1
|
Liu M, Yao Y, Tan F, Wang J, Hu R, Du J, Jiang Y, Yuan X. Sodium-glucose co-transporter 2 (SGLT-2) inhibitors ameliorate renal ischemia-reperfusion injury (IRI) by modulating autophagic processes. Transl Res 2025; 277:27-38. [PMID: 39761911 DOI: 10.1016/j.trsl.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 12/24/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025]
Abstract
Renal ischemia-reperfusion injury (IRI) is a common clinical condition that currently lacks effective treatment options. Inhibitors targeting the sodium-glucose co-transporter-2 (SGLT-2), recognized for their role in managing hyperglycemia, have demonstrated efficacy in enhancing the health outcomes for diabetic patients grappling with chronic kidney disease. Nevertheless, the precise impact of SGLT-2 inhibitors on renal ischemia-reperfusion injury (IRI) and the corresponding transcriptomic alterations remain to be elucidated. In our research, we developed a model of IRI using male C57BL/6 mice by clamping the unilateral renal artery and administering empagliflozin Transcriptomic alterations were analyzed using RNA sequencing (RNA-Seq), complemented by proteomic analysis to investigate the effects of empagliflozin. Histological assessments revealed increased renal inflammatory cell infiltration, widespread renal tubular injury, and elevated autophagosomes formation in the IRI group compared to controls. These pathological changes were significantly attenuated following empagliflozin treatment. Besides, renal function impairment can be alleviated in empagliflozin-treated group. RNA-Seq analysis identified lysosomal autophagy as a key biological process in IRI mice. Empagliflozin exerted a renoprotective effect by downregulating lysosome-associated membrane proteins, primarily LAMP1, LAMP2, and LAMP4 (CD68), through the PI3K-Akt, MAPK, and mTOR signaling pathways, thereby inhibiting autophagic processes. In conclusion, this study highlights enhanced inflammation and disrupted metabolism as hallmark transcriptomic signatures of renal. Furthermore, it demonstrates the renoprotective effects of empagliflozin in alleviating renal IRI by modulating autophagic processes.
Collapse
Affiliation(s)
- Mengmeng Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Yuanqing Yao
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Fangyan Tan
- Department of Nephrology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Jing Wang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Rong Hu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Jianlin Du
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Yonghong Jiang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China.
| | - Xin Yuan
- Department of Nephrology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China.
| |
Collapse
|
2
|
Jiang B, Liu J, Qu Z, Wang Y, Wang Y, Li Z, Jin X, Lao Y, He R, Yang S. Mechanism of dihydroartemisinin in the treatment of ischaemia/reperfusion-induced acute kidney injury via network pharmacology, molecular dynamics simulation and experiments. Int Immunopharmacol 2025; 144:113705. [PMID: 39626534 DOI: 10.1016/j.intimp.2024.113705] [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: 09/11/2024] [Revised: 11/12/2024] [Accepted: 11/21/2024] [Indexed: 12/15/2024]
Abstract
OBJECTIVE To investigate whether dihydroartemisinin (DHA) attenuates ischaemia-reperfusion injury (IRI)-induced acute kidney injury (AKI) in mice by inhibiting oxidative stress and inflammation and to explore its potential molecular mechanisms. MATERIALS AND METHODS Network pharmacology analysis was used to screen relevant targets, and molecular docking of DHA with core targets was performed. Molecular dynamics simulation of the target with the lowest binding free energy, NQO1-DHA.The renal protective effect of DHA on the IRI-induced AKI mouse model was evaluated. The expression levels of NQO1, Nrf2 and other proteins were detected by Western blotting. The expression levels of Nrf2 and others were detected by immunohistochemistry (IHC) and immunofluorescence (IF). RESULTS Through network pharmacological analysis, we obtained that PI3K/AKT and MAPK signaling pathway may be related to DHA in the treatment of AKI.Molecular dynamics simulation indicated that NQO1 is an important target protein for DHA to exert nephroprotective effects.Moreover, the potential molecular mechanisms were verified by experiments.DHA reduced the serum creatinine (Scr) and urea nitrogen (BUN) levels in AKI mice, significantly improved AKI pathology, alleviated oxidative stress and inflammatory injury, which may be related to its activation of the Nrf2 pathway and regulation of macrophage polarization. CONCLUSIONS Through network pharmacology, molecular dynamics simulation and experimental validation, we initially investigated that DHA alleviate AKI by ameliorating oxidative stress and inflammatory damage, which may be related to its activation of the Nrf2 pathway and the regulation of macrophage polarisation, which lays the foundation for subsequent in-depth study of the specific mechanism of action.
