101
|
Guan S, Chen X, Chen Y, Xie W, Liang H, Zhu X, Yang Y, Fang W, Huang Y, Zhao H, Zhuang W, Liu S, Huang M, Wang X, Zhang L. FOXM1 Variant Contributes to Gefitinib Resistance via Activating Wnt/β-Catenin Signal Pathway in Patients with Non-Small Cell Lung Cancer. Clin Cancer Res 2022; 28:3770-3784. [PMID: 35695863 DOI: 10.1158/1078-0432.ccr-22-0791] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/04/2022] [Accepted: 06/08/2022] [Indexed: 12/09/2022]
Abstract
PURPOSE Although gefitinib prolonged the progression-free survival (PFS) of patients with non-small cell lung cancer (NSCLC), unpredictable resistance limited its clinical efficacy. Novel predictive biomarkers with explicit mechanisms are urgently needed. EXPERIMENTAL DESIGN A total of 282 patients with NSCLC with gefitinib treatment were randomly assigned in a 7:3 ratio to exploratory (n = 192) and validation (n = 90) cohorts. The candidate polymorphisms were selected with Haploview4.2 in Hapmap and genotyped by a MassARRAY system, and the feature variables were identified through Randomforest Survival analysis. Tanswell and clonogenic assays, base editing and cell-derived tumor xenograft model were performed to uncover the underlying mechanism. RESULTS We found that the germline missense polymorphism rs3742076 (A>G, S628P), located in transactivation domain of FOXM1, was associated with PFS in exploratory (median PFS: GG vs. GA&AA, 9.20 vs. 13.37 months, P = 0.00039, HR = 2.399) and validation (median PFS: GG vs. GA&AA, 8.13 vs. 13.80 months, P = 0.048, HR = 2.628) cohorts. We elucidated that rs3742076_G conferred resistance to gefitinib by increasing protein stability of FOXM1 and facilitating an aggressive phenotype in vitro and in vivo through activating wnt/β-catenin signaling pathway. Meanwhile, FOXM1 level was highly associated with prognosis in patients with EGFR-mutant NSCLC. Mechanistically, FOXM1 rs3742076_G upregulated wnt/β-catenin activity by directly binding to β-catenin in cytoplasm and promoting transcription of β-catenin in nucleus. Remarkably, inhibition of β-catenin markedly reversed rs3742076_G-induced gefitinib resistance and aggressive phenotypes. CONCLUSIONS These findings characterized rs3742076_G as a gain-of-function polymorphism in mediating gefitinib resistance and tumor aggressiveness, and highlighted the variant as a predictive biomarker in guiding gefitinib treatment.
Collapse
Affiliation(s)
- Shaoxing Guan
- Laboratory of Drug Metabolism and Pharmacokinetics, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou City, Guangzhou, P. R. China
| | - Xi Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Youhao Chen
- Laboratory of Drug Metabolism and Pharmacokinetics, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou City, Guangzhou, P. R. China
| | - Wen Xie
- Department of Pharmaceutical Sciences and Center for Pharmacogenetics, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania
| | - Heng Liang
- Laboratory of Drug Metabolism and Pharmacokinetics, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou City, Guangzhou, P. R. China
| | - Xia Zhu
- Laboratory of Drug Metabolism and Pharmacokinetics, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou City, Guangzhou, P. R. China
| | - Yunpeng Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Wenfeng Fang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Yan Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Hongyun Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Wei Zhuang
- Laboratory of Drug Metabolism and Pharmacokinetics, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou City, Guangzhou, P. R. China
| | - Shu Liu
- Laboratory of Drug Metabolism and Pharmacokinetics, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou City, Guangzhou, P. R. China
| | - Min Huang
- Laboratory of Drug Metabolism and Pharmacokinetics, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou City, Guangzhou, P. R. China
| | - Xueding Wang
- Laboratory of Drug Metabolism and Pharmacokinetics, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou City, Guangzhou, P. R. China
| | - Li Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| |
Collapse
|
102
|
Micro RNA-411 Expression Improves Cardiac Phenotype Following Myocardial Infarction in Mice. JACC Basic Transl Sci 2022; 7:859-875. [PMID: 36317138 PMCID: PMC9617134 DOI: 10.1016/j.jacbts.2022.05.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 05/13/2022] [Accepted: 05/16/2022] [Indexed: 01/23/2023]
Abstract
Induction of endogenous regenerative capacity has emerged as one promising approach to repair damaged hearts following myocardial infarction (MI). Re-expression of factors that are exclusively expressed during embryonic development may reactivate the ability of adult cardiomyocytes to regenerate. Here, we identified miR-411 as a potent inducer of cardiomyocyte proliferation. Overexpression of miR-411 in the heart significantly increased cardiomyocyte proliferation and survival in a model MI. We found that miR-411 enhances the activity of YAP, the main downstream effector of the Hippo pathway, in cardiomyocytes. In conclusion, miR-411 induces cardiomyocyte regeneration and improves cardiac function post-MI likely by modulating the Hippo/YAP pathway.
Collapse
Key Words
- CVEC, cardiac vascular endothelial cells
- EdU, 5-ethynyl-2'-deoxyuridine
- Hippo pathway
- LAD, left anterior descending coronary artery
- MI, myocardial infarction
- MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
- NFAT, nuclear factor of activated T cells
- NRCF, neonatal rat cardiac fibroblast
- NRCM, neonatal rat cardiomyocytes
- PCR, polymerase chain reaction
- PEI, polyethylenimine
- cTnI, cardiac troponin I
- cardiac remodeling
- heart failure
- miRNA, microRNA
- microRNA-411
- myocardial infarction
- pHH3, phosphohistone H3
- qPCR, quantitative PCR
Collapse
|
103
|
Zhang M, Cai X, Liu J, Zhou J, Shi Q, Jiang Y, Kang N, Zhen X, Wu M, Qiu P, Yan G, Sun H, Li D. A novel lncRNA lncSAMD11-1: 1 interacts with PIP4K2A to promote endometrial decidualization by stabilizing FoxO1 nuclear localization. Int J Biochem Cell Biol 2022; 151:106280. [PMID: 35987479 DOI: 10.1016/j.biocel.2022.106280] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/09/2022] [Accepted: 08/14/2022] [Indexed: 11/29/2022]
Abstract
Decidualization is essential for a successful pregnancy and determines embryo implantation and pregnancy maintenance. Abnormal decidualization is one of the main causes of recurrent implantation failure (RIF). Studies have shown that large amounts of long noncoding RNAs (lncRNAs) are abnormally expressed in endometrial samples from patients with RIF. However, the functional contributions of lncRNAs to decidualization in RIF have not been explored. In this study, we found that lncSAMD11-1:1 was significantly declined in the endometria of patients with RIF. The knockdown of lncSAMD11-1:1 in human endometrial stromal cells (hESCs) restrained decidualization and embryo implantation in vitro, while the overexpression of lncSAMD11-1:1 facilitated hESC decidualization and embryo implantation in vitro and ameliorated decidualization in RIF patients. Mechanistically, lncSAMD11-1:1 and phosphatidylinositol-5-phosphate 4-kinase type 2 alpha (PIP4K2A) translocated out of nucleus and bound to each other during decidualization, thereby inhibiting the phosphorylation of AKT and promoting FoxO1 nuclear localization. These data suggest that lncSAMD11-1:1 might be a critical novel lncRNA functionally required for human decidualization, and the dysregulation of lncSAMD11-1:1 in the endometrium may be a new predisposing factor of RIF.
Collapse
Affiliation(s)
- Mei Zhang
- Center for Reproductive Medicine and Obstetrics & Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People's Republic of China; Center for Molecular Reproductive Medicine, Nanjing University, Nanjing 210008, People's Republic of China
| | - Xinyu Cai
- Center for Reproductive Medicine and Obstetrics & Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People's Republic of China
| | - Jingyu Liu
- Center for Reproductive Medicine and Obstetrics & Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People's Republic of China; Center for Molecular Reproductive Medicine, Nanjing University, Nanjing 210008, People's Republic of China
| | - Jidong Zhou
- Center for Reproductive Medicine and Obstetrics & Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People's Republic of China; Center for Molecular Reproductive Medicine, Nanjing University, Nanjing 210008, People's Republic of China
| | - Qingqing Shi
- Center for Reproductive Medicine and Obstetrics & Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People's Republic of China; Center for Molecular Reproductive Medicine, Nanjing University, Nanjing 210008, People's Republic of China
| | - Yue Jiang
- Center for Reproductive Medicine and Obstetrics & Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People's Republic of China; Center for Molecular Reproductive Medicine, Nanjing University, Nanjing 210008, People's Republic of China
| | - Nannan Kang
- Center for Reproductive Medicine and Obstetrics & Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People's Republic of China; Center for Molecular Reproductive Medicine, Nanjing University, Nanjing 210008, People's Republic of China
| | - Xin Zhen
- Center for Reproductive Medicine and Obstetrics & Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People's Republic of China; Center for Molecular Reproductive Medicine, Nanjing University, Nanjing 210008, People's Republic of China
| | - Min Wu
- Center for Reproductive Medicine and Obstetrics & Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People's Republic of China; Center for Molecular Reproductive Medicine, Nanjing University, Nanjing 210008, People's Republic of China
| | - Panpan Qiu
- Center for Reproductive Medicine and Obstetrics & Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People's Republic of China; Center for Molecular Reproductive Medicine, Nanjing University, Nanjing 210008, People's Republic of China
| | - Guijun Yan
- Center for Reproductive Medicine and Obstetrics & Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People's Republic of China; State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, People's Republic of China; Center for Molecular Reproductive Medicine, Nanjing University, Nanjing 210008, People's Republic of China
| | - Haixiang Sun
- Center for Reproductive Medicine and Obstetrics & Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People's Republic of China; State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211166, People's Republic of China; Center for Molecular Reproductive Medicine, Nanjing University, Nanjing 210008, People's Republic of China.
| | - Dong Li
- Center for Reproductive Medicine and Obstetrics & Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People's Republic of China; Center for Molecular Reproductive Medicine, Nanjing University, Nanjing 210008, People's Republic of China.
| |
Collapse
|
104
|
Jing J, Feng J, Yuan Y, Guo T, Lei J, Pei F, Ho TV, Chai Y. Spatiotemporal single-cell regulatory atlas reveals neural crest lineage diversification and cellular function during tooth morphogenesis. Nat Commun 2022; 13:4803. [PMID: 35974052 PMCID: PMC9381504 DOI: 10.1038/s41467-022-32490-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 08/02/2022] [Indexed: 11/10/2022] Open
Abstract
Cranial neural crest cells are an evolutionary innovation of vertebrates for craniofacial development and function, yet the mechanisms that govern the cell fate decisions of postmigratory cranial neural crest cells remain largely unknown. Using the mouse molar as a model, we perform single-cell transcriptome profiling to interrogate the cell fate diversification of postmigratory cranial neural crest cells. We reveal the landscape of transcriptional heterogeneity and define the specific cellular domains during the progression of cranial neural crest cell-derived dental lineage diversification, and find that each domain makes a specific contribution to distinct molar mesenchymal tissues. Furthermore, IGF signaling-mediated cell-cell interaction between the cellular domains highlights the pivotal role of autonomous regulation of the dental mesenchyme. Importantly, we reveal cell-type-specific gene regulatory networks in the dental mesenchyme and show that Foxp4 is indispensable for the differentiation of periodontal ligament. Our single-cell atlas provides comprehensive mechanistic insight into the cell fate diversification process of the cranial neural crest cell-derived odontogenic populations.
Collapse
Affiliation(s)
- Junjun Jing
- grid.42505.360000 0001 2156 6853Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033 USA ,grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Chengdu, Sichuan 610041 China
| | - Jifan Feng
- grid.42505.360000 0001 2156 6853Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033 USA
| | - Yuan Yuan
- grid.42505.360000 0001 2156 6853Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033 USA
| | - Tingwei Guo
- grid.42505.360000 0001 2156 6853Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033 USA
| | - Jie Lei
- grid.42505.360000 0001 2156 6853Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033 USA
| | - Fei Pei
- grid.42505.360000 0001 2156 6853Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033 USA
| | - Thach-Vu Ho
- grid.42505.360000 0001 2156 6853Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA 90033 USA
| | - Yang Chai
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
105
|
Choi Y, Luo Y, Lee S, Jin H, Yoon HJ, Hahn Y, Bae J, Lee HH. FOXL2 and FOXA1 cooperatively assemble on the TP53 promoter in alternative dimer configurations. Nucleic Acids Res 2022; 50:8929-8946. [PMID: 35920317 PMCID: PMC9410875 DOI: 10.1093/nar/gkac673] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 07/13/2022] [Accepted: 07/26/2022] [Indexed: 11/16/2022] Open
Abstract
Although both the p53 and forkhead box (FOX) family proteins are key transcription factors associated with cancer progression, their direct relationship is unknown. Here, we found that FOX family proteins bind to the non-canonical homotypic cluster of the p53 promoter region (TP53). Analysis of crystal structures of FOX proteins (FOXL2 and FOXA1) bound to the p53 homotypic cluster indicated that they interact with a 2:1 stoichiometry accommodated by FOX-induced DNA allostery. In particular, FOX proteins exhibited distinct dimerization patterns in recognition of the same p53-DNA; dimer formation of FOXA1 involved protein–protein interaction, but FOXL2 did not. Biochemical and biological functional analyses confirmed the cooperative binding of FOX proteins to the TP53 promoter for the transcriptional activation of TP53. In addition, up-regulation of TP53 was necessary for FOX proteins to exhibit anti-proliferative activity in cancer cells. These analyses reveal the presence of a discrete characteristic within FOX family proteins in which FOX proteins regulate the transcription activity of the p53 tumor suppressor via cooperative binding to the TP53 promoter in alternative dimer configurations.
Collapse
Affiliation(s)
- Yuri Choi
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
| | - Yongyang Luo
- School of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| | - Seunghwa Lee
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea
| | - Hanyong Jin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, Jilin Province, China
| | - Hye-Jin Yoon
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
| | - Yoonsoo Hahn
- Department of Life Science, Chung-Ang University, Seoul 06974, Korea
| | - Jeehyeon Bae
- School of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| | - Hyung Ho Lee
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
106
|
Fibroblast Growth Factors and Cellular Communication Network Factors: Intimate Interplay by the Founding Members in Cartilage. Int J Mol Sci 2022; 23:ijms23158592. [PMID: 35955724 PMCID: PMC9369280 DOI: 10.3390/ijms23158592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/28/2022] [Accepted: 07/28/2022] [Indexed: 02/04/2023] Open
Abstract
Fibroblast growth factors (FGFs) constitute a large family of signaling molecules that act in an autocrine/paracrine, endocrine, or intracrine manner, whereas the cellular communication network factors (CCN) family is composed of six members that manipulate extracellular signaling networks. FGFs and CCNs are structurally and functionally distinct, except for the common characteristics as matricellular proteins. Both play significant roles in the development of a variety of tissues and organs, including the skeletal system. In vertebrates, most of the skeletal parts are formed and grow through a process designated endochondral ossification, in which chondrocytes play the central role. The growth plate cartilage is the place where endochondral ossification occurs, and articular cartilage is left to support the locomotive function of joints. Several FGFs, including FGF-2, one of the founding members of this family, and all of the CCNs represented by CCN2, which is required for proper skeletal development, can be found therein. Research over a decade has revealed direct binding of CCN2 to FGFs and FGF receptors (FGFRs), which occasionally affect the biological outcome via FGF signaling. Moreover, a recent study uncovered an integrated regulation of FGF and CCN genes by FGF signaling. In this review, after a brief introduction of these two families, molecular and genetic interactions between CCN and FGF family members in cartilage, and their biological effects, are summarized. The molecular interplay represents the mutual involvement of the other in their molecular functions, leading to collaboration between CCN2 and FGFs during skeletal development.
