1
|
Luo L, Ge M, Sun Y, Hu Y, Yang X, Zhang R. New findings on the antagonism of the environmental chemical toxicity 2-ethylhexyl diphenyl phosphate: Glycyrrhizic acid as an Nrf2 activator targets Nrf2/ROS/STAT3 signalling crosstalk to alleviate thymic injury in chicks. Poult Sci 2025; 104:104918. [PMID: 40024011 PMCID: PMC11919435 DOI: 10.1016/j.psj.2025.104918] [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: 12/16/2024] [Revised: 01/25/2025] [Accepted: 02/16/2025] [Indexed: 03/04/2025] Open
Abstract
2-Ethylhexyl diphenyl phosphate (EHDPHP) is a pervasive environmental pollutant known to induce oxidative damage in organisms; however, its precise mechanisms of toxicity remain unclear. Furthermore, limited research has been conducted on potential therapeutic agents to counteract EHDPHP toxicity. Glycyrrhizic acid (GA), a triterpenoid saponin compound with recognized antioxidant, anti-inflammatory, and immunomodulatory properties, represents a promising candidate for mitigating EHDPHP-induced oxidative injury. In chickens, the thymus is the main immune organ. This study aims to investigate the mechanism of EHDPHP-induced thymus damage and the role and mechanism of GA intervention in this process. A potential 'EHDPHP/GA-Target-Oxidative Stress (OS)' network was constructed using network biology. A model of EHDPHP-induced chicken thymic injury was established by continuous oral administration of EHDPHP (160 mg/kg) for 42 days. Moreover, the mechanism of action by which GA antagonizes EHDPHP-induced oxidative damage was explored using MDCC-MSB1 cells in vitro. Network biology analyses showed that 'EHDPHP/GA-OS' targets were mainly enriched in the adipocytokine and apoptotic signaling pathways. Molecular docking demonstrated the binding interactions of GA and EHDPHP with Kelch-like ECH-associated protein 1 (Keap1), nuclear factor erythroid 2-related factor 2 (Nrf2), and signal transducer and activator of transcription 3 (STAT3) proteins. Both in vitro and in vivo experiments revealed that GA attenuated EHDPHP-induced damage to thymus and MDCC-MSB1 cells, as evidenced by reductions in oxidative stress markers (ROS, MDA, T-AOC, SOD, and GSH-Px), inflammation factors (NF-κB, IL-6, and TNF-α), and the apoptotic factor (Caspase 3) expression. GA treatment increased the expression of Nrf2 and HO-1 while reducing the expression of Keap1, JAK1, Phospho-JAK1 (P-JAK1), STAT3, and Phospho-STAT3 (P-STAT3). Furthermore, the protective effect of GA against EHDPHP-induced MDCC-MSB1 cell injury, as well as its inhibition of the JAK1/STAT3 pathway, was diminished by the Nrf2 inhibitor ML385. These findings suggest that GA exerts its protective effects through Nrf2 and mitigates EHDPHP-induced thymic injury in chickens by modulating the crosstalk between the Nrf2/ROS/STAT3 signaling pathways. Overall, this study highlights the novel role of GA in treating EHDPHP-induced injury and underscores its potential application in diseases treatable with Nrf2 activators.
Collapse
Affiliation(s)
- Linghuan Luo
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Harbin 150030, PR China
| | - Ming Ge
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Harbin 150030, PR China
| | - Yiming Sun
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Harbin 150030, PR China
| | - Yihan Hu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Harbin 150030, PR China
| | - Xiaoyi Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Harbin 150030, PR China
| | - Ruili Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Harbin 150030, PR China.
| |
Collapse
|
2
|
Huang M, Wang W, Cao L, Liu J, Du C, Zhang J. An allosteric inhibitor targeting the STAT3 coiled-coil domain selectively suppresses proliferation of breast cancer. RSC Med Chem 2025:d4md00926f. [PMID: 40190419 PMCID: PMC11969721 DOI: 10.1039/d4md00926f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/14/2025] [Indexed: 04/09/2025] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) remains a challenging and attractive therapeutic target in cancer research. The coiled-coil domain (CCD) of STAT3 represents a novel site for targeted intervention, distinct from the Src-homology 2 domain, and plays a crucial role in regulating the earlier activation and biological function of STAT3 in cell proliferation, survival and invasion of breast cancer cells. We previously reported K116, N'-(1-(2,4-dihydroxyphenyl)ethylidene)thiophene-2-carbohydrazide, as a potent allosteric inhibitor specifically targeting the STAT3 CCD. This study aimed to investigate the antiproliferation effect of K116 on breast cancer cells in vitro and in vivo. The results showed that K116 inhibited the proliferation of breast cancer cell lines in a dose-dependent manner by reducing the phosphorylation of STAT3 Lyr705 and did not inhibit the proliferation of HGC-27 and A549 cells nor their STAT3 Lyr705 phosphorylation. Compared with Stattic (STAT3 SH2 inhibitor), K116 selectively inhibited the proliferation of breast cancer cells. Furthermore, K116 (20 μM) directly monitored STAT3 stabilization and engagement within MDA-MB-468 cells, without affecting STAT1, STAT5, and Akt1. K116 induced apoptosis and inhibited migration as well as pY705STAT3 nuclear translocation and transcriptional activity of STAT3. In addition, K116 (30 mg kg-1) markedly suppressed tumor growth and inhibited STAT3 activity in a 4T1 cell-derived murine breast cancer model. Overall, our results provided pharmacological evidence supporting future clinical investigation of K116 as a promising STAT3 CCD allosteric inhibitor for breast cancer treatment.
Collapse
Affiliation(s)
- Min Huang
- Institute of Medical Research, Northwestern Polytechnical University Xi'an 710072 China
| | - Wei Wang
- Department of Pathology, Qinghai Provincial People's Hospital Xining 810000 China
| | - Liyue Cao
- Institute of Medical Research, Northwestern Polytechnical University Xi'an 710072 China
| | - Jiaxin Liu
- Institute of Medical Research, Northwestern Polytechnical University Xi'an 710072 China
| | - Can Du
- Department of Pathology, Qinghai Provincial People's Hospital Xining 810000 China
| | - Jian Zhang
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine Shanghai 200025 China
| |
Collapse
|
3
|
Bono A, La Monica G, Alamia F, Lauria A, Martorana A. A novel in silico approach for identifying multi-target JAK/STAT inhibitors as anticancer agents. J Mol Graph Model 2025; 135:108913. [PMID: 39615248 DOI: 10.1016/j.jmgm.2024.108913] [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: 06/05/2024] [Revised: 10/11/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025]
Abstract
Apoptosis, or programmed cell death, plays a pivotal role in maintaining cellular homeostasis by eliminating damaged or surplus cells. Dysregulation of signaling pathways, such as JAK/STAT, is implicated in various diseases, rendering them attractive therapeutic targets for potential new anticancer drugs. Concurrently, it is imperative to preserve essential proteins like TNF-α and p53 to maintain normal cellular life/death balance. In light of these considerations, this study employs an innovative in silico hybrid and hierarchical virtual screening approach aimed at identifying JAK/STAT multi-target inhibitors as potential anticancer agents for several tumoral diseases. Initially, the Biotarget Predictor Tool is utilized in a combined ON/OFF-target/Multitarget mode using the extensive National Cancer Institute (NCI) database, previously filtered by ADME evaluation tools. Subsequently, Molecular Docking studies are conducted on JAK2, JAK3, and STAT3, facilitating the identification of the most promising compound, 755435. Finally, Molecular Dynamics Simulations validate the high stability of the potential multitarget inhibitor 755435 in complex with JAK2, JAK3, and STAT3.
