1
|
Zhong M, Zhang P, Yao S. LTBP2 silence suppresses glioblastoma proliferation and tumor growth of xenograft tumor mice through modulating JAK2/STAT2 signaling pathway. Tissue Cell 2025; 93:102697. [PMID: 39752891 DOI: 10.1016/j.tice.2024.102697] [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: 10/01/2024] [Revised: 11/29/2024] [Accepted: 12/18/2024] [Indexed: 03/05/2025]
Abstract
Glioblastoma is considered the most malignant central nervous system tumor. This study aimed to investigate effects of latent transforming growth factor-β binding protein-2 (LTBP2) on glioblastoma growth and associated mechanisms. LTBP2 gene transcription in glioblastoma was determined using RT-PCR. LTBP2 gene silencing lentiviral vectors were synthesized, and the highest efficient vector was selected to package sh-LTBP2 lentivirus. Xenograft tumor model was constructed by injecting sh-LTBP2-infected U87MG cells into mice, and tumor growth was evaluated. Proliferation, colony formation and migration of U87MG were verified with CCK-8, colony-formation assay and migration assay, and cell cycle was examined. Western blot and immunohistochemistry were performed to examine Janus kinase 2 (JAK2) and signal transducers and activators of transcription 2 (STAT2) phosphorylation. LTBP2 gene transcription in SVGp12 cells was significantly lower compared to U87MG, U251 and T98G cells (P < 0.001). LTBP2 gene silence suppressed U87MG cell proliferation, migration and colony formation compared to U87MG group and sh-NC group (P < 0.05). LTBP2 gene silence regulated phases of cell cycle in U87MG cells. JAK2/STAT2 participated in LTBP2 silence-induced decreased U87MG proliferation. LTBP2 gene silence suppressed tumor growth by modulating JAK2/STAT2 pathway in xenograft tumor mice. In conclusion, LTBP2 silence inhibited proliferation and migration of U87MG cells and tumor growth of xenograft tumor mice through modulating JAK2/STAT2 signaling pathway.
Collapse
Affiliation(s)
- Musheng Zhong
- Zunyi Medical University, Zhuhai, China; Pingsha Community Health Service center, Zhuhai, China
| | - Peng Zhang
- Department of Neurosurgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Shengtao Yao
- Department of Neurosurgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
2
|
Zhu T, Bei F, He R, Gong X, Chen Y, Yin Z, Wang J, Sun Y, Zhang Y. Genetic Diseases and Invasive Infections in Infants 100 Days or Younger. Pediatr Infect Dis J 2023:00006454-990000000-00432. [PMID: 37171972 DOI: 10.1097/inf.0000000000003939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
BACKGROUND Understanding the association of genetic diseases with invasive infections in neonates or infants is important, given the clinical and public health implications of genetic diseases. METHODS We conducted a retrospective case-control study over a 5-year period to investigate the association between genetic diseases and invasive infections in neonates or infants. The case group included 56 patients with laboratory-confirmed invasive infections and a genetic etiology identified by exome sequencing. Another 155 patients without a genetic etiology were selected as controls from the same pool of patients. RESULTS An overview of genetic diseases that predispose patients to develop invasive infections were outlined. We identified 7 independent predictors for genetic conditions, including prenatal findings [adjusted odds ratio (aOR), 38.44; 95% confidence interval (CI): 3.94-374.92], neonatal intensive care unit admission (aOR, 46.87; 95% CI: 6.30-348.93), invasive ventilation (aOR, 6.66; 95% CI: 3.07-14.46), bacterial infections (aOR, 0.21; 95% CI: 0.06-0.69), fever (aOR, 0.15; 95% CI: 0.08-0.30), anemia (aOR, 6.64; 95% CI: 3.02-14.59) and neutrophilia (aOR, 0.98; 95% CI: 0.96-0.99). The area under the curve for the predictive model was 0.921 (95% CI: 0.876-0.954). We also found that a genetic etiology [hazard ratio (HR), 7.25; 95% CI: 1.71-30.81], neurological manifestations (HR, 3.56; 95% CI: 1.29-9.88) and septic shock (HR, 13.83; 95% CI: 3.18-60.10) were associated with severe outcomes. CONCLUSIONS Our study established predictive variables and risk factors for an underlying genetic etiology and its mortality in neonates or infants with invasive infections. These findings could lead to risk-directed screening and treatment strategies, which may improve patient outcomes.