Collapse
|
107
|
Liu R, Sun Y, Chen S, Hong Y, Lu Z. FOXD3 and GAB2 as a pair of rivals antagonistically control hepatocellular carcinogenesis. FEBS J 2022; 289:4536-4548. [DOI: 10.1111/febs.16403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 09/05/2021] [Accepted: 02/15/2022] [Indexed: 02/07/2023]
Affiliation(s)
- Ruimin Liu
- School of Pharmaceutical Sciences State Key Laboratory of Cellular Stress Biology Xiamen University Xiamen China
| | - Yan Sun
- School of Pharmaceutical Sciences State Key Laboratory of Cellular Stress Biology Xiamen University Xiamen China
| | - Shuai Chen
- School of Pharmaceutical Sciences State Key Laboratory of Cellular Stress Biology Xiamen University Xiamen China
| | - Yun Hong
- School of Pharmaceutical Sciences State Key Laboratory of Cellular Stress Biology Xiamen University Xiamen China
| | - Zhongxian Lu
- School of Pharmaceutical Sciences State Key Laboratory of Cellular Stress Biology Xiamen University Xiamen China
- Fujian Provincial Key Laboratory of Innovative Drug Target Research School of Pharmaceutical Sciences Xiamen China
| |
Collapse
|
108
|
Chen X, Xu J, Bao W, Li H, Wu W, Liu J, Pi J, Tomlinson B, Chan P, Ruan C, Zhang Q, Zhang L, Fan H, Morrisey E, Liu Z, Zhang Y, Lin L, Liu J, Zhuang T. Endothelial Foxp1 Regulates Neointimal Hyperplasia Via Matrix Metalloproteinase-9/Cyclin Dependent Kinase Inhibitor 1B Signal Pathway. J Am Heart Assoc 2022; 11:e026378. [PMID: 35904197 PMCID: PMC9375493 DOI: 10.1161/jaha.122.026378] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background The endothelium is essential for maintaining vascular physiological homeostasis and the endothelial injury leads to the neointimal hyperplasia because of the excessive proliferation of vascular smooth muscle cells. Endothelial Foxp1 (forkhead box P1) has been shown to control endothelial cell (EC) proliferation and migration in vitro. However, whether EC-Foxp1 participates in neointimal formation in vivo is not clear. Our study aimed to investigate the roles and mechanisms of EC-Foxp1 in neointimal hyperplasia. Methods and Results The wire injury femoral artery neointimal hyperplasia model was performed in Foxp1 EC-specific loss-of-function and gain-of-function mice. EC-Foxp1 deletion mice displayed the increased neointimal formation through elevation of vascular smooth muscle cell proliferation and migration, and the reduction of EC proliferation hence reendothelialization after injury. In contrast, EC-Foxp1 overexpression inhibited the neointimal formation. EC-Foxp1 paracrine regulated vascular smooth muscle cell proliferation and migration via targeting matrix metalloproteinase-9. Also, EC-Foxp1 deletion impaired EC repair through reduction of EC proliferation via increasing cyclin dependent kinase inhibitor 1B expression. Delivery of cyclin dependent kinase inhibitor 1B-siRNA to ECs using RGD (Arg-Gly-Asp)-peptide magnetic nanoparticle normalized the EC-Foxp1 deletion-mediated impaired EC repair and attenuated the neointimal formation. EC-Foxp1 regulates matrix metalloproteinase-9/cyclin dependent kinase inhibitor 1B signaling pathway to control injury induced neointimal formation. Conclusions Our study reveals that targeting EC-Foxp1-matrix metalloproteinase-9/cyclin dependent kinase inhibitor 1B pathway might provide future novel therapeutic interventions for restenosis.
Collapse
Affiliation(s)
- Xiaoli Chen
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Jianfei Xu
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Wenzhen Bao
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Hongda Li
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Wenrun Wu
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Jiwen Liu
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Jingjiang Pi
- Department of CardiologyShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Brian Tomlinson
- Faculty of MedicineMacau University of Science and TechnologyMacauChina
| | - Paul Chan
- Division of CardiologyDepartment of Internal MedicineWan Fang HospitalTaipei Medical UniversityTaipeiTaiwan
| | - Chengchao Ruan
- Department of Physiology and Pathophysiology School of Basic Medical SciencesFudan UniversityShanghaiChina
| | - Qi Zhang
- Department of CardiologyShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Lin Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Huimin Fan
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Edward Morrisey
- Department of Cell and Developmental Biology (R.W., E.E.M.)Department of Medicine (E.E.M.)Penn Cardiovascular Institute (E.E.M.), and Penn Institute for Regenerative Medicine (E.E.M.)University of PennsylvaniaPhiladelphiaPennsylvania
| | - Zhongmin Liu
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Yuzhen Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Li Lin
- Department of CardiologyShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Jie Liu
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Tao Zhuang
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaResearch Center for Translational MedicineShanghai East HospitalTongji University School of MedicineShanghaiChina,Department of Physiology and Pathophysiology School of Basic Medical SciencesFudan UniversityShanghaiChina,Shanghai Jinshan Eye Disease Prevention and Treatment InstituteShanghai Jinshan Nuclear and Chemical Injury Emergency Treatment CenterJinshan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
109
|
Machuka EM, Juma J, Muigai AWT, Amimo JO, Pelle R, Abworo EO. Transcriptome profile of spleen tissues from locally-adapted Kenyan pigs (Sus scrofa) experimentally infected with three varying doses of a highly virulent African swine fever virus genotype IX isolate: Ken12/busia.1 (ken-1033). BMC Genomics 2022; 23:522. [PMID: 35854219 PMCID: PMC9294756 DOI: 10.1186/s12864-022-08754-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 07/08/2022] [Indexed: 11/10/2022] Open
Abstract
Background African swine fever (ASF) is a lethal hemorrhagic disease affecting domestic pigs resulting in up to 100% mortality rates caused by the ASF virus (ASFV). The locally-adapted pigs in South-western Kenya have been reported to be resilient to disease and harsh climatic conditions and tolerate ASF; however, the mechanisms by which this tolerance is sustained remain largely unknown. We evaluated the gene expression patterns in spleen tissues of these locally-adapted pigs in response to varying infective doses of ASFV to elucidate the virus-host interaction dynamics. Methods Locally adapted pigs (n = 14) were experimentally infected with a high dose (1x106HAD50), medium dose (1x104HAD50), and low dose (1x102HAD50) of the highly virulent genotype IX ASFV Ken12/busia.1 (Ken-1033) isolate diluted in PBS and followed through the course of infection for 29 days. The in vivo pig host and ASFV pathogen gene expression in spleen tissues from 10 pigs (including three from each infective group and one uninfected control) were analyzed in a dual-RNASeq fashion. We compared gene expression between three varying doses in the host and pathogen by contrasting experiment groups against the naïve control. Results A total of 4954 differentially expressed genes (DEGs) were detected after ASFV Ken12/1 infection, including 3055, 1771, and 128 DEGs in the high, medium, and low doses, respectively. Gene ontology and KEGG pathway analysis showed that the DEGs were enriched for genes involved in the innate immune response, inflammatory response, autophagy, and apoptosis in lethal dose groups. The surviving low dose group suppressed genes in pathways of physiopathological importance. We found a strong association between severe ASF pathogenesis in the high and medium dose groups with upregulation of proinflammatory cytokines and immunomodulation of cytokine expression possibly induced by overproduction of prostaglandin E synthase (4-fold; p < 0.05) or through downregulation of expression of M1-activating receptors, signal transductors, and transcription factors. The host-pathogen interaction resulted in induction of expression of immune-suppressive cytokines (IL-27), inactivation of autophagy and apoptosis through up-regulation of NUPR1 [5.7-fold (high dose) and 5.1-fold (medium dose) [p < 0.05] and IL7R expression. We detected repression of genes involved in MHC class II antigen processing and presentation, such as cathepsins, SLA-DQB1, SLA-DOB, SLA-DMB, SLA-DRA, and SLA-DQA in the medium and high dose groups. Additionally, the host-pathogen interaction activated the CD8+ cytotoxicity and neutrophil machinery by increasing the expression of neutrophils/CD8+ T effector cell-recruiting chemokines (CCL2, CXCL2, CXCL10, CCL23, CCL4, CXCL8, and CXCL13) in the lethal high and medium dose groups. The recovered pigs infected with ASFV at a low dose significantly repressed the expression of CXCL10, averting induction of T lymphocyte apoptosis and FUNDC1 that suppressed neutrophilia. Conclusions We provide the first in vivo gene expression profile data from locally-adapted pigs from south-western Kenya following experimental infection with a highly virulent ASFV genotype IX isolate at varying doses that mimic acute and mild disease. Our study showed that the locally-adapted pigs induced the expression of genes associated with tolerance to infection and repression of genes involved in inflammation at varying levels depending upon the ASFV dose administered. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08754-8.
Collapse
Affiliation(s)
- Eunice Magoma Machuka
- Animal and Human Health Program, International Livestock Research Institute (ILRI), P.O. Box 30709-00100, Nairobi, Kenya. .,Pan African University Institute for Basic Sciences Technology and Innovation (PAUSTI), P.O Box 62000-00200, Nairobi, Kenya.
| | - John Juma
- Animal and Human Health Program, International Livestock Research Institute (ILRI), P.O. Box 30709-00100, Nairobi, Kenya
| | | | - Joshua Oluoch Amimo
- Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Avenue, Wooster, OH, 44691, USA
| | - Roger Pelle
- Biosciences eastern and central Africa, International Livestock Research Institute (BecA-ILRI) Hub, P.O. Box 30709-00100, Nairobi, Kenya.
| | - Edward Okoth Abworo
- Animal and Human Health Program, International Livestock Research Institute (ILRI), P.O. Box 30709-00100, Nairobi, Kenya
| |
Collapse
|
110
|
Yu M, Yu H, Mu N, Wang Y, Ma H, Yu L. The Function of FoxK Transcription Factors in Diseases. Front Physiol 2022; 13:928625. [PMID: 35903069 PMCID: PMC9314541 DOI: 10.3389/fphys.2022.928625] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
Forkhead box (FOX) transcription factors play a crucial role in the regulation of many diseases, being an evolutionarily conserved superfamily of transcription factors. In recent years, FoxK1/2, members of its family, has been the subject of research. Even though FoxK1 and FoxK2 have some functional overlap, increasing evidence indicates that the regulatory functions of FoxK1 and FoxK2 are not the same in various physiological and disease states. It is important to understand the biological function and mechanism of FoxK1/2 for better understanding pathogenesis of diseases, predicting prognosis, and finding new therapeutic targets. There is, however, a lack of comprehensive and systematic analysis of the similarities and differences of FoxK1/2 roles in disease, prompting us to perform a literature review.
Collapse
Affiliation(s)
- Mujun Yu
- School of Life Sciences, Yan'an University, Yan'an, China
| | - Haozhen Yu
- School of Basic Medical Sciences, Shaanxi University of Traditional Chinese Medicine, Xianyang, China
| | - Nan Mu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Yishi Wang
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Heng Ma
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Lu Yu
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
111
|
CDI Exerts Anti-Tumor Effects by Blocking the FoxM1-DNA Interaction. Biomedicines 2022; 10:biomedicines10071671. [PMID: 35884976 PMCID: PMC9313426 DOI: 10.3390/biomedicines10071671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/30/2022] [Accepted: 07/06/2022] [Indexed: 12/03/2022] Open
Abstract
The Forkhead box protein M1 (FoxM1) is an appealing target for anti-cancer therapeutics as this cell proliferation-associated transcription factor is overexpressed in most human cancers. FoxM1 is involved in tumor invasion, angiogenesis, and metastasis. To discover novel inhibitors that disrupt the FoxM1-DNA interaction, we identified CDI, a small molecule that inhibits the FoxM1–DNA interaction. CDI was identified through an assay based on the time-resolved fluorescence energy transfer response of a labeled consensus oligonucleotide that was bound to a recombinant FoxM1-dsDNA binding domain (FoxM1-DBD) protein and exhibited potent inhibitory activity against FoxM1-DNA interaction. CDI suppressed cell proliferation and induced apoptosis in MDA-MB-231 cells obtained from a breast cancer patient. Furthermore, it decreased not only the mRNA and protein expression of FoxM1 but also that of downstream targets such as CDC25b. Additionally, global transcript profiling of MDA-MB-231 cells by RNA-Seq showed that CDI decreases the expression of FoxM1-regulated genes. The docking and MD simulation results indicated that CDI likely binds to the DNA interaction site of FoxM1-DBD and inhibits the function of FoxM1-DBD. These results of CDI being a possible effective inhibitor of FoxM1-DNA interaction will encourage its usage in pharmaceutical applications.
Collapse
|
112
|
Geng G, Li Q, Guo X, Ni Q, Xu Y, Ma Z, Wang Y, Ming M. FOXO3a‑modulated DEPDC1 promotes malignant progression of nephroblastoma via the Wnt/β‑catenin signaling pathway. Mol Med Rep 2022; 26:272. [PMID: 35795985 PMCID: PMC9309542 DOI: 10.3892/mmr.2022.12788] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/27/2022] [Indexed: 11/22/2022] Open
Abstract
DEP domain containing 1 (DEPDC1) and forkhead box transcription factor 3a (FOXO3a) serve a role in tumor cells. To the best of our knowledge, however, the expression of DEPDC1 and FOXO3a in nephroblastoma and their role and potential mechanisms in nephroblastoma cells have not been reported. The aim of the present study was to characterize the expression of DEPDC1 and FOXO3a in nephroblastoma, as well as the underlying mechanisms. The expression levels of DEPDC1 and FOXO3a were detected using reverse transcription-quantitative PCR and western blotting. Cell viability, proliferation, invasion and migration were detected using Cell Counting Kit-8, colony formation, Transwell and wound healing assays, respectively. The activity of DEPDC1 promoter was detected by dual-luciferase reporter assay and the association between FOXO3a and DEPDC1 was detected using immunoprecipitation. DEPDC1 expression was significantly increased in nephroblastoma cells, particularly WiT49 cells. Compared with the negative control, DEPDC1 knockdown significantly inhibited proliferation, invasion and migration of WiT49 cells, while DEPDC1 overexpression (Ov) reversed these effects. By contrast, expression of FOXO3a was decreased in WiT49 cells and immunoprecipitation showed that FOXO3a bound to the DEPDC1 promoter. Ov-FOXO3a inhibited WiT49 cell proliferation, invasion and migration, as well as protein expression levels of phosphorylated-glycogen synthase kinase-3β, Wnt3a and β-catenin, while DEPDC1 Ov reversed the inhibitory effects of FOXO3a Ov on WiT49 cells. In conclusion, DEPDC1 promoted malignant progression of nephroblastoma via the Wnt/β-catenin signaling pathway; this may be regulated by FOXO3a.