Collapse
Affiliation(s)
- Beibei Jiang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Jiahui Liu
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Ziyi Qu
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Yanqing Wang
- Department of Anatomy,School of Basic Medical Sciences,Guangzhou University of Chinese Medicine, Guangdong, China
| | - Yuzhi Wang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Zhongtang Li
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Xiaoming Jin
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Yunlan Lao
- Central Hospital of Guangdong Provincial Nongken, Guangdong, China
| | - Riming He
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China.
| | - Shudong Yang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China.
| |
Collapse
|
3
|
Zheng Z, Ke L, Ye S, Shi P, Yao H. Pharmacological Mechanisms of Cryptotanshinone: Recent Advances in Cardiovascular, Cancer, and Neurological Disease Applications. Drug Des Devel Ther 2024; 18:6031-6060. [PMID: 39703195 PMCID: PMC11658958 DOI: 10.2147/dddt.s494555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/26/2024] [Indexed: 12/21/2024] Open
Abstract
Cryptotanshinone (CTS) is an important active ingredient of Salvia miltiorrhiza Bge. In recent years, its remarkable pharmacological effects have triggered extensive and in-depth studies. The aim of this study is to retrieve the latest research progress on CTS and provide prospects for future research. The selection of literature for inclusion, data extraction and methodological quality assessment were discussed. Studies included (1) physicochemical and ADME/Tox properties, (2) pharmacological effects and mechanism, (3) conclusion and bioinformatics analysis. A total of 915 titles and abstracts were screened, resulting in 184 papers used in this review; CTS has shown therapeutic effects on a variety of diseases by modulating multiple molecular pathways. For example, CTS primarily targets NF-κB pathway and MAPK pathway to have a therapeutic role in cardiovascular diseases; in cancer, CTS shows superior efficacy through the PI3K/Akt/mTOR pathway and the JAK/STAT pathway; CTS act on the Nrf2/HO-1 pathway to combat neurological diseases. In addition, key targets of CTS were predicted by bioinformatics analysis, referring to disease ontology (DO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and gene ontology (GO) enrichment analysis, with R Studio; AKT1, MAPK1, STAT3, P53 and EGFR are predicted to be the key targets of CTS against diseases. The key proteins were then docked by Autodock software to preliminarily assess their binding activities. This review provided new insights into research of CTS and its potential applications in the future, and especially the targets and directly binding modes for CTS are waiting to be investigated.
