1
|
Wang M, Shi Z, Wang F, Wang C, Wang H. Integrating structure-activity relationships, computational approaches, and experimental validation to unlock the therapeutic potential of indole-3-carbinol and its derivatives. Biochem Pharmacol 2025; 238:116968. [PMID: 40318812 DOI: 10.1016/j.bcp.2025.116968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/09/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
Indole-3-carbinol (I3C) a bioactive compound derived from cruciferous vegetables, has garnered significant attention for its role in cancer prevention and its broad-spectrum biological activities, including anti-inflammatory properties and the modulation of critical signaling pathways. This review explores the structure-activity relationship (SAR) of I3C and its derivatives, emphasizing their molecular mechanisms and therapeutic potential. Key cellular targets, such as estrogen receptors, and pathways, including NF-κB, Wnt/β-catenin, and PI3K/Akt, are highlighted for their roles in apoptosis, autophagy, and the disruption of mitogenic signaling. The SAR analysis reveals the influence of molecular modifications, particularly in dimeric forms like diindolylmethane (DIM) on pharmacokinetics and bioactivity. Computational approaches, including molecular docking, molecular dynamics simulations, and density functional theory (DFT) provide insights into ligand-receptor interactions, binding energetics, and electronic properties, facilitating biological activity predictions. Experimental evidence from in vitro assays and synergistic studies underscores the cytotoxic efficacy and combinatorial benefits of I3C with conventional chemotherapeutics. Challenges in clinical translation, such as bioavailability and targeted delivery, are addressed, highlighting the potential of integrating computational and experimental findings to develop novel I3C-based therapeutics. This comprehensive analysis positions I3C as a promising scaffold for designing effective agents against cancer and other diseases.
Collapse
Affiliation(s)
- Mingjie Wang
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Anhui Innovative Center for Drug Basic Research of Metabolic Diseases, School of Pharmacy, Wannan Medical College, Wuhu, PR China.
| | - Zihan Shi
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Anhui Innovative Center for Drug Basic Research of Metabolic Diseases, School of Pharmacy, Wannan Medical College, Wuhu, PR China.
| | - Fangfang Wang
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Anhui Innovative Center for Drug Basic Research of Metabolic Diseases, School of Pharmacy, Wannan Medical College, Wuhu, PR China
| | - Cunqin Wang
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Anhui Innovative Center for Drug Basic Research of Metabolic Diseases, School of Pharmacy, Wannan Medical College, Wuhu, PR China.
| | - Hongting Wang
- Anhui Provincial Engineering Research Center for Polysaccharide Drugs, Anhui Innovative Center for Drug Basic Research of Metabolic Diseases, School of Pharmacy, Wannan Medical College, Wuhu, PR China.
| |
Collapse
|
2
|
Lin J, Zhou J, Ye K, Xie F. Prunella vulgaris: A potential molecule for the treatment of hepatocellular carcinoma. Medicine (Baltimore) 2025; 104:e42267. [PMID: 40295240 PMCID: PMC12040002 DOI: 10.1097/md.0000000000042267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 03/03/2025] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Prunella vulgaris (PV) is widely used in treating various diseases, but its relationship with hepatocellular carcinoma (HCC) remains unclear. This study systematically evaluates PV's therapeutic potential in HCC and explores its molecular mechanisms. Active compounds and molecular targets of PV were obtained from the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform database, and HCC-related targets were identified using the Gene Expression Omnibus database. A drug-disease target network was built to identify key hub genes, which were further investigated through immune analysis, single-cell RNA sequencing, molecular docking, and in vitro experiments. We identified 185 drug targets and 635 HCC-related targets, with 15 potential PV targets linked to HCC progression. In vitro validation confirmed significant expression of these targets in HCC cells. Mechanistic analysis indicated that these hub genes may influence HCC progression through pathways like tumor protein 53 signaling and are associated with immune cell subsets, including CD8+ T cells and natural killer cells. This study identifies key bioactive components of PV for HCC treatment and reveals their molecular mechanisms. Dysregulation of these targets correlates with HCC pathogenesis, suggesting their potential as novel biomarkers. Future research will focus on further validation in vitro and in vivo to explore the clinical applicability of these targets and the synergistic potential of PV in combination with other treatments.