Collapse
Affiliation(s)
- Tianwen Zhu
- From the Department of Neonatology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fei Bei
- Department of Neonatology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruoqi He
- From the Department of Neonatology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaohui Gong
- Department of Neonatology, Shanghai Children's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Chen
- From the Department of Neonatology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhanghua Yin
- From the Department of Neonatology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Wang
- Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China; and
| | - Yu Sun
- Department of Pediatric Endocrinology/Genetics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institute for Pediatric Research, Shanghai, China
| | - Yongjun Zhang
- From the Department of Neonatology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Wang DP, Wu LH, Li R, He N, Zhang QY, Zhao CY, Jiang T. A Novel Aldisine Derivative Exhibits Potential Antitumor Effects by Targeting JAK/STAT3 Signaling. Mar Drugs 2023; 21:md21040218. [PMID: 37103357 PMCID: PMC10141377 DOI: 10.3390/md21040218] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
The JAK/STAT3 signaling pathway is aberrantly hyperactivated in many cancers, promoting cell proliferation, survival, invasiveness, and metastasis. Thus, inhibitors targeting JAK/STAT3 have enormous potential for cancer treatment. Herein, we modified aldisine derivatives by introducing the isothiouronium group, which can improve the antitumor activity of the compounds. We performed a high-throughput screen of 3157 compounds and identified compounds 11a, 11b, and 11c, which contain a pyrrole [2,3-c] azepine structure linked to an isothiouronium group through different lengths of carbon alkyl chains and significantly inhibited JAK/STAT3 activities. Further results showed that compound 11c exhibited the optimal antiproliferative activity and was a pan-JAKs inhibitor capable of inhibiting constitutive and IL-6-induced STAT3 activation. In addition, compound 11c influenced STAT3 downstream gene expression (Bcl-xl, C-Myc, and Cyclin D1) and induced the apoptosis of A549 and DU145 cells in a dose-dependent manner. The antitumor effects of 11c were further demonstrated in an in vivo subcutaneous tumor xenograft experiment with DU145 cells. Taken together, we designed and synthesized a novel small molecule JAKs inhibitor targeting the JAK/STAT3 signaling pathway, which has predicted therapeutic potential for JAK/STAT3 overactivated cancer treatment.
Collapse
Affiliation(s)
- Dong-Ping Wang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| | - Li-Hong Wu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266100, China
| | - Rui Li
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266100, China
| | - Na He
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266100, China
| | - Qian-Yue Zhang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266100, China
| | - Chen-Yang Zhao
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Department of Cancer Biology, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44195, USA
- Correspondence: (C.-Y.Z.); (T.J.)
| | - Tao Jiang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
- Correspondence: (C.-Y.Z.); (T.J.)
| |
Collapse
|
4
|
Xu J, Zhang X, Zhou M, Lu P, Xu Y, Wu L, Zhang Q, Wu Z, Xu X, Shi P, Wei Q, Li X, Song Q. Bioactive compound C498-0670 alleviates LPS-induced sepsis via JAK/STAT and NFκB signaling pathways. Front Immunol 2023; 14:1132265. [PMID: 37122731 PMCID: PMC10140310 DOI: 10.3389/fimmu.2023.1132265] [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: 12/27/2022] [Accepted: 03/27/2023] [Indexed: 05/02/2023] Open
Abstract
The JAK/STAT and NFκB signaling pathways are two major inflammatory signaling pathways that are usually activated simultaneously in the body's inflammatory response to bacterial or viral infections. Hyperactivation of these two prominent signaling pathways is associated with various immune-related diseases and mortality, pointing to an urgent need for drug development targeting JAK/STAT and/or NFκB signaling. In this study, we screened 18,840 compounds using our well-established dual STAT-NFκB driven luciferase reporter based high-throughput screening system and identified a bioactive compound C498-0670, which inhibits both JAK/STAT and NFκB signaling. C498-0670 inhibits the activation of STATs and p-IKKα/β in both the immortalized cell lines and primary peritoneal macrophages, while suppressing the expression of LPS-induced inflammatory mediators in vitro. In addition, the overall anti-inflammatory effects of C498-0670 were investigated using transcriptome sequencing and bioinformatics approaches. C498-0670 was predicted to alleviate sepsis/septic shock by disease/function analysis using IPA software, which was further verified in the LPS-induced mouse sepsis model in vivo. C498 reduced LPS-induced liver and kidney damage, myeloid cell infiltration, and pro-inflammatory cytokine and chemokine production in vivo. Furthermore, the SPR-HPLC-MS-based target fishing approach was used to identify the putative drug targets, and the high affinities of JAK2 (JAK/STAT signaling), NFKBIA (NFκB signaling), and IL-1β, NLRP1b (inflammasome signaling) for C498-0670 were verified by molecular docking approach. These results suggest that C498-0670 can be used as a dual-target inhibitor of JAK/STAT and NFκB signaling pathways for the treatment of various inflammatory diseases, especially septic shock.