Collapse
Affiliation(s)
- Ziyao Zheng
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, People’s Republic of China
| | - Liyuan Ke
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, People’s Republic of China
| | - Shumin Ye
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, People’s Republic of China
| | - Peiying Shi
- Department of Traditional Chinese Medicine Resource and Bee Products, College of Animal Sciences (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou, 350002, People’s Republic of China
| | - Hong Yao
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, People’s Republic of China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, 350122, People’s Republic of China
| |
Collapse
|
4
|
Zhu G, Li D, Wang X, Guo Q, Zhao Y, Hou W, Li J, Zheng Q. Drug monomers from Salvia miltiorrhiza Bge. promoting tight junction protein expression for therapeutic effects on lung cancer. Sci Rep 2023; 13:22928. [PMID: 38129556 PMCID: PMC10739844 DOI: 10.1038/s41598-023-50163-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023] Open
Abstract
Salvia miltiorrhiza Bge. is a traditional Chinese medicine (TCM) that has been used for treatment of various diseases, including cancer by activating blood circulation and removing blood stasis. Tanshinone (TanIIA) and cryptotanshinone (CPT) are major lipophilic compounds extracted from the root of Salvia miltiorrhiza Bge., which are considered to be the effective compounds affecting the efficacy of the anti-tumor therapy of Salvia miltiorrhiza Bge. We have explored the mechanism of CPT and TanIIA exerting inhibition in non-small cell lung cancer (NSCLC) to provide experimental data support for guiding the translational development and clinical application of anti-tumor components of TCM. The subcutaneous tumor model and in vitro culture model of A549 cells was constructed to evaluate CPT and TanIIA's tumour-inhibitory effect respectively. RNA sequencing (RNA-seq) and bioinformatics analysis were conducted to identify differentially expressed genes (DEGs) and signalling pathways related to CPT and TanIIA treatment. qRT-PCR and Western blot were used to explore the mechanism of CPT and TanIIA intervention on NSCLC. Both CPT and TanIIA significantly inhibited the proliferation of A549 tumor cells and tumor growth in animal models. After intervention, the migration ability decreased and the level of apoptosis increased. RNA-seq results showed that both CPT and TanIIA could cause gene differential expression, miR-21-5p as one of the most significant gene expression differences between the two groups, and could act on cell connectivity. CPT and TanIIA play a regulatory role in regulating tight junction proteins (Occludin and ZO1), and Occludin mRNA and protein levels were reduced in an in vitro miR-21-5p overexpression A549 cell model. The mechanisms may be related to the reduction of miR-21-5p expression to increase the level of promoted tight junction protein expression for the purpose of inhibiting proliferation and invasion of NSCLC.
Collapse
Affiliation(s)
- Guanghui Zhu
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Daorui Li
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Xueqian Wang
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Qiujun Guo
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Yuanchen Zhao
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Wei Hou
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Jie Li
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Qi Zheng
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| |
Collapse
|
5
|
Deficiency of mindin reduces renal injury after ischemia reperfusion. Mol Med 2022; 28:152. [PMID: 36510147 PMCID: PMC9743537 DOI: 10.1186/s10020-022-00578-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 11/21/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Acute renal injury (AKI) secondary to ischemia reperfusion (IR) injury continues to be a significant perioperative problem and there is no effective treatment. Mindin belongs to the mindin/F-spondin family and involves in inflammation, proliferation, and cell apoptosis. Previous studies have explored the biological functions of mindin in liver and brain ischemic injury, but its role in AKI is unknown. METHOD To investigate whether mindin has a pathogenic role, mindin knockout (KO) and wild-type (WT) mice were used to establish renal IR model. After 30 min of ischemia and 24 h of reperfusion, renal histology, serum creatinine, and inflammatory response were examined to assess kidney injury. In vitro, proinflammatory factors and inflammatory signaling pathways were measured in mindin overexpression or knockdown and vector cells after hypoxia/reoxygenation (HR). RESULTS Following IR, the kidney mindin level was increased in WT mice and deletion of mindin provided significant protection for mice against IR-induced renal injury as manifested by attenuated the elevation of serum creatinine and blood urea nitrogen along with less severity for histological alterations. Mindin deficiency significantly suppressed inflammatory cell infiltration, TNF-α and MCP-1 production following renal IR injury. Mechanistic studies revealed that mindin deficiency inhibits TLR4/JNK/NF-κB signaling activation. In vitro, the expression levels of TNF-α and MCP-1 were increased in mindin overexpression cells compared with vector cells following HR. Moreover, TLR4/JNK/NF-κB signaling activation was elevated in the mindin overexpression cells in response to HR stimulation while mindin knockdown inhibited the activation of TLR4/JNK/ NF-κB signaling after HR in vitro. Further study showed that mindin protein interacted directly with TLR4 protein. And more, mindin protein was confirmed to be expressed massively in renal tubule tissues of human hydronephrosis patients. CONCLUSION These data demonstrate that mindin is a critical modulator of renal IR injury through regulating inflammatory responses. TLR4/JNK/NF-κB signaling most likely mediates the biological function of mindin in this model of renal ischemia.
