1
|
Bai M, Huang Y, Suo X, Wang L, Han W, Zhang W. BET bromodomain inhibitors PFI-1 and CPI-203 suppress the development of follicular lymphoma via regulating Wnt/β-catenin signaling. Heliyon 2024; 10:e27149. [PMID: 38463851 PMCID: PMC10920712 DOI: 10.1016/j.heliyon.2024.e27149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 02/08/2024] [Accepted: 02/25/2024] [Indexed: 03/12/2024] Open
Abstract
Objective Follicular lymphoma (FL) is an indolent B-cell lymphoproliferative disorder, characterized by a lymphoid follicular pattern of growth. PFI-1 or CPI-203 has been known to effectively promote the inhibition of primary effusion lymphoma progression. This study aimed at investigating the anti-tumor properties of PFI-1 and CPI-203 on FL cells and uncover the underlying mechanism of action. Methods FL cells were treated with PFI-1 and CPI-203, and the treated cells were evaluated for their cell viability, cell cycle and apoptosis using CCK8, flow cytometry, and Western blot assays. A xenograft mouse model was used for assessing the in vivo effects of CPI-203 on tumorigenesis. Results PFI-1 or CPI-203 showed potential inhibitory effects on the cell viability of DOHH2 and RL cells in a dose-response-dependent manner. Furthermore, PFI-1 and CPI-203 inhibited cell growth, induced apoptosis of FL cells in vitro, and facilitated the translocation of β-catenin into cytoplasm both in vitro and in vivo. After engrafted with FL cells, CPI-203-treated mice got a longer duration of survival and a smaller tumor size than control mice. Mechanistically, PFI-1 and CPI-203 impede the activity of β-catenin and its downstream molecules by regulating the DVL2/GSK3β axis. Conclusion In conclusion, PFI-1 and CPI-203 may serve as potential anti-tumor inhibitors for the therapy of FL.
Collapse
Affiliation(s)
- Min Bai
- Hematology department, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, China
| | - Yunpeng Huang
- Hematology department, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, China
| | - Xinrui Suo
- Department of Prevention and Health, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, China
| | - Lieyang Wang
- Hematology department, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, China
| | - Weie Han
- Hematology department, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, China
| | - Weihua Zhang
- Hematology Department, The First Hospital, Shanxi Medical University, Taiyuan, 030001, China
| |
Collapse
|
2
|
Chen R, An J, Wang Y, Yang L, Lin Q, Wang Y. LINC01589 serves as a potential tumor-suppressor and immune-related biomarker in endometrial cancer: A review. Medicine (Baltimore) 2023; 102:e33536. [PMID: 37058060 PMCID: PMC10101251 DOI: 10.1097/md.0000000000033536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 03/24/2023] [Indexed: 04/15/2023] Open
Abstract
Currently, increasing attention is being paid to biomarkers in endometrial cancer. Immune infiltration of the tumor microenvironment has been shown to significantly affect the overall survival (OS) of uterine corpus endometrial carcinoma (UCEC) patients. LINC01589 is a long non-coding RNA (lncRNA) that is rarely reported in cancer and is assumed to play a role in immune regulation. We therefore evaluated the role of LINC01589 in UCEC using the Cancer Genome Atlas (TCGA) database. We analyzed the expression of LINC01589 using the gene expression profiles of LINC01589 in the UCEC projects in TCGA. Comparisons between the differentially expressed genes (DEGs) of the cancer and adjacent normal tissues of the UCEC projects revealed that LINC01589 expression was decreased in UCEC tissues. A multivariate cox regression analysis indicated that LINC01589 upregulation could serve as an independent prognostic factor for survival. Furthermore, there was a positive correlation between LINC01589 expression and B cell, T cell, NK cell, monocytic lineage, and myeloid dendritic cell infiltration in UCEC patients. In addition, 5 clusters of hub genes were detected by comparison of different expression levels of LINC01589 in the UCEC groups. The analysis of the reactome pathway using gene set enrichment analysis (GSEA) revealed immune-related pathways, including CD22-mediated B cell receptor (BCR) regulation and antigen-activated BCRs, leading to the generation of second messengers and complement cascade pathways that were significantly enriched in the high LINC01589 expression group. Thus, LINC01589 may serve as a prognostic biomarker, as it is associated with immune infiltration in UCEC.