Collapse
Affiliation(s)
- Jing Xu
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao Pilot National Laboratory for Marine Science and Technology, Qingdao, Shandong, China
| | - Xinxin Zhang
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao Pilot National Laboratory for Marine Science and Technology, Qingdao, Shandong, China
| | - Mingming Zhou
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao Pilot National Laboratory for Marine Science and Technology, Qingdao, Shandong, China
| | - Peizhe Lu
- Department of Neuroscience, University of Michigan, Ann Arbor, MI, United States
| | - Yuting Xu
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Lihong Wu
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Qianyue Zhang
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Zhihua Wu
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao Pilot National Laboratory for Marine Science and Technology, Qingdao, Shandong, China
| | - Xiaoyu Xu
- College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Pengfei Shi
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao Pilot National Laboratory for Marine Science and Technology, Qingdao, Shandong, China
| | - Qingda Wei
- School of Medicine, Zhengzhou University, Zhengzhou, China
| | - Xiaoyu Li
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
- *Correspondence: Xiaoyu Li, ; Qiaoling Song,
| | - Qiaoling Song
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao Pilot National Laboratory for Marine Science and Technology, Qingdao, Shandong, China
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- *Correspondence: Xiaoyu Li, ; Qiaoling Song,
| |
Collapse
|
5
|
Liu R, Tian Y, Wang J, Wang Z, Li X, Zhao C, Yao R, Li S, Yuan L, Yang J, Shi D. Visible light-initiated radical 1,3-difunctionalization of β,γ-unsaturated ketones. SCIENCE ADVANCES 2022; 8:eabq8596. [PMID: 36490351 PMCID: PMC9733936 DOI: 10.1126/sciadv.abq8596] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 11/03/2022] [Indexed: 06/17/2023]
Abstract
Radical-mediated 1,2-difunctionalization of olefins is a well-established synthetic technique widely used in the rapid construction of structurally diverse molecular entities. However, radical-mediated 1,3-difunctionalization reactions are rare, and the substrates are generally limited to strained skeletons. Here, we report a practical approach for 1,3-difunctionalization of available β,γ-unsaturated ketones via a radical cascade process including visible light-irradiated radical addition, thermodynamic stability-driven 1,2-carbonyl migration from unactivated all-carbon quaternary center, and terminal C-radical varied transformations. Various highly functionalized alkyl skeletons with different valuable functional groups at positions 1 and 3 and the carbonyl group at position 2 have been synthesized through a radical chain pathway or Cu-catalyzed Ritter-type reaction. Moreover, this protocol provides a real case of diversity-oriented radical rearrangement for drug discovery. We identified a previously unknown chemotype of dual inhibitors for hypoxia-inducible factor (HIF) and WNT signaling pathways from products. These small-molecule inhibitors could suppress HIF and WNT signaling-dependent HCT116 cell growth in 2D and 3D culture systems.