Collapse
|
6
|
Chen J, Yuan S, Zhou J, Huang X, Wu W, Cao Y, Liu H, Hu Q, Li X, Guan X, Yin S, Jiang J, Zhou Y, Zhou J. Danshen injection induces autophagy in podocytes to alleviate nephrotic syndrome via the PI3K/AKT/mTOR pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 107:154477. [PMID: 36215790 DOI: 10.1016/j.phymed.2022.154477] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/12/2022] [Accepted: 09/24/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Danshen injection (DSI) is an agent extracted from the Salvia miltiorrhiza Bunge, a natural drug commonly used to alleviate kidney diseases. However, the material basis and therapeutic effects of DSI on nephrotic syndrome (NS) remain unclear. PURPOSE To investigate the material basis of DSI and the therapeutic effects and underlying mechanisms of NS. METHODS NS models were established using adriamycin-induced BALB/c mice and lipopolysaccharide-induced mouse podocytes (MPC-5). Following DSI and prednisone administration, kidney coefficients, 24 h urine protein, blood urea nitrogen, and serum creatinine levels were tested. Histomorphology was observed by periodic acid-Schiff staining and hematoxylin and eosin staining of the kidney sections. The glomerular basement membrane and autophagosomes of the kidneys were observed using transmission electron microscopy. Nephrin and desmin levels in the glomeruli were tested using immunohistochemistry. The viability of MPC-5 cells was tested using cell counting kit-8 after chloroquine and rapamycin administration in combination with DSI. The in vivo and in vitro protein levels of phosphatidylinositol 3-kinase (PI3K), AKT, phosphorylated AKT (Ser473), mammalian target of rapamycin (mTOR), microtubule-associated protein light chain 3 (LC3), beclin1, cleaved caspase-3, and caspase-3 were detected using western blotting. RESULTS Our results showed that DSI contained nine main components: caffeic acid, danshensu, lithospermic acid, rosmarinic acid, salvianolic acid A, salvianolic acid B, salvianolic acid C, salvianolic acid D, and 3, 4-Dihydroxybenzaldehyde. In in vivo studies, the NS mice showed renal function and pathological impairment. Podocytes were damaged, with decreased levels of autophagy and apoptosis, accompanied by inhibition of the PI3K/AKT/mTOR signaling. DSI administration resulted in improved renal function and pathology in NS mice, with the activation of autophagy and PI3K/AKT/mTOR signaling in the kidneys. Additionally, podocytes were less damaged and intracellular autophagosomes were markedly increased. In vitro studies have shown that DSI activated MPC-5 autophagy and reduced apoptosis via the PI3K/AKT/mTOR pathway. CONCLUSION Collectively, this study demonstrated that DSI activated podocyte autophagy and reduced apoptosis via the PI3K/AKT/mTOR signaling, ultimately attenuating NS. Our study clarified the main components of DSI and elucidated its therapeutic effects and potential mechanisms for NS, providing new targets and agents for the clinical treatment of NS.
Collapse
Affiliation(s)
- Junqi Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Shengliang Yuan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Jie Zhou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Xiuye Huang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Wenjia Wu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Yiwen Cao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Hong Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Qinghong Hu
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Xiaojie Li
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Xueping Guan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Simin Yin
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Jiaying Jiang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Yuan Zhou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangdong, China.
| | - Jiuyao Zhou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangdong, China.