Collapse
Affiliation(s)
- Ruixin Chen
- Department of Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Jian An
- Department of Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Yan Wang
- Department of Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Lingling Yang
- Department of Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Qingping Lin
- Department of Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Yanlong Wang
- Department of Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
3
|
Zhao Y, Niu Q, Yang S, Yang J, Zhang Z, Geng S, Fan J, Liu Z, Guan G, Liu Z, Zhou J, Hu H, Luo J, Yin H. Inhibition of BET Family Proteins Suppresses African Swine Fever Virus Infection. Microbiol Spectr 2022; 10:e0241921. [PMID: 35758684 PMCID: PMC9430462 DOI: 10.1128/spectrum.02419-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 05/23/2022] [Indexed: 11/20/2022] Open
Abstract
African swine fever (ASF), an acute, severe, highly contagious disease caused by African swine fever virus (ASFV) infection in domestic pigs and boars, has a mortality rate of up to 100%. Because effective vaccines and treatments for ASF are lacking, effective control of the spread of ASF remains a great challenge for the pig industry. Host epigenetic regulation is essential for the viral gene transcription. Bromodomain and extraterminal (BET) family proteins, including BRD2, BRD3, BRD4, and BRDT, are epigenetic "readers" critical for gene transcription regulation. Among these proteins, BRD4 recognizes acetylated histones via its two bromodomains (BD1 and BD2) and recruits transcription factors, thereby playing a pivotal role in transcriptional regulation and chromatin remodeling during viral infection. However, how BET/BRD4 regulates ASFV replication and gene transcription is unknown. Here, we randomly selected 12 representative BET family inhibitors and compared their effects on ASFV infection in pig primary alveolar macrophages (PAMs). These were found to inhibit viral infection by interfering viral replication. The four most effective inhibitors (ARV-825, ZL0580, I-BET-762, and PLX51107) were selected for further antiviral activity analysis. These BET/BRD4 inhibitors dose dependently decreased the ASFV titer, viral RNA transcription, and protein production in PAMs. Collectively, we report novel function of BET/BRD4 inhibitors in inducing suppression of ASFV infection, providing insights into the role of BET/BRD4 in the epigenetic regulation of ASFV and potential new strategies for ASF prevention and control. IMPORTANCE Due to the continuing spread of the ASFV in the world and the lack of commercial vaccines, the development of improved control strategies, including antiviral drugs, is urgently needed. BRD4 is an important epigenetic factor and has been commonly used for drug development for tumor treatment. Furthermore, the latest research showed that BET/BRD4 inhibition could suppress replication of virus. In this study, we first showed the inhibitory effect of agents targeting BET/BRD4 on ASFV infection with no significant host cytotoxicity. Then, we found four BET/BRD4 inhibitors that can inhibit ASFV replication, RNA transcription, and protein synthesis. Our findings support the hypothesis that BET/BRD4 can be considered as attractive host targets in antiviral drug discovery against ASFV.
Collapse
Affiliation(s)
- Yaru Zhao
- African Swine Fever Regional Laboratory, and State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, People’s Republic of China
| | - Qingli Niu
- African Swine Fever Regional Laboratory, and State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, People’s Republic of China
| | - Saixia Yang
- African Swine Fever Regional Laboratory, and State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, People’s Republic of China
| | - Jifei Yang
- African Swine Fever Regional Laboratory, and State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, People’s Republic of China
| | - Zhonghui Zhang
- African Swine Fever Regional Laboratory, and State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, People’s Republic of China
| | - Shuxian Geng
- African Swine Fever Regional Laboratory, and State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, People’s Republic of China
| | - Jie Fan
- African Swine Fever Regional Laboratory, and State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, People’s Republic of China
| | - Zhijie Liu
- African Swine Fever Regional Laboratory, and State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, People’s Republic of China
| | - Guiquan Guan
- African Swine Fever Regional Laboratory, and State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, People’s Republic of China
| | - Zhiqing Liu
- Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, China