Collapse
Affiliation(s)
- Jie Lin
- Department of Hepatobiliary and Pancreatic Surgery, The Neijiang First People’s Hospital, Neijiang, Sichuan, China
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Jiale Zhou
- Department of Hepatobiliary and Pancreatic Surgery, Chengdu Medical College, Chengdu, Sichuan, China
| | - Kailin Ye
- Department of Hepatobiliary and Pancreatic Surgery, Chengdu Medical College, Chengdu, Sichuan, China
| | - Fei Xie
- Department of Hepatobiliary and Pancreatic Surgery, The Neijiang First People’s Hospital, Neijiang, Sichuan, China
| |
Collapse
|
3
|
Yang S, Zhang Y, Zheng C. β-Sitosterol Mitigates Apoptosis, Oxidative Stress and Inflammatory Response by Inactivating TLR4/NF-кB Pathway in Cell Models of Diabetic Nephropathy. Cell Biochem Biophys 2025; 83:1249-1262. [PMID: 39424766 DOI: 10.1007/s12013-024-01559-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2024] [Indexed: 10/21/2024]
Abstract
Podocyte injury plays a pivotal role in the pathogenesis of diabetic nephropathy (DN), leading to proteinuria formation. β-Sitosterol is a natural compound with anti-inflammatory, anti-diabetic, nephroprotective and antioxidant properties. The studyaimed to explore whether and how β-Sitosterol protected podocytes against high glucose (HG)-induced inflammatory andoxidative injury. DN cell models were established by stimulating podocytes or renal tubular epithelial cells (HK-2) cells with 25 mM glucose. Cell viability and apoptosis were evaluated using cell counting kit-8 assays and flow cytometry analyses. Westernblotting was used to quantify protein levels of genes related to podocyte injury, HK-2 cell damage, inflammation, and TLR4/NF-кB pathway. Contents of oxidative stress biomarkers were evaluated by corresponding commercial kits while proinflammatorycytokine levels were determined by enzyme-linked immunosorbent assay. Immunofluorescence staining was performed todetect intracellular levels of reactive oxygen species (ROS) and Nrf2 nuclear translocation. Experimental results revealed that HG treatment induced podocyte dysfunction by impairing cell viability while accelerating theapoptosis, and the changes were reversed by β-sitosterol treatment. Moreover, β-sitosterol repressed HG-evoked oxidative stressby reducing ROS and malondialdehyde (MDA) levels while increasing activities of antioxidant enzymes. The reduction ofproinflammatory cytokines mediated by β-sitosterol in HG-stimulated podocytes suggested the anti-inflammatory role of β-sitosterol. Additionally, the activation of the TLR4/NF-кB signaling induced by HG was inhibited by β-sitosterol in podocytes.Inactivation of the TLR4 using TAK-242 enhanced the protective effects of β-sitosterol against HG-mediated oxidative stressand inflammation. Similarly, β-sitosterol also protected HK-2 cells from HG-induced oxidative stress, inflammation, andapoptosis. In summary, β-sitosterol exerts anti-inflammatory, anti-oxidative, and anti-apoptotic activities in HG-induced podocytes or HK-2 cells by inhibiting TLR4/NF-кB signaling.
Collapse
Affiliation(s)
| | | | - Chenghong Zheng
- Hubei University of Chinese Medicine, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, China.