| |
Collapse
|
7
|
Zhao Q, Bai J, Chen Y, Liu X, Zhao S, Ling G, Jia S, Zhai F, Xiang R. An optimized herbal combination for the treatment of liver fibrosis: Hub genes, bioactive ingredients, and molecular mechanisms. JOURNAL OF ETHNOPHARMACOLOGY 2022; 297:115567. [PMID: 35870684 DOI: 10.1016/j.jep.2022.115567] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/30/2022] [Accepted: 07/15/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Liver fibrosis is a chronic liver disease that can lead to cirrhosis, liver failure, and hepatocellular carcinoma, and it is associated with long-term adverse outcomes and mortality. As a primary resource for complementary and alternative medicine, traditional Chinese medicine (TCM) has accumulated a large number of effective formulas for the treatment of liver fibrosis in clinical practice. However, studies on how to systematically optimize TCM formulas are still lacking. AIM OF THE REVIEW To provide a methodological reference for the systematic optimization of TCM formulae against liver fibrosis and explored the underlying molecular mechanisms; To provide an efficient method for searching for lead compounds from natural sources and developing from herbal medicines; To enable clinicians and patients to make more reasonable choices and promote the effective treatment toward those patients with liver fibrosis. MATERIALS AND METHODS TCM formulas related to treating liver fibrosis were collected from the Web of Science, PubMed, the China National Knowledge Infrastructure (CNKI), Wan Fang, and the Chinese Scientific Journals Database (VIP). Furthermore, the TCM compatibility patterns were mined using association analysis. The core TCM combinations were found by designing an optimized formulas algorithm. Finally, the hub target proteins, potential molecular mechanisms, and active compounds were explored through integrative pharmacology and docking-based inverse virtual screening (IVS) approaches. RESULTS We found that the herbs for reinforcing deficiency, activating blood, removing blood stasis, and clearing heat were the basis of TCM formulae patterns. Furthermore, the combination of Salviae Miltiorrhizae (Salvia miltiorrhiza Bunge; Chinese salvia/Danshen), Astragali Radix (Astragalus membranaceus (Fisch.) Bunge; Astragalus/Huangqi), and Radix Bupleuri (Bupleurum chinense DC.; Bupleurum/Chaihu) was identified as core groups. A total of six targets (TNF, STAT3, EGFR, IL2, ICAM1, PTGS2) play a pivotal role in TCM-mediated liver fibrosis inhibition. (-)-Cryptotanshinone, Tanshinaldehyde, Ononin, Thymol, Daidzein, and Formononetin were identified as active compounds in TCM. And mechanistically, TCM could affect the development of liver fibrosis by regulating inflammation, immunity, angiogenesis, antioxidants, and involvement in TNF, MicroRNAs, Jak-STAT, NF-kappa B, and C-type lectin receptors (CLRs) signaling pathways. Molecular docking results showed that key components had good potential to bind to the target genes. CONCLUSION In summary, this study provides a methodological reference for the systematic optimization of TCM formulae and exploration of underlying molecular mechanisms.
Collapse
Affiliation(s)
- Qianqian Zhao
- Faculty of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Jinwei Bai
- School of Medical Equipment, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Yiwei Chen
- Faculty of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Xin Liu
- Faculty of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Shangfeng Zhao
- Faculty of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Guixia Ling
- School of Medical Equipment, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Shubing Jia
- Faculty of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Fei Zhai
- School of Medical Equipment, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Rongwu Xiang
- School of Medical Equipment, Shenyang Pharmaceutical University, Shenyang, 110016, China; Liaoning Professional Technology Innovation Center on Medical Big Data and Artificial Intelligence, Shenyang, 110016, China.
| |
Collapse
|
8
|
Repression of inflammatory pathways with Boswellia for alleviation of liver injury after renal ischemia reperfusion. Life Sci 2022; 306:120799. [PMID: 35863426 DOI: 10.1016/j.lfs.2022.120799] [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: 04/22/2022] [Revised: 07/01/2022] [Accepted: 07/08/2022] [Indexed: 11/22/2022]
Abstract
AIM Acute kidney injury (AKI) is a sudden incident that is linked with a high lethality rate commonly due to distant organ injury. This study aims to explore the role of standardized Boswellia serrata (containing 35 % boswellic acid) in attenuating kidney and liver damage in a model of rats with renal insult. MAIN METHODS Sprague-Dawley rats, exposed to renal injury via ischemia-reperfusion model, were administered a daily regimen of 1000 or 2000 mg/kg Boswellia for seven days then rats were sacrificed on day eight. Alanine aminotransferase, aspartate aminotransferase, serum creatinine and blood urea nitrogen, were assayed. TLR9, oxidative stress markers; namely MDA and GSH, inflammatory cytokines; namely, IL-6, IL-1β, and TNF-α, as well as NF-κB were also measured. KEY FINDINGS Renal ischemia-reperfusion injury (IRI) impaired renal and liver function significantly, but Boswellia attenuated this impairment in a dose-dependent fashion. Histopathological assessment of kidney and liver confirmed that Boswellia decreased damage severity. A marked increase in TLR9, NF-κB, IL-6, IL-1β, TNF-α, and MDA besides decreased GSH levels were observed in the kidney and liver after renal IRI. Boswellia attenuated increases in TLR9, NF-κB, IL-1β, TNF-α, and IL-6 and boosted antioxidant defences via decreasing MDA and increasing GSH in kidney and liver. Anti-inflammatory and antioxidant effects of Boswellia were mostly comparable to those of silymarin. SIGNIFICANCE We conclude that the anti-inflammatory and antioxidant effects of Boswellia could be beneficial in ameliorating kidney and liver damage after AKI and that TLR9 might be the connection that signals liver injury in response to renal damage.