Collapse
Affiliation(s)
- Geng Geng
- Department of Pediatric Surgery, Taian City Central Hospital, Taian, Shandong 271000, P.R. China
| | - Qinghao Li
- Department of Pediatric Surgery, Taian City Central Hospital, Taian, Shandong 271000, P.R. China
| | - Xingqing Guo
- Department of Pediatric Respirology and Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, P.R. China
| | - Qingbin Ni
- Department of Pediatric Surgery, Taian City Central Hospital, Taian, Shandong 271000, P.R. China
| | - Yongtao Xu
- Department of Pediatric Surgery, Taian City Central Hospital, Taian, Shandong 271000, P.R. China
| | - Zhaolong Ma
- Department of Pediatric Surgery, Taian City Central Hospital, Taian, Shandong 271000, P.R. China
| | - Yongjin Wang
- Department of Pediatric Surgery, Taian City Central Hospital, Taian, Shandong 271000, P.R. China
| | - Ming Ming
- Department of Pediatric Surgery, Taian City Central Hospital, Taian, Shandong 271000, P.R. China
| |
Collapse
|
113
|
Udoh UAS, Banerjee M, Rajan PK, Sanabria JD, Smith G, Schade M, Sanabria JA, Nakafuku Y, Sodhi K, Pierre SV, Shapiro JI, Sanabria JR. Tumor-Suppressor Role of the α1-Na/K-ATPase Signalosome in NASH Related Hepatocellular Carcinoma †. Int J Mol Sci 2022; 23:ijms23137359. [PMID: 35806364 PMCID: PMC9266688 DOI: 10.3390/ijms23137359] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related mortality worldwide, with an estimate of 0.84 million cases every year. In Western countries, because of the obesity epidemic, non-alcoholic steatohepatitis (NASH) has become the major cause of HCC. Intriguingly, the molecular mechanisms underlying tumorigenesis of HCC from NASH are largely unknown. We hypothesized that the growing uncoupled metabolism during NASH progression to HCC, manifested by lower cell redox status and an apoptotic ‘switch’ activity, follows a dysregulation of α1-Na/K-ATPase (NKA)/Src signalosome. Our results suggested that in NASH-related malignancy, α1-NKA signaling causes upregulation of the anti-apoptotic protein survivin and downregulation of the pro-apoptotic protein Smac/DIABLO via the activation of the PI3K → Akt pro-survival pathway with concomitant inhibition of the FoxO3 circuit, favoring cell division and primary liver carcinogenesis. Signalosome normalization using an inhibitory peptide resets apoptotic activity in malignant cells, with a significant decrease in tumor burden in vivo. Therefore, α1-NKA signalosome exercises in HCC the characteristic of a tumor suppressor, suggesting α1-NKA as a putative target for clinical therapy.
Collapse
Affiliation(s)
- Utibe-Abasi S. Udoh
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Moumita Banerjee
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Pradeep K. Rajan
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Juan D. Sanabria
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Gary Smith
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Mathew Schade
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Jacqueline A. Sanabria
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Yuto Nakafuku
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Komal Sodhi
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Sandrine V. Pierre
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Joseph I. Shapiro
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
| | - Juan R. Sanabria
- Department of Surgery, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25701, USA; (U.-A.S.U.); (M.B.); (P.K.R.); (J.D.S.); (G.S.); (M.S.); (J.A.S.); (Y.N.); (K.S.); (J.I.S.)
- Marshall Institute for Interdisciplinary Research, Marshall University Joan C. Edwards School of Medicine, Huntington, WV 25703, USA;
- Department of Nutrition and Metabolomic Core Facility, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Correspondence: or
| |
Collapse
|
114
|
Donmez C, Konac E. Silencing effects of FOXD1 inhibit metastatic potentials of the PCa via N-cadherin - Wnt/β-catenin crosstalk. Gene 2022; 836:146680. [PMID: 35738443 DOI: 10.1016/j.gene.2022.146680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/03/2022] [Accepted: 06/10/2022] [Indexed: 11/18/2022]
Abstract
The elucidation of the mechanisms controlling the metastatic processes is important for the development of new treatment methods to prevent the progression of localized disease to metastasis. Forkhead box D1 (FOXD1) is a member of the FOX transcription factor family and has been reported to play an important role in the development and progression of various cancers. However, its role in prostate cancer (PCa) remains only partially understood. Therefore, we aimed to explore the effects on the associated regulatory signal pathway of FOXD1 in prostate cancer. To clarify the roles of FOXD1 in prostate cancer, we used siRNA to suppress its expression in 22Rv1 cells with relatively higher expression of FOXD1. The effects of FOXD1 silencing on cell proliferation, migration and invasion were determined. WST-1 assays were used to determine cell proliferation. Cell migration and invasion were evaluated through wound healing and transwell assays. The possible underlying mechanism of FOXD1 silencing on 22Rv1 was evaluated by determining the expression of proteins related to EMT and Wnt/β-catenin signaling pathway. Our results showed that FOXD1 was highly expressed in prostate cancer cell lines -PC-3, DU145, LNCaP and 22Rv1- compared to normal prostate epithelial cell line RWPE-1. Additionally, silencing of FOXD1 significantly reduced proliferation, migration and invasion of 22Rv1 cells. Furthermore, silencing of FOXD1 decreased the expression of β-catenin and cyclin D1, which are involved in the Wnt/β-catenin signaling pathway. However, it did not appear to affect the expression of EMT-related proteins other than N-cadherin. Our results suggest that silencing of FOXD1 suppresses metastatic potentials of the PCa via N-cadherin - Wnt/β-catenin crosstalk. Therefore, the expression status of FOXD1 may be a new prognostic factor as well as a potential therapeutic target in prostate cancer treatment.
Collapse
Affiliation(s)
- Cigdem Donmez
- Department of Medical Biology and Genetics, Faculty of Medicine, Gazi University, Besevler, 06500 Ankara, Turkey; Department of Medical Biology, Faculty of Medicine, Zonguldak Bulent Ecevit University, Esenkoy, Kozlu, 67600 Zonguldak, Turkey
| | - Ece Konac
- Department of Medical Biology and Genetics, Faculty of Medicine, Gazi University, Besevler, 06500 Ankara, Turkey.
| |
Collapse
|
115
|
He M, Cao C, Ni Z, Liu Y, Song P, Hao S, He Y, Sun X, Rao Y. PROTACs: great opportunities for academia and industry (an update from 2020 to 2021). Signal Transduct Target Ther 2022; 7:181. [PMID: 35680848 PMCID: PMC9178337 DOI: 10.1038/s41392-022-00999-9] [Citation(s) in RCA: 97] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/25/2022] [Accepted: 04/12/2022] [Indexed: 02/07/2023] Open
Abstract
PROteolysis TArgeting Chimeras (PROTACs) technology is a new protein-degradation strategy that has emerged in recent years. It uses bifunctional small molecules to induce the ubiquitination and degradation of target proteins through the ubiquitin-proteasome system. PROTACs can not only be used as potential clinical treatments for diseases such as cancer, immune disorders, viral infections, and neurodegenerative diseases, but also provide unique chemical knockdown tools for biological research in a catalytic, reversible, and rapid manner. In 2019, our group published a review article "PROTACs: great opportunities for academia and industry" in the journal, summarizing the representative compounds of PROTACs reported before the end of 2019. In the past 2 years, the entire field of protein degradation has experienced rapid development, including not only a large increase in the number of research papers on protein-degradation technology but also a rapid increase in the number of small-molecule degraders that have entered the clinical and will enter the clinical stage. In addition to PROTAC and molecular glue technology, other new degradation technologies are also developing rapidly. In this article, we mainly summarize and review the representative PROTACs of related targets published in 2020-2021 to present to researchers the exciting developments in the field of protein degradation. The problems that need to be solved in this field will also be briefly introduced.
Collapse
Affiliation(s)
- Ming He
- Ministry of Education (MOE) Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, 100084, Beijing, P. R. China
| | - Chaoguo Cao
- Ministry of Education (MOE) Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, 100084, Beijing, P. R. China
- Tsinghua-Peking Center for Life Sciences, 100084, Beijing, P. R. China
| | - Zhihao Ni
- Ministry of Education (MOE) Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, 100084, Beijing, P. R. China
| | - Yongbo Liu
- Ministry of Education (MOE) Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, 100084, Beijing, P. R. China
| | - Peilu Song
- Ministry of Education (MOE) Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, 100084, Beijing, P. R. China
| | - Shuang Hao
- Ministry of Education (MOE) Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, 100084, Beijing, P. R. China
| | - Yuna He
- Ministry of Education (MOE) Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, 100084, Beijing, P. R. China
| | - Xiuyun Sun
- Ministry of Education (MOE) Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, 100084, Beijing, P. R. China
| | - Yu Rao
- Ministry of Education (MOE) Key Laboratory of Protein Sciences, School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, 100084, Beijing, P. R. China.
- School of Pharmaceutical Sciences, Zhengzhou University, 450001, Zhengzhou, China.
| |
Collapse
|
116
|
Foxf2 represses bone formation via Wnt2b/β-catenin signaling. Exp Mol Med 2022; 54:753-764. [PMID: 35668101 PMCID: PMC9256714 DOI: 10.1038/s12276-022-00779-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 01/25/2022] [Accepted: 02/20/2022] [Indexed: 11/08/2022] Open
Abstract
Differentiation of mesenchymal stem cells (MSCs) into osteoblasts is a critical process for proper skeletal development and acquisition/maintenance of bone mass. However, since this regulatory mechanism has not yet been fully elucidated, the treatment of severe osteoporosis and fractures is a challenge. Here, through a comprehensive analysis of gene expression during the differentiation of MSCs into osteoblasts, we show that the forkhead transcription factor Foxf2 is a crucial regulator of this process. Foxf2 expression transiently increased during MSC osteoblastic differentiation. Overexpression of Foxf2 in MSCs inhibited osteoblastic differentiation, and conversely, knockdown of Foxf2 expression promoted this process. Osteoprogenitor-specific Foxf2 knockout mice developed a high bone mass phenotype due to increased bone formation. RNA-seq analysis and molecular experiments revealed that Foxf2 regulation of bone formation is mediated by Wnt2b. Knockdown of Foxf2 in mouse femurs enhanced bone regeneration in vivo. FOXF2 expression was correlated with hip bone mineral density in postmenopausal women with low bone mass. Finally, inhibition of FOXF2 promoted osteoblastic differentiation of human MSCs. This study uncovers a critical role of Foxf2 in the differentiation of MSCs into osteoblasts and provides insight into the pathogenesis associated with bone-related diseases such as osteoporosis and nonunion after fracture.
Collapse
|
117
|
Agemura T, Hasegawa T, Yari S, Kikuta J, Ishii M. Arthritis-associated osteoclastogenic macrophage, AtoM, as a key player in pathological bone erosion. Inflamm Regen 2022; 42:17. [PMID: 35650653 PMCID: PMC9161570 DOI: 10.1186/s41232-022-00206-w] [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: 02/11/2022] [Accepted: 03/11/2022] [Indexed: 11/10/2022] Open
Abstract
Osteoclasts are myeloid lineage cells with a unique bone-destroying ability that maintains bone homeostasis together with bone formation by osteoblasts. An advanced intravital imaging system using a two-photon microscopy has enabled the observation and evaluation of osteoclast dynamics and behaviors in the bone marrow of living mice. Using this system, it has become clear that pathological osteoclasts under inflamed conditions differ from physiological osteoclasts under a steady-state. Recently, we identified novel osteoclast precursors in arthritis, called arthritis-associated osteoclastogenic macrophages (AtoMs), which differentiate into pathological osteoclasts and induce inflammatory bone destruction. In this review, we introduce the in vivo imaging of physiological and pathological osteoclasts and their differentiation mechanism.
Collapse
Affiliation(s)
- Tomoya Agemura
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Tetsuo Hasegawa
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Shinya Yari
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Junichi Kikuta
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan.,WPI-Immunology Frontier Research Center, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan.,Laboratory of Bioimaging and Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Asagi Saito, Osaka, Ibaraki, 567-0085, Japan
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan. .,WPI-Immunology Frontier Research Center, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan. .,Laboratory of Bioimaging and Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Asagi Saito, Osaka, Ibaraki, 567-0085, Japan.
| |
Collapse
|
118
|
Xu Q, Xu JL, Chen WQ, Xu WX, Song YX, Tang WJ, Xu D, Jiang MP, Tang J. Roles and mechanisms of miR-195-5p in human solid cancers. Biomed Pharmacother 2022; 150:112885. [PMID: 35453003 DOI: 10.1016/j.biopha.2022.112885] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/20/2022] [Accepted: 03/23/2022] [Indexed: 11/02/2022] Open
Abstract
Cancer persists as a worldwide disease that contributes to high morbidity and mortality rates. As a class of non-coding RNA, microRNAs (miRNAs) are one kind of important regulators in cancer and frequently implicated in tumor development and progression. Emerging experiments have suggested that miRNA-195-5p (miR-195-5p) can regulate neoplastic processes in many pathways. For instance, miR-195-5p can not only regulate proliferation, migration and invasion of tumor cells but also promote tumor cell apoptosis. Furthermore, low expression of miR-195-5p could induce drug resistance. Our review focuses on the expression of miR-195-5p in various tumors and elucidates the related mechanisms of which miR-195-5p participates in tumor biology, as well as summarizes the roles of miR-195-5p in tumor progression. We believe that miR-195-5p might have potential utility as a novel diagnostic biomarker and therapeutic target for cancer.
Collapse
Affiliation(s)
- Qi Xu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Jia-Lin Xu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Wen-Quan Chen
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Wen-Xiu Xu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Yu-Xin Song
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Wen-Juan Tang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Di Xu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Meng-Ping Jiang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Jinhai Tang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China.
| |
Collapse
|
119
|
Inci N, Kamali D, Akyildiz EO, Tahir Turanli E, Bozaykut P. Translation of Cellular Senescence to Novel Therapeutics: Insights From Alternative Tools and Models. FRONTIERS IN AGING 2022; 3:828058. [PMID: 35821852 PMCID: PMC9261353 DOI: 10.3389/fragi.2022.828058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 04/12/2022] [Indexed: 01/10/2023]
Abstract
Increasing chronological age is the greatest risk factor for human diseases. Cellular senescence (CS), which is characterized by permanent cell-cycle arrest, has recently emerged as a fundamental mechanism in developing aging-related pathologies. During the aging process, senescent cell accumulation results in senescence-associated secretory phenotype (SASP) which plays an essential role in tissue dysfunction. Although discovered very recently, senotherapeutic drugs have been already involved in clinical studies. This review gives a summary of the molecular mechanisms of CS and its role particularly in the development of cardiovascular diseases (CVD) as the leading cause of death. In addition, it addresses alternative research tools including the nonhuman and human models as well as computational techniques for the discovery of novel therapies. Finally, senotherapeutic approaches that are mainly classified as senolytics and senomorphics are discussed.