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Haitao Hu
- Department of Microbiology and Immunology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, USA
| | - Jianxun Luo
- African Swine Fever Regional Laboratory, and State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, People’s Republic of China
| | - Hong Yin
- African Swine Fever Regional Laboratory, and State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, People’s Republic of China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis, Yangzhou University, Yangzhou, People’s Republic of China
| |
Collapse
|
4
|
D’Agostino A, Di Palma T, Cecchini Gualandi S, Boni R. Fluorescence Spectroscopy for the Diagnosis of Endometritis in the Mare. Animals (Basel) 2022; 12:ani12091157. [PMID: 35565583 PMCID: PMC9101999 DOI: 10.3390/ani12091157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/18/2022] Open
Abstract
By exploiting the PMN property to produce high quantities of oxygen peroxide to neutralize pathogens, the oxygen peroxide content of uterine cells was measured to diagnose endometritis. After preliminary in vitro studies in which endometrial cells from slaughtered mares were mixed with leukocytes from peripheral blood, endometrial samples were collected by uterine flushing from mares before insemination. Staining endometrial cells with H2DCF-DA was combined with hydroethidine to normalize the fluorescence intensity with the cellular content of the sample. Stained cell smears were assumed as the gold standard of endometritis, and based on this assay, the samples were considered positive (C+) and negative (C−) for endometritis. The amount and the turbidity of fluid recovered by uterine flushing were significantly (p < 0.01) higher in C+ than in C−. Moreover, the oxygen peroxide content of the endometrial cells was significantly higher in the C+ than in the C− group (6.31 ± 1.92 vs. 3.12 ± 1.26, p = 0.001). Using the value of 4.4 as the cutoff level of this fluorescence cytology assay, it was found that only one C− sample exceeded the cutoff level (false positives = 7.7%) while three C+ samples showed values below the cutoff level (false negative = 11.5%).
Collapse
Affiliation(s)
- Andrea D’Agostino
- Department of Sciences, University of Basilicata, Campus Macchia Romana, 85100 Potenza, Italy; (A.D.); (S.C.G.)
| | | | - Stefano Cecchini Gualandi
- Department of Sciences, University of Basilicata, Campus Macchia Romana, 85100 Potenza, Italy; (A.D.); (S.C.G.)
| | - Raffaele Boni
- Department of Sciences, University of Basilicata, Campus Macchia Romana, 85100 Potenza, Italy; (A.D.); (S.C.G.)
- Correspondence: ; Tel.: +39-0971-205017
| |
Collapse
|
5
|
Baudet A, Hultmark S, Ek F, Magnusson M. Small Molecule Screening of Primary Human Acute Myeloid Leukemia Using Co-culture and Multiplexed FACS Analysis. Bio Protoc 2022; 12:e4353. [PMID: 35434186 PMCID: PMC8983156 DOI: 10.21769/bioprotoc.4353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 10/20/2021] [Accepted: 01/25/2022] [Indexed: 12/29/2022] Open
Abstract
Ex vivo culture of primary acute myeloid leukemia (AML) cells is notoriously difficult due to spontaneous differentiation and cell death, which hinders mechanistic and translational studies. To overcome this bottleneck, we have implemented a co-culture system, where the OP9-M2 stromal cells support the growth, but most notably limit the differentiation of primary AML cells, thus allowing for mechanistic studies in vitro. Additionally, the co-culture on OP9-M2 stromal is superior in preserving surface marker expression of primary (adult and pediatric) AML cells in comparison to stroma-free culture. Thus, by combining the co-culture with multicolor, high-throughput FACS, we can evaluate the effect of hundreds of small molecules on multi-parametric processes including: cell survival, stemness (leukemic stem cells), and myeloid differentiation on the primary AML cells at a single-cell level. This method streamlines the identification of potential therapeutic agents, but also facilitates combinatorial screening aiming, for instance, at dissecting the regulatory pathways in a patient-specific manner. Graphic abstract: Schematic representation of the ex vivo small molecule screening of primary human acute myeloid leukemia. Irradiated, sub-confluent OP9-M2 stromal cells are plated in half-area 96 wells plates 4-16 h prior to adding primary AML cells. Compounds are added 36-48 h later and effects on cell number, leukemic stem cell population, and myeloid differentiation are quantifed by FACS after 4 days of treatment.
Collapse
Affiliation(s)
| | | | - Fredrik Ek
- Chemical Biology & Therapeutics, Lund University, Lund, Sweden
| | - Mattias Magnusson
- Lund Stem Cell Center, Lund University, Lund, Sweden
,
*For correspondence:
| |
Collapse
|