Collapse
|
9
|
Fan LM, Zhang YQ, Chen YP, Chen LL, Xu WH, Nan LH, Xu W, Lu B, Wang Y, Chu KD, Zhang JP. Cryptotanshinone ameliorates dextran sulfate sodium-induced murine acute and chronic ulcerative colitis via suppressing STAT3 activation and Th17 cell differentiation. Int Immunopharmacol 2022; 108:108894. [DOI: 10.1016/j.intimp.2022.108894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/18/2022] [Accepted: 05/23/2022] [Indexed: 11/25/2022]
|
10
|
Renal-Protective Effects and Potential Mechanisms of Traditional Chinese Medicine after Ischemia-Reperfusion Injury. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:5579327. [PMID: 33680054 PMCID: PMC7910071 DOI: 10.1155/2021/5579327] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 01/21/2021] [Accepted: 01/30/2021] [Indexed: 02/05/2023]
Abstract
Renal ischemia-reperfusion (I/R) injury mainly causes acute kidney injury (AKI) after renal transplantation, trauma, sepsis, and hypovolemic shock. Patients with renal I/R injury are frequently associated with a poor prognosis. Traditional Chinese medicine (TCM) has been used for the prevention and treatment of various diseases in China and other Asian countries for centuries. Many studies have shown the protective effect of TCM on renal I/R injury, due to its diverse bioactive components. The potential mechanisms of TCMs on renal I/R injury include anti-inflammation, antioxidative effect, anti-cell death, downregulation of adhesion molecule expression, regulation of energy metabolism by restoring Na+-K+-ATPase activity, and mitochondrial fission. This review summarizes the major developments in the effects and underlying mechanisms of TCMs on the renal I/R injury.
Collapse
|
11
|
Li H, Gao C, Liang Q, Liu C, Liu L, Zhuang J, Yang J, Zhou C, Feng F, Sun C. Cryptotanshinone Is a Intervention for ER-Positive Breast Cancer: An Integrated Approach to the Study of Natural Product Intervention Mechanisms. Front Pharmacol 2021; 11:592109. [PMID: 33505309 PMCID: PMC7832090 DOI: 10.3389/fphar.2020.592109] [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: 09/02/2020] [Accepted: 11/30/2020] [Indexed: 12/20/2022] Open
Abstract
Background: Resistance to endocrine therapy has hampered clinical treatment in patients with ER-positive breast cancer (BRCA). Studies have confirmed that cryptotanshinone (CPT) has cytotoxic effects on BRCA cells and can significantly inhibit the proliferation and metastasis of ER-positive cancer cells. Methods: We analyzed the gene high-throughput data of ER-positive and negative BRCA to screen out key gene targets for ER-positive BRCA. Finally, the effects of CPT on BRCA cells (MCF-7 and MDA-MB-231) were examined, and quantitative RT-PCR was used to evaluate the expression of the key targets during CPT intervention. Results: A total of 169 differentially expressed genes were identified, and revealed that CPT affects the ER-positive BRCA cells by regulating CDK1, CCNA2, and ESR1. The overall experimental results initially show that MCF-7 cells were more sensitive to CPT than MDA-MB-231 cells, and the expression of ESR1 was not affected in the BRCA cells during CPT intervention, while the expression of CDK1 and CCNA2 were significantly down-regulated. Conclusion: CPT can inhibit the proliferation and migration of BRCA cells by regulating CDK1, CCNA2, and ESR1, especially in ER-positive BRCA samples. On the one hand, our research has discovered the possible mechanism that CPT can better interfere with ER+ BRCA; on the other hand, the combination of high-throughput data analysis and network pharmacology provides valuable information for identifying the mechanism of drug intervention in the disease.