Collapse
Affiliation(s)
- Alessia Bono
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche "STEBICEF" - University of Palermo, Viale delle Scienze - Ed. 17, 90128, Palermo, Italy
| | - Gabriele La Monica
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche "STEBICEF" - University of Palermo, Viale delle Scienze - Ed. 17, 90128, Palermo, Italy
| | - Federica Alamia
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche "STEBICEF" - University of Palermo, Viale delle Scienze - Ed. 17, 90128, Palermo, Italy
| | - Antonino Lauria
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche "STEBICEF" - University of Palermo, Viale delle Scienze - Ed. 17, 90128, Palermo, Italy; NBFC, National Biodiversity Future Center, Piazza Marina 61, Palermo, 90133, Italy.
| | - Annamaria Martorana
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche "STEBICEF" - University of Palermo, Viale delle Scienze - Ed. 17, 90128, Palermo, Italy
| |
Collapse
|
4
|
Xia L, Li H, Long L, Ruan W, Ma J, Xu S, Qiao D. Research progress on the pathogenesis of psoriasis and its small molecule inhibitors. Arch Pharm (Weinheim) 2025; 358:e2400621. [PMID: 39686874 DOI: 10.1002/ardp.202400621] [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] [Received: 07/31/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 12/18/2024]
Abstract
Psoriasis is a prevalent chronic systemic immune disease characterized by T-cellmediated hyperproliferation of keratinized cells. Among its various manifestations, plaque-type psoriasis is the most common. Treatment options for psoriasis encompass topical medications, biological therapies, phototherapy techniques, and others. However, traditional treatments are associated with numerous side effects. In contrast, targeted therapy has garnered increasing attention due to its high selectivity, strong safety profile, and favorable therapeutic outcomes. Patients with psoriasis lesions exhibit elevated levels of proinflammatory cytokines compared with the general population. These proinflammatory cytokines have been implicated in mediating psoriasis pathogenesis by inducing keratinocyte proliferation through multiple signaling pathways within the body. This study will delve into the Janus kinase-signal transducers and activators of transcription, phosphatidylinositol 3 kinase (PI3K)-protein kinase B (PKB, also known as AKT), and nuclear factor Kappa-light-chain-enhancer of activated B cells signaling pathways to elucidate their roles in mediating psoriasis pathogenesis. In addition, we will summarize potential targets relevant to the treatment of psoriasis and discuss the design and activity assessment of their inhibitors. It also provides new insights for further in-depth study of psoriasis and development of novel molecularly targeted inhibitors.
Collapse
Affiliation(s)
- Lulu Xia
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| | - Hongxin Li
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| | - Li Long
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| | - Wei Ruan
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| | - Jiajia Ma
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| | - Shan Xu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| | - Dan Qiao
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| |
Collapse
|
5
|
Cai W, Jiang B, Yin Y, Ma L, Li T, Chen J. Identification of STAT3 phosphorylation inhibitors using generative deep learning, virtual screening, molecular dynamics simulations, and biological evaluation for non-small cell lung cancer therapy. Mol Divers 2024:10.1007/s11030-024-11067-5. [PMID: 39715975 DOI: 10.1007/s11030-024-11067-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 11/22/2024] [Indexed: 12/25/2024]
Abstract
The development of phosphorylation-suppressing inhibitors targeting Signal Transducer and Activator of Transcription 3 (STAT3) represents a promising therapeutic strategy for non-small cell lung cancer (NSCLC). In this study, a generative model was developed using transfer learning and virtual screening, leveraging a comprehensive dataset of STAT3 inhibitors to explore the chemical space for novel candidates. This approach yielded a chemically diverse library of compounds, which were prioritized through molecular docking and molecular dynamics (MD) simulations. Among the identified candidates, the HG110 molecule demonstrated potent suppression of STAT3 phosphorylation at Tyr705 and inhibited its nuclear translocation in IL6-stimulated H441 cells. Rigorous MD simulations further confirmed the stability and interaction profiles of top candidates within the STAT3 binding site. Notably, HG106 and HG110 exhibited superior binding affinities and stable conformations, with favorable interactions involving key residues in the STAT3 binding pocket, outperforming known inhibitors. These findings underscore the potential of generative deep learning to expedite the discovery of selective STAT3 inhibitors, providing a compelling pathway for advancing NSCLC therapies.
Collapse
Affiliation(s)
- Weiji Cai
- School of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Road, Yinchuan, 750004, Ningxia, China
- Key Laboratory of Fertility Maintenance Ministry of Education, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Beier Jiang
- Navy Medical Research Institute, Naval Medical University, Shanghai, 200433, China
| | - Yichen Yin
- School of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Road, Yinchuan, 750004, Ningxia, China
- Key Laboratory of Fertility Maintenance Ministry of Education, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Lei Ma
- School of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Road, Yinchuan, 750004, Ningxia, China
- Key Laboratory of Fertility Maintenance Ministry of Education, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Tao Li
- Department of Oncology, General Hospital of the Ningxia Medical University, Yinchuan, 750004, China.
| | - Jing Chen
- School of Basic Medical Sciences, Ningxia Medical University, 1160 Shengli Road, Yinchuan, 750004, Ningxia, China.
- Key Laboratory of Fertility Maintenance Ministry of Education, Ningxia Medical University, Yinchuan, 750004, Ningxia, China.
| |
Collapse
|
6
|
Song Z, Chen H, Wang X, Zhang Z, Li H, Zhao H, Liu Y, Han Q, Zhang J. Napabucasin-loaded PLGA nanoparticles trigger anti-HCC immune responses by metabolic reprogramming of tumor-associated macrophages. J Transl Med 2024; 22:1125. [PMID: 39707412 DOI: 10.1186/s12967-024-05917-x] [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] [Received: 08/13/2024] [Accepted: 11/25/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND JAK/STAT3 is one of the critical signaling pathways involved in the occurrence and development of hepatocellular carcinoma (HCC). BBI608 (Napabucasin), as a novel small molecule inhibitor of STAT3, has shown previously excellent anti-HCC effects in vitro and in mouse models. However, low bioavailability, high cytotoxicity and other shortcomings limit its clinical application. In this study, PLGA was selected to prepare Napabucasin PLGA nanoparticles (NPs) by solvent evaporation method, overcoming these limitations and improving the passive targeting effect that nanoparticle mediated. Base on this, we systematically evaluated the anti-HCC effect of Napabucasin-PLGA NPs and explored the underlying mechanisms. METHODS Napabucasin-PLGA NPs were prepared by solvent evaporation method. CCK-8 assay, Annexin V/PI double staining, RT-qPCR, colony formation assay, and Western blotting were performed to evaluate the anti-HCC effect of Napabucasin-PLGA NPs in vitro. Proliferation assay and migration assay were used to detect the effects of Napabucasin-PLGA NPs-treated HCC-TAMs on tumor biological characteristics of HCC cells. Flow cytometry was used to detect anti-HCC immune responses induced by Napabucasin-PLGA NPs in vivo. RESULTS Our results demonstrated that Napabucasin-PLGA NPs could improve the bioavailability of Napabucasin and enhance Napabucasin-mediated the anti-HCC effects in vitro and in vivo with no significant drug toxicity. In addition to the direct inhibitory effects on the tumor biological characteristics of HCC cells, Napabucasin-PLGA NPs could promote the polarization of macrophages from tumor-promoting M2-type to anti-tumor M1-type, improving the tumor immune microenvironment and augmenting T cell-mediated anti-tumor responses. The underlining mechanisms showed Napabucasin-PLGA NPs suppressed the STAT3/FAO signaling axis in HCC-induced tumor-associated macrophages (TAMs). CONCLUSIONS These findings demonstrated Napabucasin-PLGA NPs is a potential therapeutic candidate for HCC, and provided a new theoretical and experimental basis for further development and clinical application of Napabucasin.
