotoxicity recently published in the literature and provides suggestions for future studies.
Collapse
Affiliation(s)
| | | | - Ji Li
- Department of Clinical Pharmacy, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (E.E.A.); (F.Y.)
| |
Collapse
|
16
|
Licorice Extracts Attenuate Nephrotoxicity Induced by Brucine Through Suppression of Mitochondria Apoptotic Pathway and STAT3 Activation. Curr Med Sci 2019; 39:890-898. [DOI: 10.1007/s11596-019-2126-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 05/06/2019] [Indexed: 02/07/2023]
|
17
|
Liu B, Lin J, Bai L, Zhou Y, Lu R, Zhang P, Chen D, Li H, Song J, Liu X, Wu Y, Wu J, Liang C, Zhou J. Paeoniflorin Inhibits Mesangial Cell Proliferation and Inflammatory Response in Rats With Mesangial Proliferative Glomerulonephritis Through PI3K/AKT/GSK-3β Pathway. Front Pharmacol 2019; 10:978. [PMID: 31551783 PMCID: PMC6745507 DOI: 10.3389/fphar.2019.00978] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 07/31/2019] [Indexed: 12/11/2022] Open
Abstract
Mesangial proliferative glomerulonephritis (MPGN) is the most common type of chronic kidney disease in China, characterized by mesangial cell proliferation and inflammatory response. Paeoniflorin, an effective composition extracted from Radix Paeoniae Alba, has been used for various kinds of kidney diseases. However, there are no studies reporting the effects of paeoniflorin on MPGN. The present study aims to investigate whether paeoniflorin plays a role in MPGN and confirm the underlying molecular mechanisms. Our results manifested that paeoniflorin strongly restrained 24 h urinary protein and promoted renal function and dyslipidemia in a MPGN rat model. Moreover, paeoniflorin attenuated mesangial cell proliferation and inflammation both in MPGN rats and human mesangial cells (HMCs) treated with lipopolysaccharide (LPS). In detail, paeoniflorin decreased the number of mesangial cells and expressions of proliferation marker Ki67 in MPGN rats. Paeoniflorin also inhibited HMC proliferation and blocked cell cycle progression. In addition, the contents of inflammatory factors and the expressions of macrophage marker iNOS were decreased after paeoniflorin treatment. Furthermore, we found that the protective effect of paeoniflorin was accompanied by a strong inhibition of the phosphatidylinositol 3-kinase (PI3K)/AKT/glycogen synthase kinase (GSK)-3β pathway. Paeoniflorin enhanced the inhibitory effect of PI3K inhibitor LY294002 and suppressed the activated effect of PI3K agonist insulin-like growth factor 1 (IGF-1) on PI3K/AKT/GSK-3β pathway. In conclusion, these results demonstrated that paeoniflorin ameliorates MPGN by inhibiting mesangial cell proliferation and inflammatory response through the PI3K/AKT/GSK-3β pathway.
Collapse
Affiliation(s)
- Bihao Liu
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jin Lin
- College of Chinese Materia Medica, Guangdong Food and Drug Vocational College, Guangzhou, China
| | - Lixia Bai
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuan Zhou
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ruirui Lu
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peichun Zhang
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dandan Chen
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Honglian Li
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianping Song
- Science and Technology Industrial Park, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xusheng Liu
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yidan Wu
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Junbiao Wu
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chunling Liang
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiuyao Zhou
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
18
|
Sun J, Zheng S, Yang N, Chen B, He G, Zhu T. Dexmedetomidine inhibits apoptosis and expression of COX-2 induced by lipopolysaccharide in primary human alveolar epithelial type 2 cells. Biochem Biophys Res Commun 2019; 517:89-95. [DOI: 10.1016/j.bbrc.2019.07.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 07/06/2019] [Indexed: 01/23/2023]
|
19
|
Valkovic AL, Bathgate RA, Samuel CS, Kocan M. Understanding relaxin signalling at the cellular level. Mol Cell Endocrinol 2019; 487:24-33. [PMID: 30592984 DOI: 10.1016/j.mce.2018.12.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 12/19/2018] [Accepted: 12/22/2018] [Indexed: 02/07/2023]
Abstract
The peptide hormone relaxin mediates many biological actions including anti-fibrotic, vasodilatory, angiogenic, anti-inflammatory, anti-apoptotic, and organ protective effects across a range of tissues. At the cellular level, relaxin binds to the G protein-coupled receptor relaxin family peptide receptor 1 (RXFP1) to activate a variety of downstream signal transduction pathways. This signalling cascade is complex and also varies in diverse cellular backgrounds. Moreover, RXFP1 signalling shows crosstalk with other receptors to mediate some of its physiological functions. This review summarises known signalling pathways induced by acute versus chronic treatment with relaxin across a range of cell types, it describes RXFP1 crosstalk with other receptors, signalling pathways activated by other ligands targeting RXFP1, and it also outlines physiological relevance of RXFP1 signalling outputs. Comprehensive understanding of the mechanism of relaxin actions in fibrosis, vasodilation, as well as organ protection, will further support relaxin's clinical potential.