Collapse
Affiliation(s)
- Huayao Li
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chundi Gao
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qing Liang
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, China
| | - Cun Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lijuan Liu
- Departmen of Oncology, Weifang Traditional Chinese Hospital, Weifang, China.,Department of Oncology, Affilited Hospital of Weifang Medical University, Weifang, China
| | - Jing Zhuang
- Departmen of Oncology, Weifang Traditional Chinese Hospital, Weifang, China.,Department of Oncology, Affilited Hospital of Weifang Medical University, Weifang, China
| | - Jing Yang
- Departmen of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| | - Chao Zhou
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.,Departmen of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| | - Fubin Feng
- Departmen of Oncology, Weifang Traditional Chinese Hospital, Weifang, China.,Department of Basic Medical Science, Qingdao University, Qingdao, China
| | - Changgang Sun
- Departmen of Oncology, Weifang Traditional Chinese Hospital, Weifang, China.,Chinese Medicine Innovation Institute, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
12
|
Wu YH, Wu YR, Li B, Yan ZY. Cryptotanshinone: A review of its pharmacology activities and molecular mechanisms. Fitoterapia 2020; 145:104633. [DOI: 10.1016/j.fitote.2020.104633] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 05/03/2020] [Accepted: 05/15/2020] [Indexed: 02/07/2023]
|
13
|
Liu Y, Wang H, Gao J, Wen Z, Peng L. Cryptotanshinone ameliorates the pathogenicity of Streptococcus suis by targeting suilysin and inflammation. J Appl Microbiol 2020; 130:736-744. [PMID: 32750224 DOI: 10.1111/jam.14810] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/22/2020] [Accepted: 07/29/2020] [Indexed: 12/22/2022]
Abstract
AIMS Streptococcus suis is a highly zoonotic pathogen that is a serious threat to human health and the development of the pig industry worldwide. The virulence factors produced during S. suis infection play an important role, and the pore-forming activity of suilysin is considered an important virulence-related factor, especially in meningitis. Treatment of S. suis infection with traditional antibiotics is becoming increasingly challenging due to bacterial resistance. The purpose of this study is to verify the role of cryptotanshinone in the process of S. suis infection and provide a new drug precursor for the treatment of S. suis infection. METHODS AND RESULTS In this study, we used circular dichroism spectroscopy to demonstrate that cryptotanshinone alters the secondary structure of suilysin. The results of the antibacterial activity and haemolysis assays showed cryptotanshinone could inhibit the pore-forming activity of suilysin without affecting bacterial growth or its expression. We also showed that cryptotanshinone reduces bacterial damage and penetration in vitro, reduce the S. suis-induced inflammatory response and provide protection against bacterial infections in vivo and in vitro. CONCLUSIONS Cryptotanshinone is a potential compound precursor for treating S. suis infection. SIGNIFICANCE AND IMPACT OF THE STUDY Cryptotanshinone may be a promising leading compound for S. suis infection and related diseases.