Collapse
Affiliation(s)
- Nurcan Inci
- Graduate School of Natural and Applied Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Dilanur Kamali
- Graduate School of Natural and Applied Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Erdogan Oguzhan Akyildiz
- Graduate School of Natural and Applied Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Eda Tahir Turanli
- Graduate School of Natural and Applied Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Perinur Bozaykut
- Graduate School of Natural and Applied Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| |
Collapse
|
120
|
Yang F, Xiao Z, Zhang S. FOXP2 regulates thyroid cancer cell proliferation and apoptosis via transcriptional activation of RPS6KA6. Exp Ther Med 2022; 23:434. [PMID: 35607372 PMCID: PMC9121208 DOI: 10.3892/etm.2022.11361] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 01/04/2022] [Indexed: 11/06/2022] Open
Abstract
The transcription factor, forkhead box P2 (FOXP2) has tumor-suppressive effects in several types of cancer. However, the regulatory role and underlying mechanism of FOXP2 in thyroid cancer (THCA) is not completely understood. In the present study, the mRNA expression levels of FOXP2 and ribosomal protein S6 kinase A6 (RPS6KA6) were evaluated using the GEPIA database and THCA cell lines. The association between FOXP2 and RPS6KA6 was analyzed using the LinkedOmics, and GEPIA databases. Then, the binding sites of FOXP2 and the RPS6KA6 promotor was predicted using the JASPAR database, and verified using a dual-luciferase reporter assay and chromatin immunoprecipitation. In addition, functional assays investigating FOXP2 and RPS6KA6 were conducted in the TPC-1 cell line. The data showed that FOXP2 and RPS6KA6 mRNA expression levels were decreased in the THCA tissues, and cell lines. Overexpression of FOXP2 inhibited cell proliferation and promoted apoptosis in the THCA cell lines. Furthermore, RPS6KA6 mRNA expression levels were reduced in THCA and were correlated with FOXP2 expression level. Mechanistic studies revealed that FOXP2 binds directly to the promotor region of RPS6KA6 and modulated the expression level of RPS6KA6 transcriptionally. In addition, rescue experiments showed that knockdown of RPS6KA6 expression reversed the effects of FOXP2 overexpression on THCA cell proliferation and apoptosis, and the regulation of FOXP2/RPS6KA6 may be associated with the PI3K/AKT pathway. In summary, FOXP2 was associated with the proliferation and apoptosis of human THCA cells via the transcriptional activation of RPS6KA6. The FOXP2/RPS6KA6 axis could be a promising target for the treatment of THCA.
Collapse
Affiliation(s)
- Feibiao Yang
- Department of Thyroid and Breast Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
| | - Zhangsheng Xiao
- Department of Thyroid and Breast Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
| | - Songze Zhang
- Department of Thyroid and Breast Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
| |
Collapse
|
121
|
Xu T, Jiang Y, Yuan S, Zhang L, Chen X, Zhao W, Cai L, Xiao B, Jia L. Andrographolide Inhibits ER-Positive Breast Cancer Growth and Enhances Fulvestrant Efficacy via ROS-FOXM1-ER-α Axis. Front Oncol 2022; 12:899402. [PMID: 35615146 PMCID: PMC9124841 DOI: 10.3389/fonc.2022.899402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/13/2022] [Indexed: 12/02/2022] Open
Abstract
Estrogen receptor (ER)-positive breast cancer is the main subtype of breast cancer (BRCA) with high incidence and mortality. Andrographolide (AD), a major active component derived from the traditional Chinese medicine Andrographis paniculate, has substantial anti-cancer effect in various tumors. However, the antitumor efficacy and the underlying molecular mechanisms of AD on ER-positive breast cancer are poorly understood. In the present study, we demonstrated that andrographolide (AD) significantly inhibited the growth of ER-positive breast cancer cells. Mechanistically, AD suppressed estrogen receptor 1 (ESR1, encodes ER-α) transcription to inhibit tumor growth. Further studies revealed that AD induced ROS production to down-regulate FOXM1-ER-α axis. Conversely, inhibiting ROS production with N-acetylcysteine (NAC) elevated AD-decreased ER-α expression, which could be alleviated by FOXM1 knockdown. In addition, AD in combination with fulvestrant (FUL) synergistically down-regulated ER-α expression to inhibit ER-positive breast cancer both in vitro and in vivo. These findings collectively indicate that AD suppresses ESR1 transcription through ROS-FOXM1 axis to inhibit ER-positive breast cancer growth and suggest that AD might be a potential therapeutic agent and fulvestrant sensitizer for ER-positive breast cancer treatment.
Collapse
|
122
|
Bévant K, Desoteux M, Angenard G, Pineau R, Caruso S, Louis C, Papoutsoglou P, Sulpice L, Gilot D, Zucman-Rossi J, Coulouarn C. TGFβ-induced FOXS1 controls epithelial-mesenchymal transition and predicts a poor prognosis in liver cancer. Hepatol Commun 2022; 6:1157-1171. [PMID: 34825776 PMCID: PMC9035581 DOI: 10.1002/hep4.1866] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/12/2021] [Accepted: 10/26/2021] [Indexed: 01/13/2023] Open
Abstract
Transforming growth factor beta (TGF-β) plays a key role in tumor progression, notably as a potent inducer of epithelial-mesenchymal transition (EMT). However, all of the molecular effectors driving TGFβ-induced EMT are not fully characterized. Here, we report that forkhead box S1 (FOXS1) is a SMAD (mothers against decapentaplegic)-dependent TGFβ-induced transcription factor, which regulates the expression of genes required for the initial steps of EMT (e.g., snail family transcription repressor 1) and to maintain a mesenchymal phenotype in hepatocellular carcinoma (HCC) cells. In human HCC, we report that FOXS1 is a biomarker of poorly differentiated and aggressive tumor subtypes. Importantly, FOXS1 expression level and activity are associated with a poor prognosis (e.g., reduced patient survival), not only in HCC but also in colon, stomach, and kidney cancers. Conclusion: FOXS1 constitutes a clinically relevant biomarker for tumors in which the pro-metastatic arm of TGF-β is active (i.e., patients who may benefit from targeted therapies using inhibitors of the TGF-β pathway).
Collapse
Affiliation(s)
- Kevin Bévant
- InsermUniv RennesUMR_S 1242ChemistryOncogenesis, Stress SignalingCentre de Lutte contre le Cancer Eugène MarquisService de Chirurgie Hépatobiliaire et DigestiveCHU RennesRennesFrance.,InsermUniv RennesInraeUMR_S 1241NuMeCan (Nutrition, Metabolisms and Cancer)RennesFrance
| | - Matthis Desoteux
- InsermUniv RennesUMR_S 1242ChemistryOncogenesis, Stress SignalingCentre de Lutte contre le Cancer Eugène MarquisService de Chirurgie Hépatobiliaire et DigestiveCHU RennesRennesFrance.,InsermUniv RennesInraeUMR_S 1241NuMeCan (Nutrition, Metabolisms and Cancer)RennesFrance
| | - Gaëlle Angenard
- InsermUniv RennesInraeUMR_S 1241NuMeCan (Nutrition, Metabolisms and Cancer)RennesFrance
| | - Raphaël Pineau
- InsermUniv RennesUMR_S 1242ChemistryOncogenesis, Stress SignalingCentre de Lutte contre le Cancer Eugène MarquisService de Chirurgie Hépatobiliaire et DigestiveCHU RennesRennesFrance
| | - Stefano Caruso
- Centre de Recherche des CordeliersInsermSorbonne UniversitéUniversité de ParisUniversité Paris 13Functional Genomics of Solid Tumors LaboratoryParisFrance
| | - Corentin Louis
- InsermUniv RennesUMR_S 1242ChemistryOncogenesis, Stress SignalingCentre de Lutte contre le Cancer Eugène MarquisService de Chirurgie Hépatobiliaire et DigestiveCHU RennesRennesFrance.,InsermUniv RennesInraeUMR_S 1241NuMeCan (Nutrition, Metabolisms and Cancer)RennesFrance
| | - Panagiotis Papoutsoglou
- InsermUniv RennesUMR_S 1242ChemistryOncogenesis, Stress SignalingCentre de Lutte contre le Cancer Eugène MarquisService de Chirurgie Hépatobiliaire et DigestiveCHU RennesRennesFrance.,InsermUniv RennesInraeUMR_S 1241NuMeCan (Nutrition, Metabolisms and Cancer)RennesFrance
| | - Laurent Sulpice
- InsermUniv RennesUMR_S 1242ChemistryOncogenesis, Stress SignalingCentre de Lutte contre le Cancer Eugène MarquisService de Chirurgie Hépatobiliaire et DigestiveCHU RennesRennesFrance.,InsermUniv RennesInraeUMR_S 1241NuMeCan (Nutrition, Metabolisms and Cancer)RennesFrance
| | - David Gilot
- InsermUniv RennesUMR_S 1242ChemistryOncogenesis, Stress SignalingCentre de Lutte contre le Cancer Eugène MarquisService de Chirurgie Hépatobiliaire et DigestiveCHU RennesRennesFrance
| | - Jessica Zucman-Rossi
- Centre de Recherche des CordeliersInsermSorbonne UniversitéUniversité de ParisUniversité Paris 13Functional Genomics of Solid Tumors LaboratoryParisFrance.,European Hospital Georges PompidouAP-HPParisFrance
| | - Cédric Coulouarn
- InsermUniv RennesUMR_S 1242ChemistryOncogenesis, Stress SignalingCentre de Lutte contre le Cancer Eugène MarquisService de Chirurgie Hépatobiliaire et DigestiveCHU RennesRennesFrance.,InsermUniv RennesInraeUMR_S 1241NuMeCan (Nutrition, Metabolisms and Cancer)RennesFrance
| |
Collapse
|
123
|
Vereczkei A, Barta C, Magi A, Farkas J, Eisinger A, Király O, Belik A, Griffiths MD, Szekely A, Sasvári-Székely M, Urbán R, Potenza MN, Badgaiyan RD, Blum K, Demetrovics Z, Kotyuk E. FOXN3 and GDNF Polymorphisms as Common Genetic Factors of Substance Use and Addictive Behaviors. J Pers Med 2022; 12:jpm12050690. [PMID: 35629112 PMCID: PMC9144496 DOI: 10.3390/jpm12050690] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 12/15/2022] Open
Abstract
Epidemiological and phenomenological studies suggest shared underpinnings between multiple addictive behaviors. The present genetic association study was conducted as part of the Psychological and Genetic Factors of Addictions study (n = 3003) and aimed to investigate genetic overlaps between different substance use, addictive, and other compulsive behaviors. Association analyses targeted 32 single-nucleotide polymorphisms, potentially addictive substances (alcohol, tobacco, cannabis, and other drugs), and potentially addictive or compulsive behaviors (internet use, gaming, social networking site use, gambling, exercise, hair-pulling, and eating). Analyses revealed 29 nominally significant associations, from which, nine survived an FDRbl correction. Four associations were observed between FOXN3 rs759364 and potentially addictive behaviors: rs759364 showed an association with the frequency of alcohol consumption and mean scores of scales assessing internet addiction, gaming disorder, and exercise addiction. Significant associations were found between GDNF rs1549250, rs2973033, CNR1 rs806380, DRD2/ANKK1 rs1800497 variants, and the “lifetime other drugs” variable. These suggested that genetic factors may contribute similarly to specific substance use and addictive behaviors. Specifically, FOXN3 rs759364 and GDNF rs1549250 and rs2973033 may constitute genetic risk factors for multiple addictive behaviors. Due to limitations (e.g., convenience sampling, lack of structured scales for substance use), further studies are needed. Functional correlates and mechanisms underlying these relationships should also be investigated.
Collapse
Affiliation(s)
- Andrea Vereczkei
- Department of Molecular Biology, Institute of Biochemistry and Molecular Biology, Semmelweis University, 1094 Budapest, Hungary; (A.V.); (A.B.); (M.S.-S.)
| | - Csaba Barta
- Department of Molecular Biology, Institute of Biochemistry and Molecular Biology, Semmelweis University, 1094 Budapest, Hungary; (A.V.); (A.B.); (M.S.-S.)
- Correspondence: (C.B.); (Z.D.)
| | - Anna Magi
- Institute of Psychology, ELTE Eötvös Loránd University, 1075 Budapest, Hungary; (A.M.); (J.F.); (A.E.); (O.K.); (A.S.); (R.U.); (E.K.)
- Doctoral School of Psychology, ELTE Eötvös Loránd University, 1075 Budapest, Hungary
| | - Judit Farkas
- Institute of Psychology, ELTE Eötvös Loránd University, 1075 Budapest, Hungary; (A.M.); (J.F.); (A.E.); (O.K.); (A.S.); (R.U.); (E.K.)
- Nyírő Gyula National Institute of Psychiatry and Addictions, 1135 Budapest, Hungary
| | - Andrea Eisinger
- Institute of Psychology, ELTE Eötvös Loránd University, 1075 Budapest, Hungary; (A.M.); (J.F.); (A.E.); (O.K.); (A.S.); (R.U.); (E.K.)
- Doctoral School of Psychology, ELTE Eötvös Loránd University, 1075 Budapest, Hungary
| | - Orsolya Király
- Institute of Psychology, ELTE Eötvös Loránd University, 1075 Budapest, Hungary; (A.M.); (J.F.); (A.E.); (O.K.); (A.S.); (R.U.); (E.K.)
| | - Andrea Belik
- Department of Molecular Biology, Institute of Biochemistry and Molecular Biology, Semmelweis University, 1094 Budapest, Hungary; (A.V.); (A.B.); (M.S.-S.)
| | - Mark D. Griffiths
- International Gaming Research Unit, Psychology Department, Nottingham Trent University, Nottingham NG1 4FQ, UK;
| | - Anna Szekely
- Institute of Psychology, ELTE Eötvös Loránd University, 1075 Budapest, Hungary; (A.M.); (J.F.); (A.E.); (O.K.); (A.S.); (R.U.); (E.K.)
| | - Mária Sasvári-Székely
- Department of Molecular Biology, Institute of Biochemistry and Molecular Biology, Semmelweis University, 1094 Budapest, Hungary; (A.V.); (A.B.); (M.S.-S.)
| | - Róbert Urbán
- Institute of Psychology, ELTE Eötvös Loránd University, 1075 Budapest, Hungary; (A.M.); (J.F.); (A.E.); (O.K.); (A.S.); (R.U.); (E.K.)
| | - Marc N. Potenza
- Departments of Psychiatry, Child Study and Neuroscience, Yale University School of Medicine, New Haven, CT 06511, USA;
- Connecticut Council on Problem Gambling, Wethersfield, CT 06109, USA
- Connecticut Mental Health Center, New Haven, CT 06519, USA
| | - Rajendra D. Badgaiyan
- Department of Psychiatry, Ichan School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Kenneth Blum
- Division of Addiction Research & Education, Center for Psychiatry, Medicine, & Primary Care (Office of the Provost), Western University Health Sciences, Pomona, CA 91766, USA;
| | - Zsolt Demetrovics
- Institute of Psychology, ELTE Eötvös Loránd University, 1075 Budapest, Hungary; (A.M.); (J.F.); (A.E.); (O.K.); (A.S.); (R.U.); (E.K.)