| |
Collapse
|
4
|
Liang Y, Xie Y, Dang Z, Li M, Yu L, Wang X, Wang P, Yang Z. Yiqi Liangxue Jiedu Prescription Inhibited the Canonical Wnt Pathway to Prevent Hepatocellular Precancerous Lesions. J Hepatocell Carcinoma 2024; 11:2293-2308. [PMID: 39582813 PMCID: PMC11585997 DOI: 10.2147/jhc.s485257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 11/12/2024] [Indexed: 11/26/2024] Open
Abstract
Purpose Yiqi Liangxue Jiedu prescription (YLJP), a Chinese medicine that is commonly used to prevent liver cancer and is authorized by a national patent (patent No. ZL202110889980.5) has a therapeutic effect on precancerous lesions; however, the underlying mechanism remains unclear. This study is aimed at determining the clinical therapeutic efficacy of YLJP in patients with precancerous liver lesions and to explore and validate its possible effector mechanism. Patients and Methods The 1-year incidence of hepatocellular carcinoma (HCC) was retrospectively analyzed in 241 patients with cirrhosis complicated by abnormal alpha-fetoprotein precancer. Network pharmacological analysis, molecular docking, and molecular dynamics simulation were used to explore the key targets and compounds of YLJP in treating HCC. Immunohistochemical methods were used to detect the expression of key proteins in tumor and cirrhotic tissues. Finally, the mechanism underlying the effects of YLJP was verified in rats with precancerous lesions. Results The 1-year incidence of HCC was lower in the YLJP group than in the Western medicine group. The Wnt pathway protein, CTNNB1, is a key target of YLJP in preventing and treating HCC, and the canonical Wnt pathway is the key signaling pathway and is overexpressed in human liver tumors. In vivo experiments showed that YLJP significantly inhibited the canonical Wnt pathway and reduced the abnormal differentiation of hepatic oval cells. The binding of CTNNB1 to oleanolic acid, stigmasterol, and beta-sitosterol was found to be stable, indicating the action of these compounds in treating HCC. Conclusion YLJP reduces the 1-year incidence of HCC, with its mechanism likely due to oleanolic acid, beta-sitosterol, and stigmasterol inhibition of the CTNNB1 activation of the β-catenin protein, which in turn regulates the Wnt signaling pathway and prevents the abnormal differentiation of hepatic oval cells into cancer cells, thus delaying the occurrence and progression of the disease.
Collapse
Affiliation(s)
- Yuling Liang
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Yuqing Xie
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Zhibo Dang
- Ethics Committee Office, Henan Province Hospital of TCM, Henan, People’s Republic of China
| | - Mengge Li
- Department of Hepatobiliary Spleen and Stomach, Henan Province Hospital of TCM, Henan, People’s Republic of China
| | - Lihua Yu
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Xinhui Wang
- Beijing Children’s Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Peng Wang
- Department of Pathology, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Zhiyun Yang
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
5
|
Ye G, Ye M, Jin X. Roles of clinical application of lenvatinib and its resistance mechanism in advanced hepatocellular carcinoma (Review). Am J Cancer Res 2024; 14:4113-4171. [PMID: 39417171 PMCID: PMC11477829 DOI: 10.62347/ujvp4361] [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: 05/09/2024] [Accepted: 09/03/2024] [Indexed: 10/19/2024] Open
Abstract
Lenvatinib (LEN) is a multi-target TKI, which plays a pivotal role in the treatment of advanced hepatocellular carcinoma (HCC). The inevitable occurrence of drug resistance still prevents curative potential and is deleterious for the prognosis, and a growing body of studies is accumulating, which have devoted themselves to unveiling its underlying resistance mechanism and made some progress. The dysregulation of crucial signaling pathways, non-coding RNA and RNA modifications were proven to be associated with LEN resistance. A range of drugs were found to influence LEN therapeutic efficacy. In addition, the superiority of LEN combination therapy has been shown to potentially overcome the limitations of LEN monotherapy in a series of research, and a range of promising indicators for predicting treatment response and prognosis have been discovered in recent years. In this review, we summarize the latest developments in LEN resistance, the efficacy and safety of LEN combination therapy as well as associated indicators, which may provide new insight into its resistance as well as ideas in the treatment of advanced HCC.