Collapse
Affiliation(s)
- Ruihua Liu
- State Key Laboratory of Microbial Technology, Shandong University, 72 Binhai Road, Qingdao 266237, Shandong, P. R. China
| | - Yang Tian
- State Key Laboratory of Microbial Technology, Shandong University, 72 Binhai Road, Qingdao 266237, Shandong, P. R. China
| | - Jie Wang
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, Shandong, P. R. China
| | - Zemin Wang
- State Key Laboratory of Microbial Technology, Shandong University, 72 Binhai Road, Qingdao 266237, Shandong, P. R. China
| | - Xiangqian Li
- State Key Laboratory of Microbial Technology, Shandong University, 72 Binhai Road, Qingdao 266237, Shandong, P. R. China
| | - Chenyang Zhao
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, Shandong, P. R. China
| | - Ruoyu Yao
- State Key Laboratory of Microbial Technology, Shandong University, 72 Binhai Road, Qingdao 266237, Shandong, P. R. China
| | - Shuo Li
- State Key Laboratory of Microbial Technology, Shandong University, 72 Binhai Road, Qingdao 266237, Shandong, P. R. China
| | - Leifeng Yuan
- State Key Laboratory of Microbial Technology, Shandong University, 72 Binhai Road, Qingdao 266237, Shandong, P. R. China
| | - Jinbo Yang
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, Shandong, P. R. China
| | - Dayong Shi
- State Key Laboratory of Microbial Technology, Shandong University, 72 Binhai Road, Qingdao 266237, Shandong, P. R. China
- Laboratory of Marine Drugs and Biological Products, Pilot National Laboratory for Marine Science and Technology, 168 Weihai Road, Qingdao 266237, Shandong, P. R. China
| |
Collapse
|
6
|
Hong L, He M, Li S, Zhao J. Predicting for anti-(mutant) SARS-CoV-2 and anti-inflammation compounds of Lianhua Qingwen Capsules in treating COVID-19. Chin Med 2022; 17:84. [PMID: 35799189 PMCID: PMC9261255 DOI: 10.1186/s13020-022-00637-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/18/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Lianhua Qingwen Capsules (LHQW) is a traditional Chinese medicine prescription commonly used to treat viral influenza in China. There has been sufficient evidence that LHQW could effectively treat COVID-19. Nevertheless, the potential anti-(mutant) SARS-CoV-2 and anti-inflammation compounds in LHQW are still vague. METHODS The compounds of LHQW and targets were collected from TCMSP, TCMID, Shanghai Institute of Organic Chemistry of CAS database, and relevant literature. Autodock Vina was used to carry out molecular docking. The pkCSM platform to predict the relevant parameters of compound absorption in vivo. The protein-protein interaction (PPI) network was constructed by the STRING database. The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis was carried out by Database for Annotation, Visualization, and Integrated Discovery (DAVID). The anti-(mutant) SARS-CoV-2 and anti-inflammation networks were constructed on the Cytoscape platform. RESULTS 280 compounds, 16 targets related to SARS-CoV-2, and 54 targets related to cytokine storm were obtained by screening. The key pathways Toll-like receptor signaling, NOD-like receptor signal pathway, and Jak-STAT signaling pathway, and the core targets IL6 were obtained by PPI network and KEGG pathway enrichment analysis. The network analysis predicted and discussed the 16 main anti-SARS-CoV-2 active compounds and 12 main anti-inflammation active compounds. Ochnaflavone and Hypericin are potential anti-mutant virus compounds in LHQW. CONCLUSIONS In summary, this study explored the potential anti-(mutant) SARS-CoV-2 and anti-inflammation compounds of LHQW against COVID-19, which can provide new ideas and valuable references for discovering active compounds in the treatment of COVID-19.