- Division of Addiction Research & Education, Center for Psychiatry, Medicine, & Primary Care (Office of the Provost), Western University Health Sciences, Pomona, CA 91766, USA;
- Correspondence: (C.B.); (Z.D.)
| | - Eszter Kotyuk
- Institute of Psychology, ELTE Eötvös Loránd University, 1075 Budapest, Hungary; (A.M.); (J.F.); (A.E.); (O.K.); (A.S.); (R.U.); (E.K.)
| |
Collapse
|
124
|
Chung YJ, Salvi A, Kalailingam P, Alnawaz M, Tan SH, Pan JY, Tan NS, Thanabalu T. N-WASP Attenuates Cell Proliferation and Migration through ERK2-Dependent Enhanced Expression of TXNIP. BIOLOGY 2022; 11:biology11040582. [PMID: 35453780 PMCID: PMC9029996 DOI: 10.3390/biology11040582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/10/2022] [Accepted: 04/08/2022] [Indexed: 11/16/2022]
Abstract
Simple Summary Neural Wiskott–Aldrich Syndrome Protein (N-WASP) regulates actin cytoskeleton remodeling and can, it has been suggested, suppress several cancers. In this study, HSC-5 cells, a mammalian cell line with reduced N-WASP expression, were used to generate control cells and HSC-5 cells with increased N-WASP expression that is comparable to that of normal keratinocytes. The two cell lines were used to elucidate the regulation of cell proliferation and migration by N-WASP. Our findings suggest that N-WASP increases ERK2-dependent phosphorylation of FOXO1 and increases TXNIP expression, which reduces cell proliferation and migration. This study is the first to propose an antiproliferative role of N-WASP, which is mediated via ERK2, and it suggests new avenues for cancer therapeutic research and treatment. Abstract Neural Wiskott–Aldrich Syndrome Protein (N-WASP) regulates actin cytoskeleton remodeling. It has been known that reduced N-WASP expression in breast and colorectal cancers is associated with poor prognosis. Here, we found reduced N-WASP expression in squamous cell carcinoma (SCC) patient samples. The SCC cell line HSC-5 with reduced N-WASP expression was used to generate HSC-5CN (control) and HSC-5NW (N-WASP overexpression) cells. HSC-5NW cells had reduced cell proliferation and migration compared to HSC-5CN cells. HSC-5NW cells had increased phospho-ERK2 (extracellular signal-regulated kinase 2), phosphorylated Forkhead box protein class O1 (FOXO1) and reduced nuclear FOXO1 staining compared to HSC-5CN cells. Proteasome inhibition stabilized total FOXO1, however, not nuclear staining, suggesting that FOXO1 could be degraded in the cytoplasm. Inhibition of ERK2 enhanced nuclear FOXO1 levels and restored cell proliferation and migration of HSC-5NW to those of HSC-5CN cells, suggesting that ERK2 regulates FOXO1 activity. The expression of thioredoxin-interacting protein (TXNIP), a FOXO1 target that inhibits thioredoxin and glucose uptake, was higher in HSC-5NW cells than in HSC-5CN cells. Knockdown of TXNIP in HSC-5NW cells restored cell proliferation and migration to those of HSC-5CN cells. Thus, we propose that N-WASP regulates cell proliferation and migration via an N-WASP-ERK2-FOXO1-TXNIP pathway.
Collapse
Affiliation(s)
- Yat Joong Chung
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore; (Y.J.C.); (A.S.); (P.K.); (M.A.); (N.S.T.)
| | - Amrita Salvi
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore; (Y.J.C.); (A.S.); (P.K.); (M.A.); (N.S.T.)
| | - Pazhanichamy Kalailingam
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore; (Y.J.C.); (A.S.); (P.K.); (M.A.); (N.S.T.)
| | - Myra Alnawaz
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore; (Y.J.C.); (A.S.); (P.K.); (M.A.); (N.S.T.)
| | - Suat Hoon Tan
- National Skin Centre, 1 Mandalay Road, Singapore 308205, Singapore; (S.H.T.); (J.Y.P.)
| | - Jiun Yit Pan
- National Skin Centre, 1 Mandalay Road, Singapore 308205, Singapore; (S.H.T.); (J.Y.P.)
| | - Nguan Soon Tan
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore; (Y.J.C.); (A.S.); (P.K.); (M.A.); (N.S.T.)
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore 308232, Singapore
| | - Thirumaran Thanabalu
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore; (Y.J.C.); (A.S.); (P.K.); (M.A.); (N.S.T.)
- Correspondence: ; Tel.: +65-6316-2832; Fax: +65-6791-3856
| |
Collapse
|
125
|
Davis LJ, Maldonado AC, Khin M, Krunic A, Burdette JE, Orjala J. Aulosirazoles B and C from the Cyanobacterium Nostoc sp. UIC 10771: Analogues of an Isothiazolonaphthoquinone Scaffold that Activate Nuclear Transcription Factor FOXO3a in Ovarian Cancer Cells. JOURNAL OF NATURAL PRODUCTS 2022; 85:540-546. [PMID: 35100504 PMCID: PMC8957594 DOI: 10.1021/acs.jnatprod.1c01030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The known solid-tumor-selective cytotoxin aulosirazole (1) was identified from bioactive extracts from the culture medium of the cyanobacterium Nostoc sp. UIC 10771. Here, we demonstrate that 1 induces the nuclear accumulation of FOXO3a in OVCAR3 using both Western blot analysis and immunofluorescence confocal microscopy. We also report the discovery of two additional analogues, aulosirazoles B (2) and C (3). Structures for compounds 2 and 3 were determined using HR-ESI-LC-MS/MS and 1D and 2D NMR experiments. Aulosirazoles B (2) and C (3) represent the first natural analogues of the FOXO-activating compound aulosirazole (1) and are the second and third isothiazole-containing metabolites reported from this phylum.
Collapse
Affiliation(s)
- Lydia J Davis
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Amanda C Maldonado
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Manead Khin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Aleksej Krunic
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Joanna E Burdette
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Jimmy Orjala
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| |
Collapse
|
126
|
Fan W, Ma H, Jin B. Expression of FOXM1 and PLK1 predicts prognosis of patients with hepatocellular carcinoma. Oncol Lett 2022; 23:146. [PMID: 35350587 PMCID: PMC8941521 DOI: 10.3892/ol.2022.13266] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 02/16/2022] [Indexed: 11/06/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most frequently encountered malignant tumor types and to improve its treatment, effective prognostic biomarkers are urgently required. Cell cycle dysregulation is a significant feature of cancer progression. The aim of the present study was to estimate the expression levels of forkhead box protein M1 (FOXM1) and polo-like kinase 1 (PLK1), both of which have essential roles in cell cycle regulation, and determine their prognostic value in HCC. To this end, FOXM1 and PLK1 expression levels were assessed in The Cancer Genome Atlas and International Cancer Genome Consortium Japan HCC cohorts, and the associations between their co-expression were determined via Pearson's correlation analysis. Furthermore, the overall survival and disease-free survival in these cohorts for different FOXM1 and PLK1 expression statuses were analyzed. In vitro knockdown experiments were also performed using Huh7 cells. The results obtained indicated overexpression of FOXM1 and PLK1 in HCC tumor tissues as well as a positive correlation between FOXM1 and PLK1 expression. The results also suggested that both FOXM1 and PLK1 are required for HCC cell proliferation. In addition, upregulation of FOXM1 and PLK1 was indicated to be associated with poor prognosis of patients with HCC. However, only their coordinated overexpression was identified as an independent prognostic factor for HCC.
Collapse
Affiliation(s)
- Weiqiang Fan
- Department of Organ Transplantation, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Huan Ma
- Department of Organ Transplantation, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Bin Jin
- Department of Organ Transplantation, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
127
|
Li C, Guan X, Jing H, Xiao X, Jin H, Xiong J, Ai S, Wang Y, Su T, Sun G, Fu T, Wang Y, Guo S, Liang P. Circular RNA circBFAR promotes glioblastoma progression by regulating a miR-548b/FoxM1 axis. FASEB J 2022; 36:e22183. [PMID: 35202487 DOI: 10.1096/fj.202101307r] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 12/30/2021] [Accepted: 01/18/2022] [Indexed: 01/04/2023]
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive type of tumor of the primary nervous system. Treatment options for GBM include surgery, chemotherapy, and radiation therapy; however, the clinical outcomes are poor, with a high rate of recurrence. An increasing number of studies have shown that circular RNAs (circRNAs) serve important roles in several types of cancer. Gene Expression Omnibus (GEO) database was utilized to identify the differentially expressed circRNAs and their biological functions. Then, we detected the circular RNA bifunctional apoptosis regulator (circBFAR) was significantly increased in three GEO datasets. However, the role of circBFAR has not been reported in GBM. In this study, the expression of circBFAR was significantly increased both in GBM tissues or cell lines and was negatively correlated with overall survival in patients with GBM. Knockdown of circBFAR inhibited proliferation and invasion both in vitro and in vivo. Increased expression of circBFAR resulted in a reduction of miR-548b expression in glioma cells. A luciferase reporter and RIP assay indicated that miR-548b was a direct target of circBFAR, and miR-548b may negatively regulate the expression of FoxM1. Rescue experiments showed that overexpression of FoxM1 could counter the effect of circBFAR silencing on the proliferation and invasion of glioma cell lines. Moreover, we identified that circBFAR regulates FoxM1 by interacting with miR-548b in glioma cells. In conclusion, the present study demonstrated that a circBFAR/miR-548b/FoxM1 axis regulates the development of GBM and highlights potentially novel therapeutic targets for the treatment of GBM.
Collapse
Affiliation(s)
- Chenlong Li
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xue Guan
- Animal Laboratory Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hanguang Jing
- Breast Surgery, Lin Yi Famous Doctor Studio, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Xu Xiao
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hua Jin
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jinsheng Xiong
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Siqi Ai
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yingjie Wang
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Tianqi Su
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Guiyin Sun
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Tianjiao Fu
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yujie Wang
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shouli Guo
- Animal Experiment Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Peng Liang
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
128
|
Fusi L, Paudel R, Meder K, Schlosser A, Schrama D, Goebeler M, Schmidt M. Interaction of transcription factor FoxO3 with histone acetyltransferase complex subunit TRRAP Modulates Gene Expression and Apoptosis. J Biol Chem 2022; 298:101714. [PMID: 35151693 PMCID: PMC8914384 DOI: 10.1016/j.jbc.2022.101714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 12/28/2021] [Accepted: 01/19/2022] [Indexed: 11/01/2022] Open
Abstract
Forkhead box O (FoxO) transcription factors are conserved proteins involved in the regulation of life span and age-related diseases, such as diabetes and cancer. Stress stimuli or growth factor deprivation promotes nuclear localization and activation of FoxO proteins, which—depending on the cellular context—can lead to cell cycle arrest or apoptosis. In endothelial cells (ECs), they further regulate angiogenesis and may promote inflammation and vessel destabilization implicating a role of FoxOs in vascular diseases. In several cancers, FoxOs exert a tumor-suppressive function by regulating proliferation and survival. We and others have previously shown that FoxOs can regulate these processes via two different mechanisms: by direct binding to forkhead-responsive elements at the promoter of target genes or by a poorly understood alternative process that does not require direct DNA binding and regulates key targets in primary human ECs. Here, we performed an interaction study in ECs to identify new nuclear FoxO3 interaction partners that might contribute to FoxO-dependent gene regulation. Mass spectrometry analysis of FoxO3-interacting proteins revealed transformation/transcription domain–associated protein (TRRAP), a member of multiple histone acetyltransferase complexes, as a novel binding partner of FoxO family proteins. We demonstrate that TRRAP is required to support FoxO3 transactivation and FoxO3-dependent G1 arrest and apoptosis in ECs via transcriptional activation of the cyclin-dependent kinase inhibitor p27kip1 and the proapoptotic B-cell lymphoma 2 family member, BIM. Moreover, FoxO–TRRAP interaction could explain FoxO-induced alternative gene regulation via TRRAP-dependent recruitment to target promoters lacking forkhead-responsive element sequences.
Collapse
|
129
|
Histone deacetylase 3 contributes to the antiviral innate immunity of macrophages by interacting with FOXK1 to regulate STAT1/2 transcription. Cell Rep 2022; 38:110302. [PMID: 35081346 DOI: 10.1016/j.celrep.2022.110302] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 11/16/2021] [Accepted: 01/05/2022] [Indexed: 12/15/2022] Open
Abstract
It is well known that interferon (IFN)-α/-β activates the JAK/STAT signaling pathway and suppresses viral replication through the induction of IFN stimulated genes (ISGs). Here, we report that knockout of HDAC3 from macrophages results in the decreased expression of STAT1 and STAT2, leading to defective antiviral immunity in cells and mice. Further studies show that HDAC3 interacts with a conserved transcription factor Forkhead Box K1 (FOXK1), co-localizes with FOXK1 at the promoter of STAT1 and STAT2, and is required for protecting FOXK1 from lysosomal system-mediated degradation. FOXK1-deficient macrophages also show low STAT1 and STAT2 expression with defective responses to viruses. Thus, our studies uncover the biological importance of HDAC3 in regulating the antiviral immunity of macrophages through interacting with FOXK1 to regulate the expression of STAT1 and STAT2.