Collapse
Affiliation(s)
- Y Liu
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - H Wang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - J Gao
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - Z Wen
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - L Peng
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
14
|
Luo Y, Song L, Wang X, Huang Y, Liu Y, Wang Q, Hong M, Yuan Z. Uncovering the Mechanisms of Cryptotanshinone as a Therapeutic Agent Against Hepatocellular Carcinoma. Front Pharmacol 2020; 11:1264. [PMID: 32903546 PMCID: PMC7438559 DOI: 10.3389/fphar.2020.01264] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 07/30/2020] [Indexed: 12/20/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a fatal and dominant form of liver cancer that currently has no effective treatment or positive prognosis. In this study, we explored the antitumor effects of cryptotanshinone (CPT) against HCC and the molecular mechanisms underlying these effects using a systems pharmacology and experimental validation approach. First, we identified a total of 296 CPT targets, 239 of which were also HCC-related targets. We elucidated the mechanisms by which CPT affects HCC through multiple network analysis, including CPT-target network analysis, protein-protein interaction network analysis, target-function network analysis, and pathway enrichment analysis. In addition, we found that CPT induced apoptosis in Huh7 and MHCC97-H ells due to increased levels of cleaved PARP, Bax, and cleaved caspase-3 and decreased Bcl-2 expression. CPT also induced autophagy in HCC cells by increasing LC3-II conversion and the expression of Beclin1 and ATG5, while decreasing the expression of p62/SQSTM1. Autophagy inhibitors (3-methyladenine and chloroquine) enhanced CPT-induced proliferation and apoptosis, suggesting that CPT-induced autophagy may protect HCC cells against cell death. Furthermore, CPT was found to inhibit the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) signaling pathway. Interestingly, activation of PI3K by insulin-like growth factor-I inhibited CPT-induced apoptosis and autophagy, suggesting that the PI3K/AKT/mTOR signaling pathway is involved in both CPT-induced apoptosis and autophagy. Finally, CPT was found to inhibit the growth of Huh7 xenograft tumors. In conclusion, we first demonstrated the antitumor effects of CPT in Huh7 and MHCC97-H cells, both in vitro and in vivo. We elucidated the potential antitumor mechanism of CPT, which involved inducing apoptosis and autophagy by inhibiting the PI3K/Akt/mTOR signaling pathway. Our findings may provide valuable insights into the clinical application of CPT, serving as a potential candidate therapeutic agent for HCC treatment.
Collapse
Affiliation(s)
- Yi Luo
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lei Song
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinyu Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yujie Huang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yongqiang Liu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ming Hong
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhongyu Yuan
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
15
|
Ashrafizadeh M, Zarrabi A, Orouei S, Saberifar S, Salami S, Hushmandi K, Najafi M. Recent advances and future directions in anti-tumor activity of cryptotanshinone: A mechanistic review. Phytother Res 2020; 35:155-179. [PMID: 33507609 DOI: 10.1002/ptr.6815] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/29/2020] [Accepted: 07/02/2020] [Indexed: 12/13/2022]
Abstract
In respect to the enhanced incidence rate of cancer worldwide, studies have focused on cancer therapy using novel strategies. Chemotherapy is a common strategy in cancer therapy, but its adverse effects and chemoresistance have limited its efficacy. So, attempts have been directed towards minimally invasive cancer therapy using plant derived-natural compounds. Cryptotanshinone (CT) is a component of salvia miltiorrihiza Bunge, well-known as Danshen and has a variety of therapeutic and biological activities such as antioxidant, anti-inflammatory, anti-diabetic and neuroprotective. Recently, studies have focused on anti-tumor activity of CT against different cancers. Notably, this herbal compound is efficient in cancer therapy by targeting various molecular signaling pathways. In the present review, we mechanistically describe the anti-tumor activity of CT with an emphasis on molecular signaling pathways. Then, we evaluate the potential of CT in cancer immunotherapy and enhancing the efficacy of chemotherapy by sensitizing cancer cells into anti-tumor activity of chemotherapeutic agents, and elevating accumulation of anti-tumor drugs in cancer cells. Finally, we mention strategies to enhance the anti-tumor activity of CT, for instance, using nanoparticles to provide targeted drug delivery.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey.,Center of Excellence for Functional Surfaces and Interfaces (EFSUN), Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul, Turkey
| | - Sima Orouei
- MSc. Student, Department of Genetics, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sedigheh Saberifar
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Saeed Salami
- DVM. Graduated, Kazerun Branch, Islamic Azad University, Kazeroon, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|