Collapse
Affiliation(s)
- Liang Hong
- grid.437123.00000 0004 1794 8068State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China ,grid.437123.00000 0004 1794 8068Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macau, China
| | - Min He
- grid.412982.40000 0000 8633 7608Department of Pharmaceutical Engineering, School of Chemical Engineering, Xiangtan University, Xiangtan, China
| | - Shaoping Li
- grid.437123.00000 0004 1794 8068State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China ,grid.437123.00000 0004 1794 8068Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macau, China
| | - Jing Zhao
- grid.437123.00000 0004 1794 8068State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China ,grid.437123.00000 0004 1794 8068Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macau, China
| |
Collapse
|
7
|
Xu L, Zhang H, Wang Y, Yang A, Dong X, Gu L, Liu D, Ding N, Jiang Y. FABP4 activates the JAK2/STAT2 pathway via Rap1a in the homocysteine-induced macrophage inflammatory response in ApoE -/- mice atherosclerosis. J Transl Med 2022; 102:25-37. [PMID: 34725437 PMCID: PMC8695379 DOI: 10.1038/s41374-021-00679-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 12/02/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory vascular disease, and inflammation plays a critical role in its formation and progression. Elevated serum homocysteine (Hcy) is an independent risk factor for atherosclerosis. Previous studies have shown that fatty acid binding protein 4 (FABP4) plays an important role in macrophage inflammation and lipid metabolism in atherosclerosis induced by Hcy. However, the underlying molecular mechanism of FABP4 in Hcy-induced macrophage inflammation remains unknown. In this study, we found that FABP4 activated the Janus kinase 2/signal transducer and activator of transcription 2 (JAK2/STAT2) pathway in macrophage inflammation induced by Hcy. Of note, we further observed that ras-related protein Rap-1a (Rap1a) induced the Tyr416 phosphorylation and membrane translocation of non-receptor tyrosine kinase (c-Src) to activate the JAK2/STAT2 pathway. In addition, the suppressor of cytokine signaling 1 (SOCS1)-a transcriptional target of signal transducer and activator of transcription (STATs) inhibited the JAK2/STAT2 pathway and Rap1a expression via a negative feedback loop. In summary, these results demonstrated that FABP4 promotes c-Src phosphorylation and membrane translocation via Rap1a to activate the JAK2/STAT2 pathway, contributing to Hcy-accelerated macrophage inflammation in ApoE-/- mice.
Collapse
Affiliation(s)
- Lingbo Xu
- Department of Pathophysiology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
- National Health Commission Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, 750004, China
| | - Huiping Zhang
- National Health Commission Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, 750004, China
- Prenatal Diagnosis Center of Ningxia Medical University General Hospital, Yinchuan, 750004, China
| | - Yanhua Wang
- National Health Commission Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, 750004, China
- Department of Gynecology, General Hospital of Ningxia Medical University, Yinchuan, 750004, China
| | - Anning Yang
- Department of Pathophysiology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
- National Health Commission Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, 750004, China
| | - Xiaoyan Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
- National Health Commission Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, 750004, China
| | - Lingyu Gu
- National Health Commission Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, 750004, China
| | - Dayue Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
- National Health Commission Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, 750004, China
| | - Ning Ding
- National Health Commission Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, 750004, China
| | - Yideng Jiang
- Department of Pathophysiology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China.
- National Health Commission Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Medical University, Yinchuan, 750004, China.
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
8
|
Yan Y, Zhang Y, Li M, Zhang Y, Zhang X, Zhang X, Xu Y, Wei W, Wang J, Xu X, Song Q, Zhao C. C644-0303, a small-molecule inhibitor of the Wnt/β-catenin pathway, suppresses colorectal cancer growth. Cancer Sci 2021; 112:4722-4735. [PMID: 34431598 PMCID: PMC8586673 DOI: 10.1111/cas.15118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/13/2021] [Accepted: 08/19/2021] [Indexed: 12/15/2022] Open
Abstract
The Wnt/β‐catenin signaling pathway plays an important role in tissue homeostasis, and its malignant activation is closely related to the occurrence and development of many cancers, especially colorectal cancer with adenomatous polyposis coli (APC) and CTNNB1 mutations. By applying a TCF/lymphoid‐enhancing factor (LEF) luciferase reporter system, the high‐throughput screening of 18 840 small‐molecule compounds was performed. A novel scaffold compound, C644‐0303, was identified as a Wnt/β‐catenin signaling inhibitor and exhibited antitumor efficacy. It inhibited both constitutive and ligand activated Wnt signals and its downstream gene expression. Functional studies showed that C644‐0303 causes cell cycle arrest, induces apoptosis, and inhibits cancer cell migration. Moreover, transcription factor array indicated that C644‐0303 could suppress various tumor‐promoting transcription factor activities in addition to Wnt/β‐catenin. Finally, C644‐0303 suppressed tumor spheroidization in a 3‐dimensional cell culture model and inhibited xenograft tumor growth in mice. In conclusion, we report a novel structural small molecular inhibitor targeting the Wnt/β‐catenin signaling pathway that has therapeutic potential for colorectal cancer treatment.