Collapse
Affiliation(s)
- Zhenwei Song
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Hongfei Chen
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Xueyao Wang
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Zhiyue Zhang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Hui Li
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Huajun Zhao
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Yang Liu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Qiuju Han
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Jian Zhang
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, China.
| |
Collapse
|
7
|
Khan K, Zahid M, Ali N, Attaullah S, Ullah M, Khan K, Muhammad I, Abusharha A, Aschner M, Khan H. STAT3 single-nucleotide variants in autoimmune thyroid disease in the Pakhtun population of Khyber Pakhtunkhwa, Pakistan. GENE REPORTS 2024; 36:101950. [PMID: 39385969 PMCID: PMC11463997 DOI: 10.1016/j.genrep.2024.101950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
The current study was conducted to assess the relationship between the STAT3 gene variants rs744166 and rs2293152 and autoimmune thyroid disorder in the Pakhtun population of the province, of Khyber Pakhtunkhwa, Pakistan. Blood was collected from 100 healthy individuals and 400 thyroid-disordered patients. Of these, one hundred were diagnosed with Hashimoto's thyroiditis (HT), while 32 were confirmed as Grave's disease (GD) patients. T3, T4, and TSH serum levels were checked to diagnose thyroid disorders. The blood was analyzed for anti-thyroid peroxidase antibodies (Anti-TPOAb) (AESKULISA- ATPO - elisa kit), (Germany), and thyroid stimulating hormone receptor antibodies (TSHRAb), TSHR Ab elisa kit (Diametra Italy), respectively. PCR was used to amplify the targeted STAT3 gene polymorphisms from rs744166 (301 bp) and rs2293152 (365 bp) sequences and then digested by specific restriction endonucleases (AluI) and AciI respectively. The disease displayed a female predominance. The genotype TC and CC of rs744166 showed a significant relationship with Grave's disease (p = 0.002, OR = 0.28, 95 % CI = 0.11-0.77) in patients. The C allele contributed significantly to the disease in GD patients. The SNP rs2293152 significantly differed between GD patients and control (p = 0.032, OR = 0.29, 95 % CI = 0.09-0.86). Similarly, the G and C alleles showed a significant (p = 0.02) difference between GD patients and the control. No significant association was found for both SNPs in Hashimoto's thyroiditis disease. It is concluded that the STAT3 gene (rs744166 and rs2293152) was found to have a potential role in autoimmunity in GD patients. Still, it needs further studies with larger sample sizes in the Pakhtun population to understand this relationship.
Collapse
Affiliation(s)
- Khayyam Khan
- Department of Zoology, Islamia College University, 25120 Peshawar, Pakistan
| | - Muhammad Zahid
- Department of Zoology, Islamia College University, 25120 Peshawar, Pakistan
| | - Niaz Ali
- Institute of Basic Medical Sciences Khyber Medical University, 25120 Peshawar, Pakistan
| | - Sobia Attaullah
- Department of Zoology, Islamia College University, 25120 Peshawar, Pakistan
| | - Mujeeb Ullah
- Department of Zoology, Islamia College University, 25120 Peshawar, Pakistan
| | - Khalid Khan
- Department of Zoology, Islamia College University, 25120 Peshawar, Pakistan
| | - Ijaz Muhammad
- Department of Zoology, Abdul Wali Khan University Mardan, 23200 Mardan, Pakistan
| | - Ali Abusharha
- Optometry Department, Applied Medical Sciences Collage, King Saud University, P. O. Box 145111, Riyadh, Saudi Arabia
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States of America
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, 23200 Mardan, Pakistan
| |
Collapse
|
8
|
Dong L, Liu C, Sun H, Wang M, Sun M, Zheng J, Yu X, Shi R, Wang B, Zhou Q, Chen Z, Xing B, Wang Y, Yao X, Mei M, Ren Y, Zhou X. Targeting STAT3 potentiates CDK4/6 inhibitors therapy in head and neck squamous cell carcinoma. Cancer Lett 2024; 593:216956. [PMID: 38735381 DOI: 10.1016/j.canlet.2024.216956] [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: 03/27/2024] [Revised: 05/02/2024] [Accepted: 05/09/2024] [Indexed: 05/14/2024]
Abstract
Anti-CDK4/6 therapy has been employed for the treatment for head and neck squamous cell carcinoma (HNSCC) with CDK4/6 hyperactivation, but the response rate is relatively low. In this study, we first showed that CDK4 and CDK6 was over-expressed and conferred poor prognosis in HNSCC. Moreover, in RB-positive HNSCC, STAT3 signaling was activated induced by CDK4/6 inhibition and STAT3 promotes RB deficiency by upregulation of MYC. Thirdly, the combination of Stattic and CDK4/6 inhibitor results in striking anti-tumor effect in vitro and in Cal27 derived animal models. Additionally, phospho-STAT3 level negatively correlates with RB expression and predicts poor prognosis in patients with HNSCC. Taken together, our findings suggest an unrecognized function of STAT3 confers to CDK4/6 inhibitors resistance and presenting a promising combination strategy for patients with HNSCC.
Collapse
Affiliation(s)
- Lin Dong
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, 300060, China; National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, 300060, China
| | - Chao Liu
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, 300060, China; National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, 300060, China
| | - Haoyang Sun
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Mo Wang
- Department of Cell Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Mengyu Sun
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, 300060, China; National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, 300060, China
| | - Jianwei Zheng
- Department of Cell Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Xiaoxue Yu
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Rong Shi
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Bo Wang
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, 300060, China; National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, 300060, China
| | - Qianqian Zhou
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, 300060, China; National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, 300060, China
| | - Zhiqiang Chen
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, 300060, China; National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, 300060, China
| | - Bofan Xing
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, 300060, China; National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, 300060, China
| | - Yu Wang
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, 300060, China; National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, 300060, China
| | - Xiaofeng Yao
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, 300060, China; National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, 300060, China
| | - Mei Mei
- Department of Cell Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
| | - Yu Ren
- Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, 300060, China; National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, 300060, China; Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
| | - Xuan Zhou
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, 300060, China; National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, 300060, China.
| |
Collapse
|
9
|
Li S, Li Q, Zhou Q, Li S, Wang S, Yao Q, Ouyang C, Liu C, Li M. Attenuating Atherosclerosis through Inhibition of the NF- κB/NLRP3/IL-1 β Pathway-Mediated Pyroptosis in Vascular Smooth Muscle Cells (VSMCs). Cardiovasc Ther 2024; 2024:1506083. [PMID: 39742016 PMCID: PMC10985643 DOI: 10.1155/2024/1506083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 02/24/2024] [Accepted: 02/26/2024] [Indexed: 01/03/2025] Open
Abstract
Objective We investigated the effects of resveratrol (Res) and MCC950 on the pyroptosis of vascular smooth muscle cells (VSMCs) and the potential pathway. Methods and Results Compared with the control (Con) group, the atherosclerosis (AS) group showed calcified nodules, which suggested that the calcification medium induced the calcification of VSMCs. VSMCs showed proliferative activity and significantly attenuated calcification under treatment with 10 μmol/L Res. The calcium salt was detected by alizarin red S staining. Res and MCC950 downregulated the calcification, inflammatory, pyroptosis, and transcription factor-related indicators all decreased by RT-qPCR with Western blot and immunofluorescence. The results showed that Res and MCC950 refrained the calcification of VSMCs and that Res has a better effect than MCC950. Plaques and calcium salt deposits were present in the carotid region in the control group. More calcium salt deposits were evident in the plaques of the Par group by HE staining and alizarin red S staining. The calcification indexes BMP2, Runx2, and related indexes declined by immunofluorescence, which showed parthenolide-inhibited AS. The related protein expressions were consistent with the expression of the cell experiments. Conclusion Our data demonstrated that inflammatory response and pyroptosis exacerbate AS and unravel the link between VSMCs and the progression of AS lesions. Res and MCC950 inhibited the calcification of VSMCs by regulating NF-κB/NLRP3/IL-1β signaling axis. P53 can exacerbate the AS lesions by acting on NLRP3 inflammasome and pyroptosis. Our findings supported the clinical applications of Res and MCC950 in VSMCs individuals to counteract pyroptosis and AS, and P53 inhibitors also can be a potential treatment for AS.