Collapse
|
130
|
Li S, Wang F. Forkhead Box S1 inhibits the progression of lung squamous cell carcinoma cells by mediating Wnt/β-catenin pathway. CHINESE J PHYSIOL 2022; 65:266-275. [DOI: 10.4103/0304-4920.359799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
131
|
Wang XW, Guo QQ, Yu Y, Zhou TT, Zhang SY, Wang Z, Liu JW, Tang J, Jiang XY, Wang SS, Guo WD, Xu HD, Sun HY, Li ZW, Song XY, Zhao JG, Cao L. The deacetylation of Foxk2 by Sirt1 reduces chemosensitivity to cisplatin. J Cell Mol Med 2021; 26:491-506. [PMID: 34866322 PMCID: PMC8743664 DOI: 10.1111/jcmm.17107] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/23/2021] [Accepted: 11/26/2021] [Indexed: 01/16/2023] Open
Abstract
In multiple types of cancer, decreased tumour cell apoptosis during chemotherapy is indicative of decreased chemosensitivity. Forkhead box K2 (FOXK2), which is essential for cell fate, regulates cancer cell apoptosis through several post‐translational modifications. However, FOXK2 acetylation has not been extensively studied. Here, we evaluated the effects of sirtiun 1 (SIRT1) on FOXK2 deacetylation. Our findings demonstrated that SIRT1 inhibition increased FOXK2‐induced chemosensitivity to cisplatin and that K223 in FOXK2 was acetylated. Furthermore, FOXK2 K223 deacetylation reduced chemosensitivity to cisplatin in vitro and in vivo. Mechanistically, FOXK2 was acetylated by the acetyltransferase cAMP response element binding protein and deacetylated by SIRT1. Furthermore, cisplatin attenuated the interaction between FOXK2 and SIRT1. Cisplatin or SIRT1 inhibition enhanced FOXK2 acetylation, thereby reducing the nuclear distribution of FOXK2. Additionally, FOXK2 K223 acetylation significantly affected the expression of cell cycle–related and apoptosis‐related genes in cisplatin‐stimulated cancer cells, and FOXK2 K223 hyperacetylation promoted mitotic catastrophe, which enhanced chemosensitivity to cisplatin. Overall, our results provided insights into the mechanisms of SIRT1‐mediated FOXK2 deacetylation, which was involved in chemosensitivity to cisplatin.
Collapse
Affiliation(s)
- Xi-Wen Wang
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China.,Key Laboratory of Liaoning Province, China Medical University, Shenyang, China.,Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Qi-Qiang Guo
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China.,Key Laboratory of Liaoning Province, China Medical University, Shenyang, China
| | - Yang Yu
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China.,Key Laboratory of Liaoning Province, China Medical University, Shenyang, China
| | - Ting-Ting Zhou
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China.,Key Laboratory of Liaoning Province, China Medical University, Shenyang, China
| | - Si-Yi Zhang
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China.,Key Laboratory of Liaoning Province, China Medical University, Shenyang, China
| | - Zhuo Wang
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China.,Key Laboratory of Liaoning Province, China Medical University, Shenyang, China
| | - Jing-Wei Liu
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China.,Key Laboratory of Liaoning Province, China Medical University, Shenyang, China
| | - Jun Tang
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiao-You Jiang
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China.,Key Laboratory of Liaoning Province, China Medical University, Shenyang, China
| | - Shan-Shan Wang
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China.,Key Laboratory of Liaoning Province, China Medical University, Shenyang, China
| | - Wen-Dong Guo
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China.,Key Laboratory of Liaoning Province, China Medical University, Shenyang, China
| | - Hong-de Xu
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China.,Key Laboratory of Liaoning Province, China Medical University, Shenyang, China
| | - Hua-Yi Sun
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China.,Key Laboratory of Liaoning Province, China Medical University, Shenyang, China
| | - Zi-Wei Li
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China.,Key Laboratory of Liaoning Province, China Medical University, Shenyang, China
| | - Xiao-Yu Song
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China.,Key Laboratory of Liaoning Province, China Medical University, Shenyang, China
| | - Jun-Gang Zhao
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Liu Cao
- College of Basic Medical Science, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China.,Key Laboratory of Liaoning Province, China Medical University, Shenyang, China
| |
Collapse
|
132
|
Puri D, Koschorz B, Engist B, Onishi-Seebacher M, Ryan D, Soujanya M, Montavon T. Foxd3 controls heterochromatin-mediated repression of repeat elements and 2-cell state transcription. EMBO Rep 2021; 22:e53180. [PMID: 34605600 PMCID: PMC8647145 DOI: 10.15252/embr.202153180] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/18/2021] [Accepted: 09/14/2021] [Indexed: 11/17/2022] Open
Abstract
Repeat element transcription plays a vital role in early embryonic development. The expression of repeats such as MERVL characterises mouse embryos at the 2‐cell stage and defines a 2‐cell‐like cell (2CLC) population in a mouse embryonic stem cell culture. Repeat element sequences contain binding sites for numerous transcription factors. We identify the forkhead domain transcription factor FOXD3 as a regulator of major satellite repeats and MERVL transcription in mouse embryonic stem cells. FOXD3 binds to and recruits the histone methyltransferase SUV39H1 to MERVL and major satellite repeats, consequentially repressing the transcription of these repeats by the establishment of the H3K9me3 heterochromatin modification. Notably, depletion of FOXD3 leads to the de‐repression of MERVL and major satellite repeats as well as a subset of genes expressed in the 2‐cell state, shifting the balance between the stem cell and 2‐cell‐like population in culture. Thus, FOXD3 acts as a negative regulator of repeat transcription, ascribing a novel function to this transcription factor.
Collapse
Affiliation(s)
- Deepika Puri
- Department of Epigenetics, Max Planck Institute for Immunobiology and Epigenetics, Freiburg, Germany.,National Centre for Cell Science, Savitribai Phule Pune University, Pune, India
| | - Birgit Koschorz
- Department of Epigenetics, Max Planck Institute for Immunobiology and Epigenetics, Freiburg, Germany
| | - Bettina Engist
- Department of Epigenetics, Max Planck Institute for Immunobiology and Epigenetics, Freiburg, Germany
| | - Megumi Onishi-Seebacher
- Department of Epigenetics, Max Planck Institute for Immunobiology and Epigenetics, Freiburg, Germany
| | - Devon Ryan
- Department of Epigenetics, Max Planck Institute for Immunobiology and Epigenetics, Freiburg, Germany
| | | | - Thomas Montavon
- Department of Epigenetics, Max Planck Institute for Immunobiology and Epigenetics, Freiburg, Germany
| |
Collapse
|
133
|
Kawaratani H, Sawai H, Onishi M, Kogiso T, Shimada N, Uojima H, Nakajima T, Matsumoto N, Ikejima K, Ishikawa T, Terai S, Motoyama H, Komori A, Hirashima N, Saito S, Eguchi Y, Nojima M, Kawai Y, Tateyama M, Yoshiji H, Tanaka Y. A genome-wide association study identifying SVEP1 variant as a predictor of response to tolvaptan for cirrhotic ascites. Liver Int 2021; 41:2944-2953. [PMID: 34309184 DOI: 10.1111/liv.15022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/03/2021] [Accepted: 07/15/2021] [Indexed: 02/13/2023]
Abstract
BACKGROUND & AIMS Tolvaptan, vasopressin V2-receptor antagonist, has been used for patients with difficult-to-treat ascites in Japan. In this study, we conducted a genome-wide association study (GWAS) in the Japanese population to identify genetic variants associated with tolvaptan's efficacy for patients with hepatic ascites. METHODS From 2014 through 2018, genomic DNA samples were obtained from 550 patients who were treated with tolvaptan. Of those, 80 cases (non-responder; increase of body weight [BW]) and 333 controls (responder; >1.5 kg decrease of BW) were included in the GWAS and replication study. RESULTS Genome-wide association study showed 5 candidate SNPs around the miR818, KIAA1109, and SVEP1 genes. After validation and performing a replication study, an SNP (rs2991364) located in the SVEP1 gene was found to have a significant genome-wide association (OR = 3.55, P = 2.01 × 10-8 ). Multivariate analyses showed that serum sodium (Na), blood urea nitrogen (BUN) and SVEP1 SNP were significantly associated with the response (OR = 0.92, P = .003; OR = 1.02, P = .02 and OR = 3.98, P = .000008, respectively). Based on a prediction model of logistic regression analysis in a population with the rs2991364 risk allele, the failure probability (=exp (score: 22.234 + BUN*0.077 + Na*-0.179) (1 + exp (score)) was determined for the detection of non-responders. Assuming a cutoff of failure probability at 38.6%, sensitivity was 84.4%, specificity was 70% and AUC was 0.774. CONCLUSION SVEP1 rs2991364 was identified as the specific SNP for the tolvaptan response. The prediction score (>38.6%) can identify tolvaptan non-responders and help to avoid a lengthy period of futile treatment.
Collapse
Affiliation(s)
- Hideto Kawaratani
- Department of Gastroenterology, Nara Medical University, Nara, Japan
| | - Hiromi Sawai
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masaya Onishi
- Department of Virology and Liver Unit, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Tomomi Kogiso
- Institute of Gastroenterology, Department of Internal Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Noritomo Shimada
- Division of Gastroenterology and Hepatology, Ootakanomori Hospital, Kashiwa, Japan
| | - Haruki Uojima
- Department of Gastroenterology, Internal Medicine, Kitasato University School of Medicine, Sagamihara, Japan
| | - Tomoaki Nakajima
- Department of Hepatology, Sapporo Kosei General Hospital, Hokkaido, Japan
| | - Naoki Matsumoto
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Kenichi Ikejima
- Department of Gastroenterology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Toru Ishikawa
- Department of Gastroenterology, Saiseikai Niigata Hospital, Niigata, Japan
| | - Shuji Terai
- Division of Gastroenterology and Hepatology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Hiroyuki Motoyama
- Department of Hepatology, Graduate School of Medicine, Osaka City University Osaka, Japan
| | - Atsumasa Komori
- Clinical Research Center, Nagasaki Medical Center, Nagasaki, Japan
| | - Noboru Hirashima
- Department of Gastroenterology, National Hospital Organization, Nagoya Medical Center, Nagoya, Japan
| | - Satoru Saito
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | | | - Masanori Nojima
- Center for Translational Research, The Institute of Medical Science, the University of Tokyo, Tokyo, Japan
| | - Yosuke Kawai
- Genome Medical Science Project (Toyama), National Center for Global Health and Medicine, Tokyo, Japan
| | - Masakuni Tateyama
- Department of Gastroenterology and Hepatology, Kumamoto University, Kumamoto, Japan
| | - Hitoshi Yoshiji
- Department of Gastroenterology, Nara Medical University, Nara, Japan
| | - Yasuhito Tanaka
- Department of Virology and Liver Unit, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.,Department of Gastroenterology and Hepatology, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
134
|
Structure-based virtual screening towards the discovery of novel FOXM1 inhibitors. Future Med Chem 2021; 14:207-219. [PMID: 34809496 DOI: 10.4155/fmc-2021-0282] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aim: Given the importance of FOXM1 in the treatment of ovarian cancer, we aimed to identify an excellent specific inhibitor and examined its underlying therapeutic effect. Materials & methods: The binding statistics for FDI-6 with FOXM1 were calculated through computer-aided drug design (CADD). We selected XST-119 through virtual screening, performed surface plasmon resonance and in vitro cell antiproliferative activity analysis and evaluated its antitumor efficacy in a mouse model. Results: XST-119 had significantly higher affinity for FOXM1 and antiproliferative activity than FDI-6. XST-119 had a definite inhibitory activity in a xenograft mouse model. Conclusion: We identified XST-119, a FOXM1 inhibitor, with better efficacy for treatment of ovarian cancer. FOXM1 binding sites for small molecules are also highlighted, which may provide the foundation for further drug discovery.
Collapse
|
135
|
Virus-induced FoxO factor facilitates replication of human cytomegalovirus. Arch Virol 2021; 167:109-121. [PMID: 34751815 DOI: 10.1007/s00705-021-05279-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 09/06/2021] [Indexed: 10/19/2022]
Abstract
Recently, it was reported that the forkhead box O (FoxO) transcription factor promotes human cytomegalovirus (HCMV) replication via direct binding to the promoters of the major immediate-early (MIE) genes, but how the FoxO factor impacts HCMV replication remains unknown. Here, it is reported that FoxO1 expression is strongly induced by HCMV infection in cells of fibroblast origin. Suppression of the FoxO1 gene by specific RNA interference significantly inhibited HCMV growth and replication, but viral DNA synthesis was not affected considerably. Interestingly, depletion or overexpression of FoxO1 had a significant effect on the expression of viral early/late transcripts. FoxO1 was found to colocalize with the pUL44 protein subunit of viral replication compartments without direct association with DNA. This study highlights how FoxO enhances HCMV gene transcription and viral replication to promote infection.
Collapse
|
136
|
Porras L, Ismail H, Mader S. Positive Regulation of Estrogen Receptor Alpha in Breast Tumorigenesis. Cells 2021; 10:cells10112966. [PMID: 34831189 PMCID: PMC8616513 DOI: 10.3390/cells10112966] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/23/2021] [Accepted: 10/24/2021] [Indexed: 12/31/2022] Open
Abstract
Estrogen receptor alpha (ERα, NR3A1) contributes through its expression in different tissues to a spectrum of physiological processes, including reproductive system development and physiology, bone mass maintenance, as well as cardiovascular and central nervous system functions. It is also one of the main drivers of tumorigenesis in breast and uterine cancer and can be targeted by several types of hormonal therapies. ERα is expressed in a subset of luminal cells corresponding to less than 10% of normal mammary epithelial cells and in over 70% of breast tumors (ER+ tumors), but the basis for its selective expression in normal or cancer tissues remains incompletely understood. The mapping of alternative promoters and regulatory elements has delineated the complex genomic structure of the ESR1 gene and shed light on the mechanistic basis for the tissue-specific regulation of ESR1 expression. However, much remains to be uncovered to better understand how ESR1 expression is regulated in breast cancer. This review recapitulates the current body of knowledge on the structure of the ESR1 gene and the complex mechanisms controlling its expression in breast tumors. In particular, we discuss the impact of genetic alterations, chromatin modifications, and enhanced expression of other luminal transcription regulators on ESR1 expression in tumor cells.
Collapse
|
137
|
Roßwag S, Sleeman JP, Thaler S. RASSF1A-Mediated Suppression of Estrogen Receptor Alpha (ERα)-Driven Breast Cancer Cell Growth Depends on the Hippo-Kinases LATS1 and 2. Cells 2021; 10:cells10112868. [PMID: 34831091 PMCID: PMC8616147 DOI: 10.3390/cells10112868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/07/2021] [Accepted: 10/12/2021] [Indexed: 11/27/2022] Open
Abstract
Around 70% of breast cancers express the estrogen receptor alpha (ERα). This receptor is of central importance for breast cancer development and estrogen-dependent tumor growth. However, the molecular mechanisms that are responsible for the control of ERα expression and function in the context of breast carcinogenesis are complex and not fully understood. In previous work, we have demonstrated that the tumor suppressor RASSF1A suppresses estrogen-dependent growth of breast cancer cells through a complex network that keeps ERα expression and function under control. We observed that RASSF1A mediates the suppression of ERα expression through modulation of the Hippo effector Yes-associated protein 1 (YAP1) activity. Here we report that RASSF1A-mediated alteration of YAP1 depends on the Hippo-kinases LATS1 and LATS2. Based on these results, we conclude that inactivation of RASSF1A causes changes in the function of the Hippo signaling pathway and altered activation of YAP1, and as a consequence, increased expression and function of ERα. Thus, the inactivation of RASSF1A might constitute a fundamental event that supports the initiation of ERα-dependent breast cancer. Furthermore, our results support the notion that the Hippo pathway is important for the suppression of luminal breast cancers, and that the tumor-suppressor function of RASSF1A depends on LATS1 and LATS2.