Collapse
Affiliation(s)
- Yu Yan
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Yidan Zhang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Mengyuan Li
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Yazhuo Zhang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Xinxin Zhang
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| | - Xiaonan Zhang
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| | - Yuting Xu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Wei Wei
- School of Life Science, Lanzhou University, Lanzhou, China
| | - Jie Wang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Xiaohan Xu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Qiaoling Song
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China.,Innovation Platform of Marine Drug Screening & Evaluation, Qingdao Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| | - Chenyang Zhao
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China.,Innovation Platform of Marine Drug Screening & Evaluation, Qingdao Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
9
|
Li M, Yan Y, Zhang X, Zhang Y, Xu X, Zhang L, Lu L, Wang J, Zhang Y, Song Q, Zhao C. Scaffold compound L971 exhibits anti-inflammatory activities through inhibition of JAK/STAT and NFκB signalling pathways. J Cell Mol Med 2021; 25:6333-6347. [PMID: 34018320 PMCID: PMC8256347 DOI: 10.1111/jcmm.16609] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 12/17/2022] Open
Abstract
JAK/STAT and NFκB signalling pathways play essential roles in regulating inflammatory responses, which are important pathogenic factors of various serious immune-related diseases, and function individually or synergistically. To find prodrugs that can treat inflammation, we performed a preliminary high-throughput screening of 18 840 small molecular compounds and identified scaffold compound L971 which significantly inhibited JAK/STAT and NFκB driven luciferase activities. L971 could inhibit the constitutive and stimuli-dependent activation of STAT1, STAT3 and IκBα and could significantly down-regulate the proinflammatory gene expression in mouse peritoneal macrophages stimulated by LPS. Gene expression profiles upon L971 treatment were determined using high-throughput RNA sequencing, and significant differentially up-regulated and down-regulated genes were identified by DESeq analysis. The bioinformatic studies confirmed the anti-inflammatory effects of L971. Finally, L971 anti-inflammatory character was further verified in LPS-induced sepsis shock mouse model in vivo. Taken together, these data indicated that L971 could down-regulate both JAK/STAT and NFκB signalling activities and has the potential to treat inflammatory diseases such as sepsis shock.
Collapse
Affiliation(s)
- Mengyuan Li
- School of Medicine and PharmacyOcean University of ChinaQingdaoChina
| | - Yu Yan
- School of Medicine and PharmacyOcean University of ChinaQingdaoChina
| | - Xinxin Zhang
- Innovation Platform of Marine Drug Screening & EvaluationQingdao National Laboratory for Marine Science and TechnologyQingdaoChina
| | - Yidan Zhang
- School of Medicine and PharmacyOcean University of ChinaQingdaoChina
| | - Xiaohan Xu
- School of Medicine and PharmacyOcean University of ChinaQingdaoChina
| | - Lei Zhang
- School of Medicine and PharmacyOcean University of ChinaQingdaoChina
| | - Liangliang Lu
- School of Life ScienceLanzhou UniversityLanzhouChina
| | - Jie Wang
- School of Medicine and PharmacyOcean University of ChinaQingdaoChina
| | - Yazhuo Zhang
- School of Medicine and PharmacyOcean University of ChinaQingdaoChina
| | - Qiaoling Song
- School of Medicine and PharmacyOcean University of ChinaQingdaoChina
- Innovation Platform of Marine Drug Screening & EvaluationQingdao National Laboratory for Marine Science and TechnologyQingdaoChina
| | - Chenyang Zhao
- School of Medicine and PharmacyOcean University of ChinaQingdaoChina
- Innovation Platform of Marine Drug Screening & EvaluationQingdao National Laboratory for Marine Science and TechnologyQingdaoChina
| |
Collapse
|