Collapse
MESH Headings
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/metabolism
- Pyroptosis/drug effects
- Signal Transduction/drug effects
- NF-kappa B/metabolism
- Vascular Calcification/pathology
- Vascular Calcification/metabolism
- Vascular Calcification/drug therapy
- Interleukin-1beta/metabolism
- Resveratrol/pharmacology
- Cells, Cultured
- Furans/pharmacology
- Sulfonamides/pharmacology
- Disease Models, Animal
- Animals
- Anti-Inflammatory Agents/pharmacology
- Male
- Indenes/pharmacology
- Plaque, Atherosclerotic
- Inflammasomes/metabolism
- Humans
- Atherosclerosis/pathology
- Atherosclerosis/metabolism
- Atherosclerosis/drug therapy
- Atherosclerosis/genetics
- Carotid Artery Diseases/pathology
- Carotid Artery Diseases/metabolism
- Carotid Artery Diseases/drug therapy
- Carotid Artery Diseases/genetics
Collapse
Affiliation(s)
- Shihuan Li
- College of Medicine, Hubei University of Science and Technology, Xianning 437100, China
- Institute of Medicine, Hubei Key Laboratory of Diabetes, Hubei University of Science and Technology, Xianning, China
| | - Qingjie Li
- College of Medicine, Hubei University of Science and Technology, Xianning 437100, China
- Institute of Medicine, Hubei Key Laboratory of Diabetes, Hubei University of Science and Technology, Xianning, China
| | - Qiaofeng Zhou
- College of Medicine, Hubei University of Science and Technology, Xianning 437100, China
- Institute of Medicine, Hubei Key Laboratory of Diabetes, Hubei University of Science and Technology, Xianning, China
| | - Suqin Li
- School of Basic Medical Science, Hubei University of Science and Technology, Xianning, China
| | - Siqi Wang
- College of Medicine, Hubei University of Science and Technology, Xianning 437100, China
- Institute of Medicine, Hubei Key Laboratory of Diabetes, Hubei University of Science and Technology, Xianning, China
| | - Qing Yao
- Institute of Medicine, Hubei Key Laboratory of Diabetes, Hubei University of Science and Technology, Xianning, China
| | - Changhan Ouyang
- College of Medicine, Hubei University of Science and Technology, Xianning 437100, China
| | - Chao Liu
- Institute of Medicine, Hubei Key Laboratory of Diabetes, Hubei University of Science and Technology, Xianning, China
| | - Mincai Li
- Institute of Medicine, Hubei Key Laboratory of Diabetes, Hubei University of Science and Technology, Xianning, China
- School of Basic Medical Science, Hubei University of Science and Technology, Xianning, China
| |
Collapse
|
10
|
Khan F, Pandey P, Verma M, Upadhyay TK. Terpenoid-Mediated Targeting of STAT3 Signaling in Cancer: An Overview of Preclinical Studies. Biomolecules 2024; 14:200. [PMID: 38397437 PMCID: PMC10886526 DOI: 10.3390/biom14020200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/01/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Cancer has become one of the most multifaceted and widespread illnesses affecting human health, causing substantial mortality at an alarming rate. After cardiovascular problems, the condition has a high occurrence rate and ranks second in terms of mortality. The development of new drugs has been facilitated by increased research and a deeper understanding of the mechanisms behind the emergence and advancement of the disease. Numerous preclinical and clinical studies have repeatedly demonstrated the protective effects of natural terpenoids against a range of malignancies. Numerous potential bioactive terpenoids have been investigated in natural sources for their chemopreventive and chemoprotective properties. In practically all body cells, the signaling molecule referred to as signal transducer and activator of transcription 3 (STAT3) is widely expressed. Numerous studies have demonstrated that STAT3 regulates its downstream target genes, including Bcl-2, Bcl-xL, cyclin D1, c-Myc, and survivin, to promote the growth of cells, differentiation, cell cycle progression, angiogenesis, and immune suppression in addition to chemotherapy resistance. Researchers viewed STAT3 as a primary target for cancer therapy because of its crucial involvement in cancer formation. This therapy primarily focuses on directly and indirectly preventing the expression of STAT3 in tumor cells. By explicitly targeting STAT3 in both in vitro and in vivo settings, it has been possible to explain the protective effect of terpenoids against malignant cells. In this study, we provide a complete overview of STAT3 signal transduction processes, the involvement of STAT3 in carcinogenesis, and mechanisms related to STAT3 persistent activation. The article also thoroughly summarizes the inhibition of STAT3 signaling by certain terpenoid phytochemicals, which have demonstrated strong efficacy in several preclinical cancer models.
Collapse
Affiliation(s)
- Fahad Khan
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 602105, India;
| | - Pratibha Pandey
- University Centre for Research and Development, Chandigarh University, Gharuan, Mohali 140413, India
| | - Meenakshi Verma
- University Centre for Research and Development, Chandigarh University, Gharuan, Mohali 140413, India
- Department of Chemistry, University Institute of Sciences, Chandigarh University, Gharuan, Mohali 140413, India
| | - Tarun Kumar Upadhyay
- Department of Biotechnology, Parul Institute of Applied Sciences and Research and Development Cell, Parul University, Vadodara 391760, India;
| |
Collapse
|
11
|
Gu Q, Xiao YB, Wang Y. Silencing suppressor of cytokine signaling 3 induces apoptosis and activates the p-STAT3/NF-κB pathway in hypoxic cultivated H9c2 cells. J Physiol Biochem 2024; 80:127-136. [PMID: 37856074 DOI: 10.1007/s13105-023-00989-7] [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] [Received: 12/20/2022] [Accepted: 10/05/2023] [Indexed: 10/20/2023]
Abstract
Suppressor of cytokine signaling 3 (SOCS3) plays a significant role in the process of myocardial adaptation to chronic hypoxia. SOCS3 finely regulates cell signaling cross-talk that occurs between NF-κB and STAT3 during the compensatory protective response. However, the role and mechanism of SOCS3 in hypoxic cardiomyocytes are not fully understood. In the study, we investigated the effect of SOCS3 on the p65 and STAT3 signaling pathways and further examined the potential molecular mechanism involved in regulating apoptosis. Our data showed that SOCS3 silencing could upregulate Ac-p65, p-p65, and p-STAT3 expression in nuclear extracts of H9c2 cells that received hypoxic treatment for 24, 48, and 72 h. SOCS3 silencing also remarkably increased the DNA-binding activity of the p65 motif in hypoxic cultivated H9c2 cells. We also found that SOCS3 knockdown increased cleaved-caspase-3, Bax, and PUMA expression and decreased cleaved PARP and Bcl-2 in expression in hypoxic H9c2 cells. Silencing of SOCS3 caused an increase in LDH leakage from injured cardiomyocytes and reduced cell viability under conditions of hypoxic stress. Furthermore, SOCS3 silencing enhanced the apoptosis of H9c2 cells at 72 h of hypoxia. These findings suggest that knockdown of SOCS3 leads to excessive activation of the NF-κB pathway, which, in turn, might promote apoptosis under conditions of chronic hypoxia.
Collapse
Affiliation(s)
- Qiang Gu
- Department of Cardiovascular Surgery, Xinqiao Hospital, Second Affiliated Hospital of Army Military Medical University, No.183 Xinqiao Street, Shapingba District, Chongqing, 400037, China.
- Vascular Injury and Repair Laboratory, Xinqiao Hospital, Second Affiliated Hospital of Army Medical University, Chongqing, 400037, China.