Collapse
Affiliation(s)
- Adam L Valkovic
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Ross Ad Bathgate
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, 3010, Australia; Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, 3052, Australia.
| | - Chrishan S Samuel
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, 3800, Australia
| | - Martina Kocan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, 3010, Australia.
| |
Collapse
|
20
|
Receptor-independent modulation of TGF-β-induced pro-fibrotic pathways by relaxin-2 in human primary tubular epithelial cells. Cell Tissue Res 2018; 374:619-627. [PMID: 30078103 DOI: 10.1007/s00441-018-2904-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 07/24/2018] [Indexed: 10/28/2022]
Abstract
Renal tubular epithelial cells actively contribute to the development of renal fibrosis and may be targeted by anti-fibrotic drugs. Relaxin-2 (RLX2) applied as recombinant protein is suggested to be renoprotective. Therefore, we investigated whether human primary tubular epithelial cells (hPTEC) obtained from various donors were target cells for the anti-fibrotic actions of RLX2. Treatment of hPTEC with RLX2 reduced the TGF-β1-induced secretion of the pro-fibrotic factor CTGF (connective tissue growth factor) and inhibited fibronectin synthesis and secretion. Furthermore, metalloproteinase MMP2 secretion was increased, with no effect on MMP9. Considerable differences were observed between hPTEC obtained from different donors. Therefore, expression of the relaxin family peptide receptor RXFP1, the major mediator of renal RLX2 effects, was analyzed. A validated antibody detected a double band of 80-90 kDa in cellular homogenates by Western blotting. Expression of the detected protein was not altered by incubation with TGF-β1 and RLX2-induced modulation of CTGF expression did not correlate with the putative receptor expression. Therefore, relaxin family receptors RXFP1-4 were assessed by RNA-seq analysis. No evidence was found for mRNA expression of any of these receptors in several hPTEC preparations. Lack of RXFP1 mRNA was confirmed by qPCR using mRNA obtained from THP-1 cells as positive control. Our data thus provide evidence for primary renal human tubular epithelial cells as targets for the anti-fibrotic actions of RLX2. However, anti-fibrotic effects were observed at micromolar concentrations of RLX2 and shown to be independent of RXFP1 expression.
Collapse
|
21
|
Aragón-Herrera A, Feijóo-Bandín S, Rodríguez-Penas D, Roselló-Lletí E, Portolés M, Rivera M, Bigazzi M, Bani D, Gualillo O, González-Juanatey JR, Lago F. Relaxin activates AMPK-AKT signaling and increases glucose uptake by cultured cardiomyocytes. Endocrine 2018; 60:103-111. [PMID: 29411306 DOI: 10.1007/s12020-018-1534-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 01/15/2018] [Indexed: 12/22/2022]
Abstract
PURPOSE Many evidences show that the hormone relaxin plays a pivotal role in the physiology and pathology of the cardiovascular system. This pleiotropic hormone exerts regulatory functions through specific receptors in cardiovascular tissues: in experimental animal models it was shown to induce coronary vasodilation, prevent cardiac damage induced by ischemia/reperfusion and revert cardiac hypertrophy and fibrosis. A tight relationship between this hormone and important metabolic pathways has been suggested, but it is at present unknown if relaxin could regulate cardiac metabolism. Our aim was to study the possible effects of relaxin on cardiomyocyte metabolism. METHODS Neonatal rat cardiomyocytes were treated with relaxin and (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide) assays (MTT) were performed to assess metabolic activity; while 2-deoxy-D-[3H] glucose and BODIPY-labelled fatty acid incorporations were analyzed to measure glucose and fatty acid uptakes, and western blot was utilized to study the intracellular signaling pathways activated by the hormone. RESULTS We observed that relaxin at 10 ng/ml was able to increase the level of metabolic activity of cultured neonatal rat cardiomyocytes; the rate of 2-deoxy-D-[3H]glucose incorporation demonstrated that relaxin also induced an increase in glucose uptake. First evidence is also offered that relaxin can activate the master energy sensor and regulator AMPK in cardiomyocytes. Moreover, the treatment of cardiomyocytes with relaxin also induced dose-dependent increases in ERK1/2, AKT, and AS160 phosphorylation. That raise in AS160 phosphorylation induced by relaxin was prevented by the pretreatment with AMPK and AKT pathways inhibitors, indicating that both molecules play important roles in the relaxin effects reported. CONCLUSION Relaxin can regulate cardiomyocyte metabolism and activate AMPK, the central sensor of energy status that maintains cellular energy homeostasis, and also ERK and AKT, two molecular sensing nodes that coordinate dynamic responses of the cell's metabolic responses.