Collapse
Affiliation(s)
- Sven Roßwag
- Department of Microvascular Biology and Pathobiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany; (S.R.); (J.P.S.)
| | - Jonathan P. Sleeman
- Department of Microvascular Biology and Pathobiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany; (S.R.); (J.P.S.)
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT) Campus Nord, 76344 Eggenstein-Leupoldshafen, Germany
| | - Sonja Thaler
- Department of Microvascular Biology and Pathobiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany; (S.R.); (J.P.S.)
- Correspondence: ; Tel.: +49-621-383-71599; Fax: +49-621-383-71451
| |
Collapse
|
138
|
FOXA1 of regulatory variant associated with risk of breast cancer through allele-specific enhancer in the Chinese population. Breast Cancer 2021; 29:247-259. [PMID: 34635981 DOI: 10.1007/s12282-021-01305-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 10/07/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND FOXA1 is a pioneer transcription factor which has been established as a carcinogenic factor and can regulate the expression of downstream target genes in breast cancer. We hypothesized that genetic variants modulating FOXA1 expression might play a role in the risk of breast cancer. METHODS Physical interaction predicted by PreSTIGE analysis and CHIA-PET data integration with cis-expression quantitative trait loci (cis-eQTL) based SNP-FOXA1 analysis were used to identify potentially regulatory variants modulating the expression of FOXA1. Then, we utilized a case-control study consisting of 855 new diagnosed breast cancer cases and 920 controls in the Chinese population to identify breast cancer associated variants. Biological assays were conducted in breast cancer cell lines to illustrate the effects of associated variants on breast cancer risk. RESULTS We identified that rs7160774 G > A variant was associated with lower risk of breast cancer (OR = 0.77, 95% confidence interval = 0.62-0.96, P = 0.022). Biological experiments indicated that rs7160774[A] allele down-regulated the expression of FOXA1 compared to the G allele by influencing transcription factor binding affinity, thus playing an important role in the development of breast cancer. CONCLUSION Our study suggested that the regulatory variant rs7160774 was associated with risk of breast cancer by long-range modulating FOXA1 expression and provided critical insights into pinpoint causal genetic variants.
Collapse
|
139
|
A pathogenic deletion in Forkhead Box L1 (FOXL1) identifies the first otosclerosis (OTSC) gene. Hum Genet 2021; 141:965-979. [PMID: 34633540 PMCID: PMC9034980 DOI: 10.1007/s00439-021-02381-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 09/30/2021] [Indexed: 12/23/2022]
Abstract
Otosclerosis is a bone disorder of the otic capsule and common form of late-onset hearing impairment. Considered a complex disease, little is known about its pathogenesis. Over the past 20 years, ten autosomal dominant loci (OTSC1-10) have been mapped but no genes identified. Herein, we map a new OTSC locus to a 9.96 Mb region within the FOX gene cluster on 16q24.1 and identify a 15 bp coding deletion in Forkhead Box L1 co-segregating with otosclerosis in a Caucasian family. Pre-operative phenotype ranges from moderate to severe hearing loss to profound sensorineural loss requiring a cochlear implant. Mutant FOXL1 is both transcribed and translated and correctly locates to the cell nucleus. However, the deletion of 5 residues in the C-terminus of mutant FOXL1 causes a complete loss of transcriptional activity due to loss of secondary (alpha helix) structure. FOXL1 (rs764026385) was identified in a second unrelated case on a shared background. We conclude that FOXL1 (rs764026385) is pathogenic and causes autosomal dominant otosclerosis and propose a key inhibitory role for wildtype Foxl1 in bone remodelling in the otic capsule. New insights into the molecular pathology of otosclerosis from this study provide molecular targets for non-invasive therapeutic interventions.
Collapse
|
140
|
Shi J, Wang J, Cheng H, Liu S, Hao X, Lan L, Wu G, Liu M, Zhao Y. FOXP4 promotes laryngeal squamous cell carcinoma progression through directly targeting LEF‑1. Mol Med Rep 2021; 24:831. [PMID: 34590150 PMCID: PMC8503739 DOI: 10.3892/mmr.2021.12471] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 08/25/2021] [Indexed: 12/14/2022] Open
Abstract
Forkhead box (FOX) proteins are multifaceted transcription factors that have been shown to be involved in cell cycle progression, proliferation and metastasis. FOXP4, a member of the FOX family, has been implicated in diverse biological processes in tumor initiation and progression. However, the molecular mechanisms of FOXP4 in laryngeal squamous cell carcinoma (LSCC) remain unknown. In the present study, differentially expressed transcripts in transforming growth factor‑β‑treated TU177 cells were screened using microarrays and it was found that FOXP4 was significantly upregulated. The high expression of FOXP4 was detected in LSCC tissues and cells, and predicted poor prognosis. The role of FOXP4 in laryngeal cancer cell proliferation, migration and invasion was determined by gain‑ and loss‑of‑function assays. Besides, FOXP4 was demonstrated to participate in the epithelial‑mesenchymal transition process at the mRNA and protein levels. Mechanically, FOXP4 directly bound to the promoter of lymphoid enhancer‑binding factor 1 and activated Wnt signaling pathway, which was confirmed via chromatin immunoprecipitation and luciferase reporter assays. Consequently, these findings provided novel mechanisms of FOXP4 in LSCC progression, which may be considered as potential therapeutic and prognostic targets for LSCC.
Collapse
Affiliation(s)
- Jian Shi
- Department of Otolaryngology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Jingtian Wang
- Department of Otolaryngology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Hongkun Cheng
- Department of Otorhinolaryngology, The Third Hospital of Handan, Handan, Hebei 056001, P.R. China
| | - Shenghui Liu
- Department of Otolaryngology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Xiaowei Hao
- Department of Otorhinolaryngology, The People's Hospital of Cixian, Handan, Hebei 056500, P.R. China
| | - Lili Lan
- Department of Otolaryngology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Gancun Wu
- Department of Otolaryngology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Meng Liu
- Department of Otolaryngology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Yan Zhao
- Department of Otolaryngology, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
141
|
Berghausen EM, Janssen W, Vantler M, Gnatzy-Feik LL, Krause M, Behringer A, Joseph C, Zierden M, Freyhaus HT, Klinke A, Baldus S, Alcazar MA, Savai R, Pullamsetti SS, Wong DW, Boor P, Zhao JJ, Schermuly RT, Rosenkranz S. Disrupted PI3K subunit p110α signaling protects against pulmonary hypertension and reverses established disease in rodents. J Clin Invest 2021; 131:136939. [PMID: 34596056 DOI: 10.1172/jci136939] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 08/18/2021] [Indexed: 11/17/2022] Open
Abstract
Enhanced signaling via RTKs in pulmonary hypertension (PH) impedes current treatment options because it perpetuates proliferation and apoptosis resistance of pulmonary arterial smooth muscle cells (PASMCs). Here, we demonstrated hyperphosphorylation of multiple RTKs in diseased human vessels and increased activation of their common downstream effector phosphatidylinositol 3'-kinase (PI3K), which thus emerged as an attractive therapeutic target. Systematic characterization of class IA catalytic PI3K isoforms identified p110α as the key regulator of pathogenic signaling pathways and PASMC responses (proliferation, migration, survival) downstream of multiple RTKs. Smooth muscle cell-specific genetic ablation or pharmacological inhibition of p110α prevented onset and progression of pulmonary hypertension (PH) as well as right heart hypertrophy in vivo and even reversed established vascular remodeling and PH in various animal models. These effects were attributable to both inhibition of vascular proliferation and induction of apoptosis. Since this pathway is abundantly activated in human disease, p110α represents a central target in PH.
Collapse
Affiliation(s)
- Eva M Berghausen
- Department of Cardiology, Heart Center at the University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC) and.,Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
| | - Wiebke Janssen
- Max-Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,University of Giessen and Marburg Lung Center (UGMLC), and German Centre for Lung Research (DZL), Giessen, Germany
| | - Marius Vantler
- Department of Cardiology, Heart Center at the University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC) and.,Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
| | - Leoni L Gnatzy-Feik
- Department of Cardiology, Heart Center at the University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC) and.,Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
| | - Max Krause
- Department of Cardiology, Heart Center at the University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC) and.,Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
| | - Arnica Behringer
- Department of Cardiology, Heart Center at the University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC) and
| | - Christine Joseph
- Department of Cardiology, Heart Center at the University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC) and
| | - Mario Zierden
- Department of Cardiology, Heart Center at the University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC) and.,Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
| | - Henrik Ten Freyhaus
- Department of Cardiology, Heart Center at the University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC) and.,Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
| | - Anna Klinke
- Department of Cardiology, Heart Center at the University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC) and.,Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
| | - Stephan Baldus
- Department of Cardiology, Heart Center at the University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC) and.,Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
| | - Miguel A Alcazar
- Center for Molecular Medicine Cologne (CMMC) and.,Institute for Lung Health, member of the DZL, UGMLC, Giessen, Germany.,Department of Pediatric and Adolecent Medicine, University of Cologne, Cologne, Germany
| | - Rajkumar Savai
- Max-Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | - Dickson Wl Wong
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Peter Boor
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Jean J Zhao
- Dana-Farber Cancer Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Ralph T Schermuly
- Max-Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,University of Giessen and Marburg Lung Center (UGMLC), and German Centre for Lung Research (DZL), Giessen, Germany
| | - Stephan Rosenkranz
- Department of Cardiology, Heart Center at the University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC) and.,Cologne Cardiovascular Research Center (CCRC), University of Cologne, Cologne, Germany
| |
Collapse
|
142
|
Hale AE, Moorman NJ. The Ends Dictate the Means: Promoter Switching in Herpesvirus Gene Expression. Annu Rev Virol 2021; 8:201-218. [PMID: 34129370 DOI: 10.1146/annurev-virology-091919-072841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Herpesvirus gene expression is dynamic and complex, with distinct complements of viral genes expressed at specific times in different infection contexts. These complex patterns of viral gene expression arise in part from the integration of multiple cellular and viral signals that affect the transcription of viral genes. The use of alternative promoters provides an increased level of control, allowing different promoters to direct the transcription of the same gene in response to distinct temporal and contextual cues. While once considered rare, herpesvirus alternative promoter usage was recently found to be far more pervasive and impactful than previously thought. Here we review several examples of promoter switching in herpesviruses and discuss the functional consequences on the transcriptional and post-transcriptional regulation of viral gene expression.
Collapse
Affiliation(s)
- Andrew E Hale
- Department of Microbiology and Immunology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA;
| | - Nathaniel J Moorman
- Department of Microbiology and Immunology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA;
| |
Collapse
|
143
|
García-Mato Á, Cervantes B, Murillo-Cuesta S, Rodríguez-de la Rosa L, Varela-Nieto I. Insulin-like Growth Factor 1 Signaling in Mammalian Hearing. Genes (Basel) 2021; 12:genes12101553. [PMID: 34680948 PMCID: PMC8535591 DOI: 10.3390/genes12101553] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 02/06/2023] Open
Abstract
Insulin-like growth factor 1 (IGF-1) is a peptide hormone belonging to the insulin family of proteins. Almost all of the biological effects of IGF-1 are mediated through binding to its high-affinity tyrosine kinase receptor (IGF1R), a transmembrane receptor belonging to the insulin receptor family. Factors, receptors and IGF-binding proteins form the IGF system, which has multiple roles in mammalian development, adult tissue homeostasis, and aging. Consequently, mutations in genes of the IGF system, including downstream intracellular targets, underlie multiple common pathologies and are associated with multiple rare human diseases. Here we review the contribution of the IGF system to our understanding of the molecular and genetic basis of human hearing loss by describing, (i) the expression patterns of the IGF system in the mammalian inner ear; (ii) downstream signaling of IGF-1 in the hearing organ; (iii) mouse mutations in the IGF system, including upstream regulators and downstream targets of IGF-1 that inform cochlear pathophysiology; and (iv) human mutations in these genes causing hearing loss.
Collapse
Affiliation(s)
- Ángela García-Mato
- Institute for Biomedical Research “Alberto Sols” (IIBm), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; (Á.G.-M.); (B.C.); (S.M.-C.)
- Rare Diseases Networking Biomedical Research Centre (CIBERER), CIBER, Carlos III Institute of Health, 28029 Madrid, Spain
| | - Blanca Cervantes
- Institute for Biomedical Research “Alberto Sols” (IIBm), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; (Á.G.-M.); (B.C.); (S.M.-C.)
- Rare Diseases Networking Biomedical Research Centre (CIBERER), CIBER, Carlos III Institute of Health, 28029 Madrid, Spain
| | - Silvia Murillo-Cuesta
- Institute for Biomedical Research “Alberto Sols” (IIBm), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; (Á.G.-M.); (B.C.); (S.M.-C.)
- Rare Diseases Networking Biomedical Research Centre (CIBERER), CIBER, Carlos III Institute of Health, 28029 Madrid, Spain
- La Paz Hospital Institute for Health Research (IdiPAZ), 28046 Madrid, Spain
| | - Lourdes Rodríguez-de la Rosa
- Rare Diseases Networking Biomedical Research Centre (CIBERER), CIBER, Carlos III Institute of Health, 28029 Madrid, Spain
- La Paz Hospital Institute for Health Research (IdiPAZ), 28046 Madrid, Spain
- Correspondence: (L.R.-d.l.R.); (I.V.-N.)
| | - Isabel Varela-Nieto
- Institute for Biomedical Research “Alberto Sols” (IIBm), Spanish National Research Council-Autonomous University of Madrid (CSIC-UAM), 28029 Madrid, Spain; (Á.G.-M.); (B.C.); (S.M.-C.)
- Rare Diseases Networking Biomedical Research Centre (CIBERER), CIBER, Carlos III Institute of Health, 28029 Madrid, Spain
- La Paz Hospital Institute for Health Research (IdiPAZ), 28046 Madrid, Spain
- Correspondence: (L.R.-d.l.R.); (I.V.-N.)
| |
Collapse
|
144
|
Chen ACH, Peng Q, Fong SW, Lee KC, Yeung WSB, Lee YL. DNA Damage Response and Cell Cycle Regulation in Pluripotent Stem Cells. Genes (Basel) 2021; 12:genes12101548. [PMID: 34680943 PMCID: PMC8535646 DOI: 10.3390/genes12101548] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 01/30/2023] Open
Abstract
Pluripotent stem cells (PSCs) hold great promise in cell-based therapy because of their pluripotent property and the ability to proliferate indefinitely. Embryonic stem cells (ESCs) derived from inner cell mass (ICM) possess unique cell cycle control with shortened G1 phase. In addition, ESCs have high expression of homologous recombination (HR)-related proteins, which repair double-strand breaks (DSBs) through HR or the non-homologous end joining (NHEJ) pathway. On the other hand, the generation of induced pluripotent stem cells (iPSCs) by forced expression of transcription factors (Oct4, Sox2, Klf4, c-Myc) is accompanied by oxidative stress and DNA damage. The DNA repair mechanism of DSBs is therefore critical in determining the genomic stability and efficiency of iPSCs generation. Maintaining genomic stability in PSCs plays a pivotal role in the proliferation and pluripotency of PSCs. In terms of therapeutic application, genomic stability is the key to reducing the risks of cancer development due to abnormal cell replication. Over the years, we and other groups have identified important regulators of DNA damage response in PSCs, including FOXM1, SIRT1 and PUMA. They function through transcription regulation of downstream targets (P53, CDK1) that are involved in cell cycle regulations. Here, we review the fundamental links between the PSC-specific HR process and DNA damage response, with a focus on the roles of FOXM1 and SIRT1 on maintaining genomic integrity.