| | - Ying-Bin Xiao
- Department of Cardiovascular Surgery, Xinqiao Hospital, Second Affiliated Hospital of Army Military Medical University, No.183 Xinqiao Street, Shapingba District, Chongqing, 400037, China
- Vascular Injury and Repair Laboratory, Xinqiao Hospital, Second Affiliated Hospital of Army Medical University, Chongqing, 400037, China
| | - Yong Wang
- Department of Cardiovascular Surgery, Xinqiao Hospital, Second Affiliated Hospital of Army Military Medical University, No.183 Xinqiao Street, Shapingba District, Chongqing, 400037, China
- Vascular Injury and Repair Laboratory, Xinqiao Hospital, Second Affiliated Hospital of Army Medical University, Chongqing, 400037, China
| |
Collapse
|
12
|
Wang KD, Zhu ML, Qin CJ, Dong RF, Xiao CM, Lin Q, Wei RY, He XY, Zang X, Kong LY, Xia YZ. Sanguinarine induces apoptosis in osteosarcoma by attenuating the binding of STAT3 to the single-stranded DNA-binding protein 1 (SSBP1) promoter region. Br J Pharmacol 2023; 180:3175-3193. [PMID: 37501645 DOI: 10.1111/bph.16202] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 06/19/2023] [Accepted: 07/20/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND AND PURPOSE Osteosarcoma, a primary malignant bone tumour prevalent among adolescents and young adults, remains a considerable challenge despite protracted progress made in enhancing patient survival rates over the last 40 years. Consequently, the development of novel therapeutic approaches for osteosarcoma is imperative. Sanguinarine (SNG), a compound with demonstrated potent anticancer properties against various malignancies, presents a promising avenue for exploration. Nevertheless, the intricate molecular mechanisms underpinning SNG's actions in osteosarcoma remain elusive, necessitating further elucidation. EXPERIMENTAL APPROACH Single-stranded DNA-binding protein 1 (SSBP1) was screened out by differential proteomic analysis. Apoptosis, cell cycle, reactive oxygen species (ROS) and mitochondrial changes were assessed via flow cytometry. Western blotting and quantitative real-time reverse transcription PCR (qRT-PCR) were used to determine protein and gene levels. The antitumour mechanism of SNG was explored at a molecular level using chromatin immunoprecipitation (ChIP) and dual luciferase reporter plasmids. KEY RESULTS Our investigation revealed that SNG exerted an up-regulated effect on SSBP1, disrupting mitochondrial function and inducing apoptosis. In-depth analysis uncovered a mechanism whereby SNG hindered the JAK/signal transducer and activator of transcription 3 (STAT3) signalling pathway, relieved the inhibitory effect of STAT3 on SSBP1 transcription, and inhibited the downstream PI3K/Akt/mTOR signalling axis, ultimately activating apoptosis. CONCLUSIONS AND IMPLICATIONS The study delved further into elucidating the anticancer mechanism of SNG in osteosarcoma. Notably, we unravelled the previously undisclosed apoptotic potential of SSBP1 in osteosarcoma cells. This finding holds substantial promise in advancing the development of novel anticancer drugs and identification of therapeutic targets.
Collapse
Affiliation(s)
- Kai-Di Wang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Miao-Lin Zhu
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing, China
| | - Cheng-Jiao Qin
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Rui-Fang Dong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Cheng-Mei Xiao
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Qing Lin
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Rong-Yuan Wei
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiao-Yu He
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xin Zang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ling-Yi Kong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yuan-Zheng Xia
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
13
|
Wang R, Du TT, Liu WQ, Liu YC, Yang YD, Hu JP, Ji M, Yang BB, Li L, Chen XG. Discovery, Optimization, and Evaluation of Novel N-(Benzimidazol-5-yl)-1,3,4-thiadiazol-2-amine Analogues as Potent STAT3 Inhibitors for Cancer Treatment. J Med Chem 2023; 66:12373-12395. [PMID: 37594012 DOI: 10.1021/acs.jmedchem.3c00863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/19/2023]
Abstract
Signal transducer and activator of transcription 3 (STAT3) is an attractive target for cancer therapy. However, identifying potent and selective STAT3 small-molecule inhibitors with drug-like properties remains challenging. Based on a scaffold combination strategy, compounds with a novel N-(benzimidazol-5-yl)-1,3,4-thiadiazol-2-amine scaffold were designed and their inhibition of the interleukin-6 (IL-6)/JAK/STAT3 pathway was tested in HEK-Blue IL-6 reporter cells. After optimization of lead compound 12, compound 40 was identified as a selective STAT3 inhibitor that directly binds the SH2 domain to inhibit STAT3 phosphorylation, translocation, and downstream gene transcription. Compound 40 exhibited antiproliferative activities against STAT3-overactivated DU145 (IC50 value = 2.97 μM) and MDA-MB-231 (IC50 value = 3.26 μM) cancer cells and induced cell cycle arrest and apoptosis. In the DU145 xenograft model, compound 40 showed in vivo antitumor efficacy following intraperitoneal administration, with a tumor growth inhibition rate of 65.3% at 50 mg/kg, indicating promise for further development.
Collapse
Affiliation(s)
- Ru Wang
- Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050,China
| | - Ting-Ting Du
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Wen-Qiang Liu
- Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050,China
| | - Yi-Chen Liu
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Ya-Dong Yang
- Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050,China
| | - Jin-Ping Hu
- Beijing Key Laboratory of Non-Clinical Drug Metabolism and PK/PD Study, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Ming Ji
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Bei-Bei Yang
- Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050,China
| | - Li Li
- Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050,China
| | - Xiao-Guang Chen
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
14
|
Zhu PL, Li JK, Jiang XL, Zhang SQ, Zhang Z, Wang Y, Zhang Z, Chen WQ, Yung KKL. A traditional prescription comprising Astragali Radix and Schisandra chinensis Fructus induces apoptosis and protective autophagy in hepatocellular carcinoma cells. JOURNAL OF ETHNOPHARMACOLOGY 2023; 312:116548. [PMID: 37100264 DOI: 10.1016/j.jep.2023.116548] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/29/2023] [Accepted: 04/23/2023] [Indexed: 05/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hepatocellular carcinoma (HCC) poses a growing challenge to global health efforts. The 5-year survival rate of HCC patients is still dismal. A traditional prescription Qi-Wei-Wan (QWW) comprising Astragali Radix and Schisandra chinensis Fructus has traditionally been used for HCC treatment according to traditional Chinese medicine theory, but the pharmacological basis is not clear. AIM OF THE STUDY This study aims to investigate the anti-HCC effects of an ethanolic extract of QWW (hereafter, QWWE) and the mechanism of action. MATERIALS AND METHODS An UPLC-Q-TOF-MS/MS method was developed to control the quality of QWWE. Two human HCC cell lines (HCCLM3 and HepG2) and a HCCLM3 xenograft mouse model were employed to investigate the anti-HCC effects of QWWE. The anti-proliferative effect of QWWE in vitro was determined by MTT, colony formation and EdU staining assays. Apoptosis and protein levels were examined by flow cytometry and Western blotting, respectively. Nuclear presence of signal transducer and activator of transcription 3 (STAT3) was examined by immunostaining. Transient transfection of pEGFP-LC3 and STAT3C plasmids was performed to assess autophagy and determine the involvement of STAT3 signaling in QWWE's anti-HCC effects, respectively. RESULTS We found that QWWE inhibited the proliferation of and triggered apoptosis in HCC cells. Mechanistically, QWWE inhibited the activation of SRC and STAT3 at Tyr416 and Tyr705, respectively; inhibited the nuclear translocation of STAT3; lowered Bcl-2 protein levels, while increased Bax protein levels in HCC cells. Over-activating STAT3 attenuated the cytotoxic and apoptotic effects of QWWE in HCC cells. Moreover, QWWE induced autophagy in HCC cells by inhibiting mTOR signaling. Blocking autophagy with autophagy inhibitors (3-methyladenine and chloroquine) enhanced the cytotoxicity, apoptotic effect and the inhibitory effect on STAT3 activation of QWWE. Intragastric administration of QWWE at 10 mg/kg and 20 mg/kg potently repressed tumor growth and inhibited STAT3 and mTOR signaling in tumor tissues, but did not significantly affect mouse body weight. CONCLUSION QWWE exhibited potent anti-HCC effects. Inhibiting the STAT3 signaling pathway is involved in QWWE-mediated apoptosis, while blocking mTOR signaling contributes to QWWE-mediated autophagy induction. Blockade of autophagy enhanced the anti-HCC effects of QWWE, indicating that the combination of an autophagy inhibitor and QWWE might be a promising therapeutic strategy for HCC management. Our findings provide pharmacological justifications for the traditional use of QWW in treating HCC.