Collapse
Affiliation(s)
- Ganghui Ye
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo UniversityNingbo 315211, Zhejiang, P. R. China
- Department of Oncology, The First Hospital of Ningbo UniversityNingbo 315020, Zhejiang, P. R. China
- Department of Radiation Oncology, Taizhou Central Hospital (Taizhou University Hospital)Taizhou 318000, Zhejiang, P. R. China
| | - Meng Ye
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo UniversityNingbo 315211, Zhejiang, P. R. China
- Department of Oncology, The First Hospital of Ningbo UniversityNingbo 315020, Zhejiang, P. R. China
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo UniversityNingbo 315211, Zhejiang, P. R. China
- Department of Oncology, The First Hospital of Ningbo UniversityNingbo 315020, Zhejiang, P. R. China
| |
Collapse
|
6
|
Liu B, Liu L, Liu Y. Targeting cell death mechanisms: the potential of autophagy and ferroptosis in hepatocellular carcinoma therapy. Front Immunol 2024; 15:1450487. [PMID: 39315094 PMCID: PMC11416969 DOI: 10.3389/fimmu.2024.1450487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/21/2024] [Indexed: 09/25/2024] Open
Abstract
Ferroptosis is a type of cell death that plays a remarkable role in the growth and advancement of malignancies including hepatocellular carcinoma (HCC). Non-coding RNAs (ncRNAs) have a considerable impact on HCC by functioning as either oncogenes or suppressors. Recent research has demonstrated that non-coding RNAs (ncRNAs) have the ability to control ferroptosis in HCC cells, hence impacting the advancement of tumors and the resistance of these cells to drugs. Autophagy is a mechanism that is conserved throughout evolution and plays a role in maintaining balance in the body under normal settings. Nevertheless, the occurrence of dysregulation of autophagy is evident in the progression of various human disorders, specifically cancer. Autophagy plays dual roles in cancer, potentially influencing both cell survival and cell death. HCC is a prevalent kind of liver cancer, and genetic mutations and changes in molecular pathways might worsen its advancement. The role of autophagy in HCC is a subject of debate, as it has the capacity to both repress and promote tumor growth. Autophagy activation can impact apoptosis, control proliferation and glucose metabolism, and facilitate tumor spread through EMT. Inhibiting autophagy can hinder the growth and spread of HCC and enhance the ability of tumor cells to respond to treatment. Autophagy in HCC is regulated by several signaling pathways, such as STAT3, Wnt, miRNAs, lncRNAs, and circRNAs. Utilizing anticancer drugs to target autophagy may have advantageous implications for the efficacy of cancer treatment.
Collapse
Affiliation(s)
- Beibei Liu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ling Liu
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yang Liu
- Day Surgery Center, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
7
|
Liang B, Xiao D, Wang SH, Xu X. Novel thiosemicarbazide-based β-carboline derivatives as α-glucosidase inhibitors: Synthesis and biological evaluation. Eur J Med Chem 2024; 275:116595. [PMID: 38875808 DOI: 10.1016/j.ejmech.2024.116595] [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: 04/19/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 06/16/2024]
Abstract
In the quest for potent α-glucosidase inhibitors to combat diabetes, a series of novel thiosemicarbazide-based β-carboline derivatives (CTL1∼36) were synthesized and evaluated. CTL1∼36 exhibited remarkable inhibitory effects against α-glucosidase, with IC50 values ranging from 2.81 to 12.40 μM, significantly surpassing the positive control acarbose (IC50 = 564.28 μM). Notably, CTL26 demonstrated the most potent inhibition (IC50 = 2.81 μM) and was characterized as a non-competitive inhibitor. Through a combination assay with fluorescence quenching, 3D fluorescence spectra, CD spectra, and molecular docking, we elucidated that CTL26 formed a complex with α-glucosidase via hydrogen bondings and hydrophobic interactions, leading to α-glucosidase conformation changes that impaired enzymatic activity. In vivo studies revealed that oral administration of CTL26 (25 and 50 mg/kg/d) reduced fasting blood glucose levels, enhanced glucose tolerance, and ameliorated lipid abnormalities in diabetic mice. These findings positioned CTL26 as a promising candidate for the development of α-glucosidase inhibitors with anti-diabetic potential.
Collapse
Affiliation(s)
- Bingwen Liang
- School of Pharmacy and Food Engineering & Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, 529020, China
| | - Di Xiao
- School of Pharmacy and Food Engineering & Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, 529020, China
| | - Shao-Hua Wang
- School of Pharmacy & State Key Laboratory of Applied Organic Chemistry & Collaborative Innovation Center for Northwestern Chinese Medicine, Lanzhou University, Lanzhou, 730000, China.
| | - Xuetao Xu
- School of Pharmacy and Food Engineering & Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, Wuyi University, Jiangmen, 529020, China.
| |
Collapse
|
8
|
Duan H, Li H, Liu T, Chen Y, Luo M, Shi Y, Zhou J, Rashed MMA, Zhai K, Li L, Wei Z. Exploring the Molecular Mechanism of Schisandrin C for the Treatment of Atherosclerosis via the PI3K/AKT/mTOR Autophagy Pathway. ACS OMEGA 2024; 9:32920-32930. [PMID: 39100354 PMCID: PMC11292807 DOI: 10.1021/acsomega.4c03738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/16/2024] [Accepted: 07/02/2024] [Indexed: 08/06/2024]
Abstract
Atherosclerosis (AS) is a common cardiovascular disease that poses a major threat to health. Schisandra chinensis is a medicinal and edible plant that is commonly used to treat cardiovascular diseases. In this paper, HPLC was used to detect and analyze 5 different components in Schisandra chinensis. Network pharmacological predictions highlight the PI3K/AKT/mTOR pathway as an important pharmacological pathway. The effective ingredient Schisandrin C was screened by the molecular docking technique. ox-LDL-induced HUVECs were used to construct the atherosclerosis model for further experimental verification. The results showed that Schisandrin C interfered with the PI3K/AKT/mTOR autophagy pathway. This study lays a foundation for the further application of Schisandrin C in the prevention and treatment of atherosclerosis in the future.