Collapse
Affiliation(s)
- A Aragón-Herrera
- Cellular and Molecular Cardiology Unit and Department of Cardiology, Institute of Biomedical Research (IDIS-SERGAS), Santiago de Compostela, Spain
| | - S Feijóo-Bandín
- Cellular and Molecular Cardiology Unit and Department of Cardiology, Institute of Biomedical Research (IDIS-SERGAS), Santiago de Compostela, Spain.
- CIBERCV, Institute of Health Carlos III, Madrid, Spain.
| | - D Rodríguez-Penas
- Cellular and Molecular Cardiology Unit and Department of Cardiology, Institute of Biomedical Research (IDIS-SERGAS), Santiago de Compostela, Spain
| | - E Roselló-Lletí
- CIBERCV, Institute of Health Carlos III, Madrid, Spain
- Cardiocirculatory Unit, Health Institute of La Fe University Hospital (IIS La Fe), Valencia, Spain
| | - M Portolés
- CIBERCV, Institute of Health Carlos III, Madrid, Spain
- Cardiocirculatory Unit, Health Institute of La Fe University Hospital (IIS La Fe), Valencia, Spain
| | - M Rivera
- CIBERCV, Institute of Health Carlos III, Madrid, Spain
- Cardiocirculatory Unit, Health Institute of La Fe University Hospital (IIS La Fe), Valencia, Spain
| | - M Bigazzi
- Prosperius Institute, Florence, Italy
| | - D Bani
- Prosperius Institute, Florence, Italy
| | - O Gualillo
- Neuroendocrine Interaccions in Rheumatology and Inflammatory Diseases Unit, Institute of Biomedical Research (IDIS-SERGAS), Santiago de Compostela, Spain
| | - J R González-Juanatey
- Cellular and Molecular Cardiology Unit and Department of Cardiology, Institute of Biomedical Research (IDIS-SERGAS), Santiago de Compostela, Spain
- CIBERCV, Institute of Health Carlos III, Madrid, Spain
| | - F Lago
- Cellular and Molecular Cardiology Unit and Department of Cardiology, Institute of Biomedical Research (IDIS-SERGAS), Santiago de Compostela, Spain
- CIBERCV, Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
22
|
Fan Y, Chen H, Peng H, Huang F, Zhong J, Zhou J. Molecular Mechanisms of Curcumin Renoprotection in Experimental Acute Renal Injury. Front Pharmacol 2017; 8:912. [PMID: 29311922 PMCID: PMC5733093 DOI: 10.3389/fphar.2017.00912] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 11/29/2017] [Indexed: 11/13/2022] Open
Abstract
As a highly perfused organ, the kidney is especially sensitive to ischemia and reperfusion. Ischemia-reperfusion (IR)-induced acute kidney injury (AKI) has a high incidence during the perioperative period in the clinic and is an important link in ischemic acute renal failure (IARF). Therefore, IR-induced AKI has important clinical significance and it is necessary to explore to develop drugs to prevent and alleviate IR-induced AKI. Curcumin [diferuloylmethane, 1,7-bis(4-hydroxy-3-methoxiphenyl)-1,6-heptadiene-3,5-dione)] is a polyphenol compound derived from Curcuma longa (turmeric) and was shown to have a renoprotective effect on ischemia-reperfusion injury (IRI) in a previous study. However, the specific mechanisms underlying the protective role of curcumin in IR-induced AKI are not completely understood. APPL1 is a protein coding gene that has been shown to be involved in the crosstalk between the adiponectin-signaling and insulin-signaling pathways. In the study, to investigate the molecular mechanisms of curcumin effects in kidney ischemia/reperfusion model, we observed the effect of curcumin in experimental models of IR-induced AKI and we found that curcumin treatment significantly increased the expression of APPL1 and inhibited the activation of Akt after IR treatment in the kidney. Our in vitro results showed that apoptosis of renal tubular epithelial cells was exacerbated with hypoxia-reoxygenation (HR) treatment compared to sham control cells. Curcumin significantly decreased the rate of apoptosis in renal tubular epithelial cells with HR treatment. Moreover, knockdown of APPL1 activated Akt and subsequently aggravated apoptosis in HR-treated renal tubular epithelial cells. Conversely, inhibition of Akt directly reversed the effects of APPL1 knockdown. In summary, our study demonstrated that curcumin mediated upregulation of APPL1 protects against ischemia reperfusion induced AKI by inhibiting Akt phosphorylation.
Collapse
Affiliation(s)
- Youling Fan
- Department of Anesthesiology, Panyu Central Hospital, Guangzhou, China
| | - Hongtao Chen
- Department of Anesthesiology, The Eighth People's Hospital of Guangzhou, Guangzhou, China
| | - Huihua Peng
- Department of Anesthesiology, Panyu Central Hospital, Guangzhou, China
| | - Fang Huang
- Department of Anesthesiology, Panyu Central Hospital, Guangzhou, China
| | - Jiying Zhong
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, China
| | - Jun Zhou
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, China
| |
Collapse
|