Collapse
Affiliation(s)
- Pei-Li Zhu
- Department of Biology, Hong Kong Baptist University (HKBU), Kowloon Tong, Kowloon, Hong Kong, China; Golden Meditech Center for NeuroRegeneration Sciences (GMCNS), HKBU, Kowloon Tong, Hong Kong, China; HKBU Institute of Research and Continuing Education, Shenzhen, China
| | - Jun-Kui Li
- Department of Biology, Hong Kong Baptist University (HKBU), Kowloon Tong, Kowloon, Hong Kong, China; Golden Meditech Center for NeuroRegeneration Sciences (GMCNS), HKBU, Kowloon Tong, Hong Kong, China; HKBU Institute of Research and Continuing Education, Shenzhen, China
| | - Xiao-Li Jiang
- Department of Biology, Hong Kong Baptist University (HKBU), Kowloon Tong, Kowloon, Hong Kong, China; Golden Meditech Center for NeuroRegeneration Sciences (GMCNS), HKBU, Kowloon Tong, Hong Kong, China
| | - Shi-Qing Zhang
- Golden Meditech Center for NeuroRegeneration Sciences (GMCNS), HKBU, Kowloon Tong, Hong Kong, China; JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Zhu Zhang
- Department of Biology, Hong Kong Baptist University (HKBU), Kowloon Tong, Kowloon, Hong Kong, China; Golden Meditech Center for NeuroRegeneration Sciences (GMCNS), HKBU, Kowloon Tong, Hong Kong, China
| | - Ying Wang
- Department of Biology, Hong Kong Baptist University (HKBU), Kowloon Tong, Kowloon, Hong Kong, China; Golden Meditech Center for NeuroRegeneration Sciences (GMCNS), HKBU, Kowloon Tong, Hong Kong, China
| | - Zhang Zhang
- Department of Biology, Hong Kong Baptist University (HKBU), Kowloon Tong, Kowloon, Hong Kong, China; Golden Meditech Center for NeuroRegeneration Sciences (GMCNS), HKBU, Kowloon Tong, Hong Kong, China
| | - Wen-Qing Chen
- Department of Biology, Hong Kong Baptist University (HKBU), Kowloon Tong, Kowloon, Hong Kong, China; Golden Meditech Center for NeuroRegeneration Sciences (GMCNS), HKBU, Kowloon Tong, Hong Kong, China
| | - Ken-Kin-Lam Yung
- Department of Biology, Hong Kong Baptist University (HKBU), Kowloon Tong, Kowloon, Hong Kong, China; Golden Meditech Center for NeuroRegeneration Sciences (GMCNS), HKBU, Kowloon Tong, Hong Kong, China; HKBU Institute of Research and Continuing Education, Shenzhen, China.
| |
Collapse
|
15
|
Geetha R, Iyer S, Keechilat P, N GI, Thankappan KK, N V S. Evaluation of premetastatic changes in lymph nodes(pN0) of oral tongue tumour: A prospective observational Study. F1000Res 2023; 12:889. [PMID: 37786649 PMCID: PMC10541534 DOI: 10.12688/f1000research.138951.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/17/2023] [Indexed: 10/04/2023] Open
Abstract
Background: Tongue tumors show intra and inter-tumoral heterogenicity with high incidence, relapse and mortality rates necessitating further research. Recurrence/metastasis that occurs after surgical resection of primary cancer is often the reason for poor survival in these patients. Lymph nodes are the most common site of metastasis in tongue tumors. Therefore, premetastatic molecular changes can be best evaluated in lymph nodes which may epitomize the earliest events in the metastasis cascades. The presence of circulating tumor cells(CTCs) in the absence of nodal disease (N0) may represent tumor aggressiveness, suggesting an immune escape which may have high metastatic potential. This trial was developed to investigate the earliest pre-metastatic changes which may regulate tumor dormancy and predict metastasis. A better understanding of organotropism or pre-metastatic changes can help in theragnostic, thereby preventing the outbreak of overt metastasis. Methods: A single-institutional prospective observational cohort study. This trial will be conducted at a tertiary care Centre (Amrita Institute of Medical Sciences Kochi). Eligible patients will be enrolled after obtaining informed consent. The dissected lymph nodes will be subjected to histopathological and immunohistochemical analyses for premetastatic niche (PMN) formation. In addition, circulating tumor cells will be evaluated before treatment and 6 months after treatment. The patients will be followed up for a period of two years to correlate the findings with the recurrence-free survival. Expected results: The pre-metastatic changes, if detected will be a predictive biomarker. It may help to define future drug targets for metastasis chemoprevention . CTCs may define the tumor aggressiveness ,there by prognostication and helps in better disease management. Ethics and dissemination: The study has received the following approval: Ethics Committee of Amrita School of Medicine (ECASM-AIMS-2022-048).Trial Registered Prospectively( CTRI/2022/03/041256 ) on 22/03/2022 under Clinical Trial Registry of India.
Collapse
Affiliation(s)
- Rajalakshmi Geetha
- Head and Neck Surgery/Oncology, Amrita Institute of Medical Sciences - Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Subramania Iyer
- Head and Neck Surgery/Oncology, Amrita Institute of Medical Sciences - Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Pavithran Keechilat
- Medical Oncology, Amrita Institute of Medical Sciences - Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | | | - Krishna Kumar Thankappan
- Head and Neck Surgery/Oncology, Amrita Institute of Medical Sciences - Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Smitha N V
- Department of Pathology, Amrita Institute of Medical Sciences -Amrita Vishwa Vidyapeetham, Kochi, India
| |
Collapse
|
16
|
Hu YL, Gar-Lee Yue G, Li XR, Xu G, Bik-San Lau C. Structurally diverse spirocyclic polycyclic polyprenylated acylphloroglucinols from Hypericum ascyron linn. and their anti-tumor activity. PHYTOCHEMISTRY 2023; 212:113727. [PMID: 37207991 DOI: 10.1016/j.phytochem.2023.113727] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 05/10/2023] [Accepted: 05/15/2023] [Indexed: 05/21/2023]
Abstract
Ten spirocyclic polycyclic polyprenylated acylphloroglucinols (PPAP), hunascynols A-J (1-10), and 12 known analogs were isolated from the aerial parts of Hypericum ascyron Linn. Compounds 1 and 2, which share a 1,2-seco-spirocyclic PPAP skeleton, could be derived from spirocyclic PPAP, with a common octahydrospiro[cyclohexan-1,5'-indene]-2,4,6-trione core, through a cascade of Retro-Claisen, keto-enol tautomerism, and esterification reactions. Aldolization of normal spirocyclic PPAP yielded 3, which has a caged framework with a 6/5/6/5/6 ring system. The structures of these compounds were determined using spectroscopy and X-ray diffraction. The inhibitory activities of all isolates were tested in three human cancer cell lines and a zebrafish model. Compounds 1 and 2 displayed moderate cytotoxicity against HCT116 cells (IC50 6.87 and 9.86 μM, respectively). The mechanisms of these compounds were evaluated using Western blot assays. Compounds 3 and 5 inhibited the growth of sub-intestinal vessels in zebrafish embryos. Further, the target genes were screened using real-time PCR.
Collapse
Affiliation(s)
- Ya-Li Hu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming, 650201, People's Republic of China; University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Grace Gar-Lee Yue
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, People's Republic of China
| | - Xing-Ren Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming, 650201, People's Republic of China
| | - Gang Xu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, and Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming, 650201, People's Republic of China.
| | - Clara Bik-San Lau
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, People's Republic of China.
| |
Collapse
|
17
|
Wang X, Feng J, Wu R, Tan J, Huang Q, Phang Y, Zhang L, Fu W, Xu H, Zheng C. Garcinol and its analogues: Synthesis, cytotoxic activity and mechanistic investigation. Bioorg Chem 2023; 133:106389. [PMID: 36731298 DOI: 10.1016/j.bioorg.2023.106389] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023]
Abstract
Garcinol is a polyisoprenylated benzophenone isolated from Garcinia. It has been reported to have a variety of intriguing biological effects, including anticancer, anti-inflammatory, and antioxidant capabilities. The purpose of this research is to thoroughly evaluate garcinol and a series of its analogues in terms of synthesis, structural diversity, biosynthesis, and potential for preventing carcinoma cell proliferation. Garcinopicrobenzophenone and eugeniaphenone, which contain a unique cyclobutyl unit at C-5, were initially synthesized using the procedures utilized in the synthesis of garcinol. All the natural analogs of garcinol were produced at completion of the synthesis, and their structures and absolute configurations were clarified. Based on the synthesis, a possible biogenetic synthesis pathway towards cambogin, 13,14-didehydroxyisogarcinol via O-cyclization, and garcinopicrobenzophenone or eugeniaphenone via C-cyclization was proposed. The cytotoxicity of polyisoprenylated benzophenones produced in our group was tested, and the structure-activity relationship was summarized. The mechanism by which garcinol, cambogin, and 21' induce apoptosis was studied. Cambogin and 21' were shown to have a greater capacity to cause apoptosis in pancreatic cancer BXPC3 cells, and the suppression of BXPC3 cells by 21' might be attributed to the target of STAT3 signaling. Garcinol could cause pyroptosis and apoptosis in pancreatic cancer cells at the same time, which was the first time that garcinol was identified as a possible chemotherapeutic agent that could significantly promote pyroptosis in cancer cells.