Collapse
Affiliation(s)
- Hong Duan
- School
of Biological and Food Engineering, Engineering Research Center for
Development and High Value Utilization of Genuine Medicinal Materials
in North Anhui Province, Suzhou University, Suzhou, Anhui 234000, China
- College
of Biological and Food Engineering, Anhui
Polytechnic University, Wuhu, Anhui 241000, China
| | - Han Li
- School
of Biological and Food Engineering, Engineering Research Center for
Development and High Value Utilization of Genuine Medicinal Materials
in North Anhui Province, Suzhou University, Suzhou, Anhui 234000, China
- College
of Biological and Food Engineering, Anhui
Polytechnic University, Wuhu, Anhui 241000, China
| | - Tianyi Liu
- School
of Biological and Food Engineering, Engineering Research Center for
Development and High Value Utilization of Genuine Medicinal Materials
in North Anhui Province, Suzhou University, Suzhou, Anhui 234000, China
| | - Yuan Chen
- School
of Biological and Food Engineering, Engineering Research Center for
Development and High Value Utilization of Genuine Medicinal Materials
in North Anhui Province, Suzhou University, Suzhou, Anhui 234000, China
- College
of Biological and Food Engineering, Anhui
Polytechnic University, Wuhu, Anhui 241000, China
| | - Mengmeng Luo
- School
of Biological and Food Engineering, Engineering Research Center for
Development and High Value Utilization of Genuine Medicinal Materials
in North Anhui Province, Suzhou University, Suzhou, Anhui 234000, China
- College
of Biological and Food Engineering, Anhui
Polytechnic University, Wuhu, Anhui 241000, China
| | - Ying Shi
- School
of Biological and Food Engineering, Engineering Research Center for
Development and High Value Utilization of Genuine Medicinal Materials
in North Anhui Province, Suzhou University, Suzhou, Anhui 234000, China
- College
of Biological and Food Engineering, Anhui
Polytechnic University, Wuhu, Anhui 241000, China
| | - Jing Zhou
- School
of Biological and Food Engineering, Engineering Research Center for
Development and High Value Utilization of Genuine Medicinal Materials
in North Anhui Province, Suzhou University, Suzhou, Anhui 234000, China
- College
of Biological and Food Engineering, Anhui
Polytechnic University, Wuhu, Anhui 241000, China
| | - Marwan M. A. Rashed
- School
of Biological and Food Engineering, Engineering Research Center for
Development and High Value Utilization of Genuine Medicinal Materials
in North Anhui Province, Suzhou University, Suzhou, Anhui 234000, China
| | - Kefeng Zhai
- School
of Biological and Food Engineering, Engineering Research Center for
Development and High Value Utilization of Genuine Medicinal Materials
in North Anhui Province, Suzhou University, Suzhou, Anhui 234000, China
- College
of Biological and Food Engineering, Anhui
Polytechnic University, Wuhu, Anhui 241000, China
| | - Lili Li
- General
Clinical Research Center, Anhui Wanbei Coal-Electricity
Group General Hospital, Suzhou 234000, China
| | - Zhaojun Wei
- School
of Biological Science and Engineering, North
Minzu University, Yinchuan 750021, China
| |
Collapse
|
9
|
Zhu H, Zhong X. Honokiol as an α-glucosidase inhibitor. Front Pharmacol 2024; 15:1425832. [PMID: 38962316 PMCID: PMC11220239 DOI: 10.3389/fphar.2024.1425832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/04/2024] [Indexed: 07/05/2024] Open
Abstract
Honokiol, a naturally occurring compound from Magnolia obovata Thunb., has many biological activities, but its anti-α-glucosidase activity is still unclear. Therefore, we determined its inhibitory effects against α-glucosidase. Activity assays showed that honokiol was a reversible mixed-type inhibitor of α-glucosidase, and its IC50 value was 317.11 ± 12.86 μM. Fluorescence results indicated that the binding of honokiol to α-glucosidase caused a reduction in α-glucosidase activity. 3D fluorescence and CD spectra results indicated that the binding of honokiol to α-glucosidase caused conformational change in α-glucosidase. Docking simulated the detailed interactions between honokiol and α-glucosidase, including hydrogen and hydrophobic bonds. All findings showed that honokiol could be used as a natural inhibitor to develop α-glucosidase agents.