Collapse
Affiliation(s)
- Xueying Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiling Feng
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Rong Wu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiaqi Tan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qing Huang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yeelin Phang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Li Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wenwei Fu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hongxi Xu
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Changwu Zheng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
18
|
Zhu M, Sun Y, Bai H, Wang Y, Yang B, Wang Q, Kuang H. Effects of saponins from Chinese herbal medicines on signal transduction pathways in cancer: A review. Front Pharmacol 2023; 14:1159985. [PMID: 37063281 PMCID: PMC10090286 DOI: 10.3389/fphar.2023.1159985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 03/16/2023] [Indexed: 03/31/2023] Open
Abstract
Cancer poses a serious threat to human health, and the search for safe and effective drugs for its treatment has aroused interest and become a long-term goal. Traditional Chinese herbal medicine (TCM), an ancient science with unique anti-cancer advantages, has achieved outstanding results in long-term clinical practice. Accumulating evidence shows that saponins are key bioactive components in TCM and have great research and development applications for their significant role in the treatment of cancer. Saponins are a class of glycosides comprising nonpolar triterpenes or sterols attached to hydrophilic oligosaccharide groups that exert antitumor effects by targeting the NF-κB, PI3Ks-Akt-mTOR, MAPK, Wnt-β-catenin, JAK-STAT3, APMK, p53, and EGFR signaling pathways. Presently, few advances have been made in physiological and pathological studies on the effect of saponins on signal transduction pathways involved in cancer treatment. This paper reviews the phytochemistry and extraction methods of saponins of TCM and their effects on signal transduction pathways in cancer. It aims to provide theoretical support for in-depth studies on the anticancer effects of saponins.
Collapse
Affiliation(s)
- Mingtao Zhu
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, China
| | - Yanping Sun
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, China
| | - Haodong Bai
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, China
| | - Yimeng Wang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, China
| | - Bingyou Yang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, China
| | - Qiuhong Wang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- *Correspondence: Qiuhong Wang, ; Haixue Kuang,
| | - Haixue Kuang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, China
- *Correspondence: Qiuhong Wang, ; Haixue Kuang,
| |
Collapse
|
19
|
Xia T, Zhang M, Lei W, Yang R, Fu S, Fan Z, Yang Y, Zhang T. Advances in the role of STAT3 in macrophage polarization. Front Immunol 2023; 14:1160719. [PMID: 37081874 PMCID: PMC10110879 DOI: 10.3389/fimmu.2023.1160719] [Citation(s) in RCA: 103] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/27/2023] [Indexed: 04/22/2023] Open
Abstract
The physiological processes of cell growth, proliferation, differentiation, and apoptosis are closely related to STAT3, and it has been demonstrated that aberrant STAT3 expression has an impact on the onset and progression of a number of inflammatory immunological disorders, fibrotic diseases, and malignancies. In order to produce the necessary biological effects, macrophages (M0) can be polarized into pro-inflammatory (M1) and anti-inflammatory (M2) types in response to various microenvironmental stimuli. STAT3 signaling is involved in macrophage polarization, and the research of the effect of STAT3 on macrophage polarization has gained attention in recent years. In order to provide references for the treatment and investigation of disorders related to macrophage polarization, this review compiles the pertinent signaling pathways associated with STAT3 and macrophage polarization from many fundamental studies.
Collapse
Affiliation(s)
- Tingting Xia
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
- Department of Dermatology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Meng Zhang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Wei Lei
- Department of Dermatology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Ruilin Yang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Shengping Fu
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Zhenhai Fan
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
- The Clinical Stem Cell Research Institute, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Ying Yang
- Department of Dermatology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Tao Zhang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
- Department of Dermatology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
- The Clinical Stem Cell Research Institute, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
- *Correspondence: Tao Zhang,
| |
Collapse
|
20
|
Sun H, Ma D, Cheng Y, Li J, Zhang W, Jiang T, Li Z, Li X, Meng H. The JAK-STAT Signaling Pathway in Epilepsy. Curr Neuropharmacol 2023; 21:2049-2069. [PMID: 36518035 PMCID: PMC10556373 DOI: 10.2174/1570159x21666221214170234] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 11/08/2022] [Accepted: 11/21/2022] [Indexed: 12/16/2022] Open
Abstract
Epilepsy is defined as spontaneous recurrent seizures in the brain. There is increasing evidence that inflammatory mediators and immune cells are involved in epileptic seizures. As more research is done on inflammatory factors and immune cells in epilepsy, new targets for the treatment of epilepsy will be revealed. The Janus kinase-signal transducer and transcriptional activator (JAKSTAT) signaling pathway is strongly associated with many immune and inflammatory diseases, At present, more and more studies have found that the JAK-STAT pathway is involved in the development and development of epilepsy, indicating the JAK-STAT pathway's potential promise as a target in epilepsy treatment. In this review, we discuss the composition, activation, and regulation of the JAK-STAT pathway and the relationship between the JAK-STAT pathway and epilepsy. In addition, we summarize the common clinical inhibitors of JAK and STAT that we would expect to be used in epilepsy treatment in the future.
Collapse
Affiliation(s)
- Huaiyu Sun
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Di Ma
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Yu Cheng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jiaai Li
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Wuqiong Zhang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Ting Jiang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Zhaoran Li
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Xuewei Li
- Department of Radiology, The First Hospital of Jilin University, Changchun, China
| | - Hongmei Meng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
21
|
Kobayashi K, Kogen H, Tamura O. Total Synthesis of Phaeosphaerides with STAT3 Inhibitory Activity. J SYN ORG CHEM JPN 2022. [DOI: 10.5059/yukigoseikyokaishi.80.755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Kenichi Kobayashi
- School of Pharmaceutical Sciences, Health Sciences University of Hokkaido
| | - Hiroshi Kogen
- Graduate School of Pharmaceutical Sciences, Meiji Pharmaceutical University
| | | |
Collapse
|
22
|
Shi X, Du TT, Zhang Z, Liu X, Yang Y, Xue N, Jiao X, Chen X, Xie P. (+)-Isocryptotanshinone derivatives and its simplified analogs as STAT3 signaling pathway inhibitors. Bioorg Chem 2022; 127:106015. [PMID: 35849894 DOI: 10.1016/j.bioorg.2022.106015] [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: 11/18/2021] [Revised: 06/13/2022] [Accepted: 07/06/2022] [Indexed: 11/15/2022]
Abstract
Isocryptotanshinone (ICTS), a natural product with potential signal transducer and activator of transcription-3 (STAT3) signaling pathway inhibitory activity, shows significant inhibitory activity against several tumors. In this study, a series of ICTS derivatives and simplified analogs containing a 1, 4-naphthoquinone core was designed, synthesized, and evaluated. The results demonstrated that most target compounds were potent STAT3 signaling pathway inhibitors based on their mechanism of inhibition of STAT3 phosphorylation. Moreover, based on the obtained data, the structure-activity relationship (SAR) was rationally deduced. Simultaneously, molecular docking of the compound 16r suggested its possible interaction mode with STAT3. To further verify anticancer activity, all target compounds were tested using HCT116, HepG2, MCF-7, A549, and U251 cell lines. Interestingly, compared with different tumor cell lines, the HCT-116 cell line was determined to be the most sensitive. Furthermore, compounds 21e, 16r, 28a, and 16e showed a dose-dependent inhibition of the growth of HCT116 cells. Thus, the SAR of ICTS derivatives and its simplified analogs was determined, and some of them were discovered to be potential anticancer candidates owing to their ability to inhibit the STAT3 signaling pathway.
Collapse
Affiliation(s)
- Xiang Shi
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Ting Ting Du
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhihui Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiaoyu Liu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ying Yang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Nina Xue
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiaozhen Jiao
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Xiaoguang Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Ping Xie
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
23
|
Lu PWA, Chou CH, Yang JS, Hsieh YH, Tsai MY, Lu KH, Yang SF. HO-3867 Induces Apoptosis via the JNK Signaling Pathway in Human Osteosarcoma Cells. Pharmaceutics 2022; 14:pharmaceutics14061257. [PMID: 35745828 PMCID: PMC9229449 DOI: 10.3390/pharmaceutics14061257] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/07/2022] [Accepted: 06/10/2022] [Indexed: 01/17/2023] Open
Abstract
Metastatic osteosarcoma often results in poor prognosis despite the application of surgical en bloc excision along with chemotherapy. HO-3867 is a curcumin analog that induces cell apoptosis in several cancers, but the apoptotic effect and its mechanisms on osteosarcoma cells are still unknown. After observing the decrease in cellular viability of three human osteosarcoma U2OS, HOS, and MG-63 cell lines, and the induction of cellular apoptosis and arrest in sub-G1 phase in U2OS and HOS cells by HO-3867, the human apoptosis array showed that heme oxygenase (HO)-1 and cleaved caspase-3 expressions had significant increases after HO-3867 treatment in U2OS cells and vice versa for cellular inhibitors of apoptosis (cIAP)1 and X-chromosome-linked IAP (XIAP). Western blot analysis verified the results and showed that HO-3867 activated the initiators of both extrinsic caspase 8 and intrinsic caspase 9, and significantly increased cleaved PARP expression in U2OS and HOS cells. Moreover, with the addition of HO-3867, ERK1/2, and JNK1/2 phosphorylation were increased in U2OS and HOS cells. Using the inhibitor of JNK (JNK in 8), HO-3867’s increases in cleaved caspases 3, 8, and 9 could be expectedly suppressed, indicating that JNK signaling is responsible for both apoptotic pathways, including extrinsic and intrinsic, in U2OS and HOS cells caused by HO-3867. Through JNK signaling, HO-3867 has proven to be effective in causing both extrinsic and intrinsic apoptotic pathways of human osteosarcoma cells.
Collapse
Affiliation(s)
| | - Chia-Hsuan Chou
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; (C.-H.C.); (J.-S.Y.); (Y.-H.H.); (M.-Y.T.)
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Jia-Sin Yang
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; (C.-H.C.); (J.-S.Y.); (Y.-H.H.); (M.-Y.T.)
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Yi-Hsien Hsieh
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; (C.-H.C.); (J.-S.Y.); (Y.-H.H.); (M.-Y.T.)
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Meng-Ying Tsai
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; (C.-H.C.); (J.-S.Y.); (Y.-H.H.); (M.-Y.T.)
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Ko-Hsiu Lu
- Department of Orthopedics, Chung Shan Medical University Hospital, Taichung 402, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
- Correspondence: (K.-H.L.); (S.-F.Y.); Tel.: +886-4-24739595-34253 (S.-F.Y.)
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; (C.-H.C.); (J.-S.Y.); (Y.-H.H.); (M.-Y.T.)
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan
- Correspondence: (K.-H.L.); (S.-F.Y.); Tel.: +886-4-24739595-34253 (S.-F.Y.)
| |
Collapse
|
24
|
Yun S, Lee YJ, Choi J, Kim ND, Han DC, Kwon BM. Acacetin Inhibits the Growth of STAT3-Activated DU145 Prostate Cancer Cells by Directly Binding to Signal Transducer and Activator of Transcription 3 (STAT3). Molecules 2021; 26:6204. [PMID: 34684783 PMCID: PMC8540902 DOI: 10.3390/molecules26206204] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/24/2021] [Accepted: 10/12/2021] [Indexed: 11/16/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) plays a critical role in the formation and growth of human cancer. Therefore, STAT3 is a therapeutic target for cancer drug discovery. Acacetin, a flavone present in various plants, inhibits constitutive and inducible STAT3 activation in STAT3-activated DU145 prostate cancer cells. Acacetin inhibits STAT3 activity by directly binding to STAT3, which we confirmed by a pull-down assay with a biotinylated compound and two level-free methods, namely, a drug affinity responsive target stability (DARTS) experiment and a cellular thermal shift assay (CETSA). Acacetin inhibits STAT3 phosphorylation at the tyrosine 705 residue and nuclear translocation in DU145 cells, which leads to the downregulation of STAT3 target genes. Acacetin then induces apoptosis in a time-dependent manner. Interestingly, acacetin induces the production of reactive oxygen species (ROS) that are not involved in the acacetin-induced inhibition of STAT3 activation because the suppressed p-STAT3 level is not rescued by treatment with GSH or NAC, which are general ROS inhibitors. We also found that acacetin inhibits tumor growth in xenografted nude mice. These results suggest that acacetin, as a STAT3 inhibitor, could be a possible drug candidate for targeting STAT3 for the treatment of cancer in humans.
Collapse
Affiliation(s)
- Sun Yun
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahakro, Daejeon 34141, Korea; (S.Y.); (Y.-J.L.); (J.C.)
- KRIBB School of Bioscience, University of Science and Technology in Korea, Daejeon 34113, Korea
| | - Yu-Jin Lee
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahakro, Daejeon 34141, Korea; (S.Y.); (Y.-J.L.); (J.C.)
| | - Jiyeon Choi
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahakro, Daejeon 34141, Korea; (S.Y.); (Y.-J.L.); (J.C.)
| | - Nam Doo Kim
- VORONOIBIO Inc., S 11th F, Thechnopark IT Center Songdo Kwahak-ro 32, Incheon 21984, Korea;
| | - Dong Cho Han
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahakro, Daejeon 34141, Korea; (S.Y.); (Y.-J.L.); (J.C.)
- KRIBB School of Bioscience, University of Science and Technology in Korea, Daejeon 34113, Korea
| | - Byoung-Mog Kwon
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahakro, Daejeon 34141, Korea; (S.Y.); (Y.-J.L.); (J.C.)
- KRIBB School of Bioscience, University of Science and Technology in Korea, Daejeon 34113, Korea
| |
Collapse
|
25
|
Discovery of Novel Polycyclic Polyprenylated Acylphloroglucinols from the Fruits of Garcinia xanthochymus as Antitumor Agents by Suppressing the STAT3 Signaling. Int J Mol Sci 2021; 22:ijms221910365. [PMID: 34638708 PMCID: PMC8508737 DOI: 10.3390/ijms221910365] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/20/2021] [Accepted: 09/22/2021] [Indexed: 12/18/2022] Open
Abstract
Pharmacologic studies have revealed that polycyclic polyprenylated acylphloroglucinols (PPAPs) collectively exhibit a broad range of biological activities, including antineoplastic potential. Here, six new PPAPs, named garcixanthochymones F–K (3, 5, 7, 8, 11, and 15), together with nine known analogues were isolated from the fruits of Garcinia xanthochymus. Their structures were elucidated based on the spectroscopic data, including UV, HRESIMS, and NMR, and quantum chemical calculations. All the isolated PPAPs were tested for anti-proliferative activity against four human tumor cell lines, including SGC7901, A549, HepG2, and MCF-7. Most of the PPAPs possessed high anti-proliferative activity with IC50 values in the range of 0.89 to 36.98 μM, and significant apoptosis was observed in MCF-7 cells exposed to compounds 2 and 5. Besides, docking results showed that compounds 2 and 5 could strongly combine with the Src homology 2 (SH2) domain of STAT3 via hydrogen bond and hydrophobic interaction, which is one of the key oncogenes and crucial therapeutic targets. Furthermore, compounds 2 and 5 efficiently downregulated the expression of p-STAT3Tyr705 and pivotal effector proteins involved in oncogenic signaling pathways of MCF-7 cells.
Collapse
|