Collapse
Affiliation(s)
- Hua Zhu
- School of Chemistry and Chemical Engineering, Mianyang Teacher’s College, Mianyang, China
| | - Xin Zhong
- Dean’s Office, Mianyang Teacher’s College, Mianyang, China
| |
Collapse
|
10
|
Liang F. Inhibition mechanism investigation of quercetagetin as a potential tyrosinase inhibitor. Front Chem 2024; 12:1411801. [PMID: 38894729 PMCID: PMC11184945 DOI: 10.3389/fchem.2024.1411801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 05/16/2024] [Indexed: 06/21/2024] Open
Abstract
Tyrosinase is one important rate limiting enzyme in melanin synthesis, directly affecting the melanin synthesis. Quercetagetin is one active ingredient from marigold. Thence, the inhibition effects of quercetagetin against tyrosinase were investigated. The results showed quercetagetin could inhibit tyrosinase activity with IC50 value of 0.19 ± 0.01 mM and the inhibition type was a reversible mixed-type. Results of fluorescence quenching showed quercetagetin could quench tyrosinase fluorescence in static process. CD and 3D fluorescence results showed the interaction of quercetagetin to tyrosinase could change tyrosinase conformation to inhibit activity. Moreover, docking revealed details of quercetagetin's interactions with tyrosinase.
Collapse
Affiliation(s)
- Faliang Liang
- Pharmacy Department, Jiang Men Maternity and Child Healthcare Hospital, Jiangmen, China
| |
Collapse
|
11
|
Wu D, Zuo Z, Sun X, Li X, Yin F, Yin W. ACSL4 promotes malignant progression of Hepatocellular carcinoma by targeting PAK2 transcription. Biochem Pharmacol 2024; 224:116206. [PMID: 38615921 DOI: 10.1016/j.bcp.2024.116206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 03/10/2024] [Accepted: 04/11/2024] [Indexed: 04/16/2024]
Abstract
Long-chain fatty acyl-Coa ligase 4 (ACSL4) is an important enzyme that converts fatty acids to fatty acyl-Coa esters, there is increasing evidence for its role in carcinogenesis. However, the precise role of ACLS4 in hepatocellular carcinoma (HCC) is not clearly understood. In the present study, we provide evidence that ACSL4 expression was specifically elevated in HCC and is associated with poor clinical outcomes. ACSL4 significantly promotes the growth and metastasis of HCC both in vitro and in vivo. RNA sequencing and functional experiments showed that the effect of ACSL4 on HCC development was heavily dependent on PAK2. ACSL4 expression is well correlated with PAK2 in HCC, and ACSL4 even transcriptionally increased PAK2 gene expression mediated by Sp1. In addition, emodin, a naturally occurring anthraquinone derivative, inhibited HCC cell growth and tumor progression by targeting ACSL4. In summary, ACSL4 plays a novel oncogene in HCC development by regulating PAK2 transcription. Targeting ACSL4 could be useful in drug development and therapy for HCC.
Collapse
Affiliation(s)
- Dandan Wu
- College of Life Sciences in Nanjing University (Xianlin Campus), State Key lab of Pharmaceutical Biotechnology (SKLPB), Nanjing University, Nanjing 210046, China
| | - Zongchao Zuo
- Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
| | - Xinning Sun
- College of Life Sciences in Nanjing University (Xianlin Campus), State Key lab of Pharmaceutical Biotechnology (SKLPB), Nanjing University, Nanjing 210046, China
| | - Xin Li
- College of Life Sciences in Nanjing University (Xianlin Campus), State Key lab of Pharmaceutical Biotechnology (SKLPB), Nanjing University, Nanjing 210046, China
| | - Fangzhou Yin
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Wu Yin
- College of Life Sciences in Nanjing University (Xianlin Campus), State Key lab of Pharmaceutical Biotechnology (SKLPB), Nanjing University, Nanjing 210046, China.
| |
Collapse
|