Collapse
Affiliation(s)
- Andy Chun Hang Chen
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China; (A.C.H.C.); (S.W.F.); (K.C.L.)
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong Shenzhen Hospital, Shenzhen 518009, China;
| | - Qian Peng
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong Shenzhen Hospital, Shenzhen 518009, China;
| | - Sze Wan Fong
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China; (A.C.H.C.); (S.W.F.); (K.C.L.)
| | - Kai Chuen Lee
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China; (A.C.H.C.); (S.W.F.); (K.C.L.)
| | - William Shu Biu Yeung
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China; (A.C.H.C.); (S.W.F.); (K.C.L.)
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong Shenzhen Hospital, Shenzhen 518009, China;
- Correspondence: (W.S.B.Y.); (Y.L.L.)
| | - Yin Lau Lee
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China; (A.C.H.C.); (S.W.F.); (K.C.L.)
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong Shenzhen Hospital, Shenzhen 518009, China;
- Correspondence: (W.S.B.Y.); (Y.L.L.)
| |
Collapse
|
145
|
Human FoxP Transcription Factors as Tractable Models of the Evolution and Functional Outcomes of Three-Dimensional Domain Swapping. Int J Mol Sci 2021; 22:ijms221910296. [PMID: 34638644 PMCID: PMC8508939 DOI: 10.3390/ijms221910296] [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: 09/06/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 01/18/2023] Open
Abstract
The association of two or more proteins to adopt a quaternary complex is one of the most widespread mechanisms by which protein function is modulated. In this scenario, three-dimensional domain swapping (3D-DS) constitutes one plausible pathway for the evolution of protein oligomerization that exploits readily available intramolecular contacts to be established in an intermolecular fashion. However, analysis of the oligomerization kinetics and thermodynamics of most extant 3D-DS proteins shows its dependence on protein unfolding, obscuring the elucidation of the emergence of 3D-DS during evolution, its occurrence under physiological conditions, and its biological relevance. Here, we describe the human FoxP subfamily of transcription factors as a feasible model to study the evolution of 3D-DS, due to their significantly faster dissociation and dimerization kinetics and lower dissociation constants in comparison to most 3D-DS models. Through the biophysical and functional characterization of FoxP proteins, relevant structural aspects highlighting the evolutionary adaptations of these proteins to enable efficient 3D-DS have been ascertained. Most biophysical studies on FoxP suggest that the dynamics of the polypeptide chain are crucial to decrease the energy barrier of 3D-DS, enabling its fast oligomerization under physiological conditions. Moreover, comparison of biophysical parameters between human FoxP proteins in the context of their minute sequence differences suggests differential evolutionary strategies to favor homoassociation and presages the possibility of heteroassociations, with direct impacts in their gene regulation function.
Collapse
|
146
|
Wu Z, Li Y, Niu Y, Lu J, Yan Z, Xu T, Guo Y, Dong Z, Guo W. FOXD3 suppresses epithelial-mesenchymal transition through direct transcriptional promotion of SMAD7 in esophageal squamous cell carcinoma. Mol Carcinog 2021; 60:859-873. [PMID: 34551139 DOI: 10.1002/mc.23350] [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: 04/23/2021] [Revised: 09/01/2021] [Accepted: 09/02/2021] [Indexed: 02/06/2023]
Abstract
The transcription factor forkhead box D3 (FOXD3) is an important member of the FOX family, which can maintain the pluripotent properties of cell clusters, neural crest, and trophoblastic progenitor cells in vivo. It has been shown that FOXD3 could affect proliferation, migration, and angiogenesis of various tumors and its deletion and overexpression in organisms will undoubtedly have important influence on the change of cell fate and the occurrence of tumors. However, the underlying functions and molecular mechanisms of FOXD3 in esophageal squamous cell carcinoma (ESCC) have not been fully clarified. According to the present study, the expression levels and functional roles of FOXD3 were investigated, and its prognostic value and molecular mechanisms in tumorigenesis and progression of ESCC were clarified. The expression level of FOXD3 was significantly downregulated in ESCC tissues and cell lines, and correlated with gender, family history of upper gastrointestinal cancer, TNM stage, depth of invasion, lymph node metastasis, and ESCC patients' survival. Moreover, FOXD3 inhibited cells migration and invasion as well as participated in TGF-β1 induced epithelial-mesenchymal transition process. Furthermore, a positive correlation between FOXD3 and SMAD family member 7 (SMAD7) was explored in ESCC. FOXD3 could directly bind to promoter regions of SMAD7 gene, leading to transcriptional promotion of SMAD7 in human esophageal cancer cells. Taken together, FOXD3 may play a tumor suppressor role in ESCC and may be applied as a new therapeutic target and prognostic marker for ESCC.
Collapse
Affiliation(s)
- Zheng Wu
- Laboratory of Pathology, Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yan Li
- Laboratory of Pathology, Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yunfeng Niu
- Laboratory of Pathology, Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Juntao Lu
- Laboratory of Pathology, Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhaoyang Yan
- Laboratory of Pathology, Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Tongxin Xu
- Laboratory of Pathology, Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yanli Guo
- Laboratory of Pathology, Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhiming Dong
- Laboratory of Pathology, Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Wei Guo
- Laboratory of Pathology, Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
147
|
Zhao H, Chen W, Zhu Y, Lou J. Hypoxia promotes pancreatic cancer cell migration, invasion, and epithelial-mesenchymal transition via modulating the FOXO3a/DUSP6/ERK axis. J Gastrointest Oncol 2021; 12:1691-1703. [PMID: 34532120 DOI: 10.21037/jgo-21-359] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 07/22/2021] [Indexed: 01/05/2023] Open
Abstract
Background Pancreatic cancer (PC) is among the most aggressive types of cancer. Hypoxia has been identified as a key risk factor for cancer progression. The forkhead box (FOX) proteins are multidirectional transcriptional factors that are strongly implicated in malignancies. However, whether FOXO3a, a member of the FOX protein family, is involved in the pro-oncogenic functions of hypoxia in PC has remained largely unelucidated. In this study, we attempted to clarify the role of FOXO3a in metastasis under hypoxic conditions and its underlying mechanism. Methods MTT and flow cytometry assays were performed to detect the cell proliferation and cell cycle distribution respectively. Transwell assays were used to determine the potential of cell migration and invasion. qPCR and western blot assays were used to assess the expression of mRNA and protein. Immunofluorescence assay was performed to evaluate the cellular localization of FOXO3a. FOXO3a overexpression plasmid was constructed to perform the rescue experiment. Results Our data indicated that PANC-1 and SW1990 cells represented enhanced cell migration and invasion abilities under hypoxia, while no statistical differences in cell proliferation and cell cycle distribution were observed. Hypoxia upregulated the messenger RNA (mRNA) and protein expressions of HIF-1α, FOXO3a, and the key epithelial-mesenchymal transition (EMT)-related (EMT) molecules N-cadherin and vimentin, as well as the phosphorylation of FOXO3a. Interestingly, hypoxia promoted the extranuclear localization of FOXO3a. Overexpression of FOXO3a not only significantly decreased the invasion, migration, and EMT of PC cell lines, but also reversed hypoxia-induced extranuclear localization. Finally, FOXO3a might act as a tumor suppressor in PC by inhibiting the ERK signaling pathway by inducing DUSP6 expression, and the ERK activator fisetin could effectively attenuate the inhibitory role of FOXO3a on ERK. Conclusions Taken together, our results identified that hypoxia-induced extranuclear localization of FOXO3a promoted cell migration and invasion of human PC by modulating the DUSP6/ERK pathway.
Collapse
Affiliation(s)
- Hua Zhao
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Wei Chen
- The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Yi Zhu
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianying Lou
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
148
|
Elaidy NF, Harb OA, Mohamed AM, Hemeda R, Taha HF, Samir A, Elsayed AM, Osman G, Hendawy EIE. Prognostic Significances of NEDD-9 and FOXL-1 Expression in Intestinal Type Gastric Carcinoma: an Immunohistochemical Study. J Gastrointest Cancer 2021; 52:728-737. [PMID: 32794109 DOI: 10.1007/s12029-020-00471-3] [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] [Indexed: 12/24/2022]
Abstract
BACKGROUND Gastric cancer (GC) is mostly diagnosed at advanced stage, so prognosis is poor. Therefore, it is necessary to understand the molecular mechanism of GC development to design new targeted treatment to improve the prognosis of gastric cancer patients. AIM OF THE WORK To assess the prognostic value of NEDD-9 and FOXL-1 expression in intestinal type gastric cancer patients, as well as their relationship to clinicopathologic features of the disease and patients outcome. PATIENTS AND METHODS This is a retrospective study; we included 50 sections from formalin-fixed, paraffin-embedded tissue samples which included intestinal type GC and adjacent non-neoplastic gastric mucosa in the same block that were subjected to immunohistochemistry with anti-NEDD-9 and anti-FOXL-1 antibody. Patients were retrospectively followed up for about 5 years for assessment of tumor progression and survival in relation to marker expression. RESULTS High NEDD-9 and low FOXL-1 expression were found in intestinal type GC more than adjacent non-neoplastic mucosa (p < 0.001). NEDD-9 high expression and FOXL-1 low expression were associated with presence of helicobacter pylori gastritis (p = 0.010, 0.049), high grade (p = 0.007, 0.004), high stage (p < 0.001), presence of distant metastases (p = 0.029, 0.021), poor DFS (p = 0.003), and OS rates (< 0.001). CONCLUSION NEDD-9 overexpression and FOXL-1 deficiency in intestinal type GC can help in prediction of tumor prognosis and it can guide the selection of patients for future therapies in gastric carcinoma.
Collapse
Affiliation(s)
- Noha F Elaidy
- Department of Pathology, Faculty of Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Ola A Harb
- Department of Pathology, Faculty of Medicine, Zagazig University, Zagazig, 44519, Egypt.
| | - Abdel Motaleb Mohamed
- Department of Clinical Oncology& Nuclear Medicine, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Rehab Hemeda
- Department of Clinical Oncology& Nuclear Medicine, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Heba F Taha
- Department of Medical Oncology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Amr Samir
- Department of Internal Medicine, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Ahmed M Elsayed
- Department of Tropical Medicine, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Gamal Osman
- Department of General Surgery, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Elsayed I El Hendawy
- Department of General Surgery, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
149
|
Lin HY, Zhu CQ, Zhang HH, Shen ZC, Zhang CX, Ye YX. The Genetic Network of Forkhead Gene Family in Development of Brown Planthoppers. BIOLOGY 2021; 10:867. [PMID: 34571744 PMCID: PMC8469257 DOI: 10.3390/biology10090867] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 11/16/2022]
Abstract
We identified 18 distinct Fox genes in the genome of the brown planthopper, Nilaparvata lugens, and further found a novel insect-specific subfamily that we temporarily named FoxT. A total of 16 genes were highly expressed in the eggs, while NlFoxL2 and NlFoxT are female- and male-specific genes, respectively. Large scale RNAi and RNA-seq analyses were used to reveal the functions and potential targets of NlFoxs. In the eggs, NlFoxA, NlFoxN1 and NlFoxN2 are indispensable to early embryogenesis by regulating different target genes; NlFoxG and NlFoxQ co-regulate NlSix3 for brain development; and NlFoxC, NlFoxJ1 and NlFoxP have complementary effects on late embryogenesis. Moreover, NlFoxA, NlFoxNl and NlFoxQ have pleiotropism. NlFoxA and NlFoxQ regulate the expression of NlCHS1 and cuticular proteins, respectively, thereby participating in the formation of cuticles. NlFoxN1, which regulates the expression of NlKrt9 is involved in the formation of intermediate filament frameworks. Our previous studies have revealed that NlFoxL2 and NlFoxO play important roles in chorion formation and wing polyphenism. Altogether, N. lugens Fox genes exhibit functional diversity in embryonic development and organogenesis. This comprehensive study combines genomics, transcriptomics and phenomics, thereby constructing a complex genetic network that spans the entire life cycle of the brown planthopper.
Collapse
Affiliation(s)
- Hai-Yan Lin
- The Rural Development Academy, Zhejiang University, Hangzhou 310058, China; (H.-Y.L.); (C.-Q.Z.)
- Institute of Insect Sciences, Zhejiang University, Hangzhou 310058, China; (H.-H.Z.); (Z.-C.S.); (C.-X.Z.)
| | - Cheng-Qi Zhu
- The Rural Development Academy, Zhejiang University, Hangzhou 310058, China; (H.-Y.L.); (C.-Q.Z.)
- Institute of Insect Sciences, Zhejiang University, Hangzhou 310058, China; (H.-H.Z.); (Z.-C.S.); (C.-X.Z.)
| | - Hou-Hong Zhang
- Institute of Insect Sciences, Zhejiang University, Hangzhou 310058, China; (H.-H.Z.); (Z.-C.S.); (C.-X.Z.)
| | - Zhi-Cheng Shen
- Institute of Insect Sciences, Zhejiang University, Hangzhou 310058, China; (H.-H.Z.); (Z.-C.S.); (C.-X.Z.)
| | - Chuan-Xi Zhang
- Institute of Insect Sciences, Zhejiang University, Hangzhou 310058, China; (H.-H.Z.); (Z.-C.S.); (C.-X.Z.)
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Plant Virology, Ningbo University, Ningbo 315211, China
| | - Yu-Xuan Ye
- The Rural Development Academy, Zhejiang University, Hangzhou 310058, China; (H.-Y.L.); (C.-Q.Z.)
- Institute of Insect Sciences, Zhejiang University, Hangzhou 310058, China; (H.-H.Z.); (Z.-C.S.); (C.-X.Z.)
| |
Collapse
|
150
|
Siques P, Pena E, Brito J, El Alam S. Oxidative Stress, Kinase Activation, and Inflammatory Pathways Involved in Effects on Smooth Muscle Cells During Pulmonary Artery Hypertension Under Hypobaric Hypoxia Exposure. Front Physiol 2021; 12:690341. [PMID: 34434114 PMCID: PMC8381601 DOI: 10.3389/fphys.2021.690341] [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: 04/02/2021] [Accepted: 07/16/2021] [Indexed: 12/23/2022] Open
Abstract
High-altitude exposure results in hypobaric hypoxia, which affects organisms by activating several mechanisms at the physiological, cellular, and molecular levels and triggering the development of several pathologies. One such pathology is high-altitude pulmonary hypertension (HAPH), which is initiated through hypoxic pulmonary vasoconstriction to distribute blood to more adequately ventilated areas of the lungs. Importantly, all layers of the pulmonary artery (adventitia, smooth muscle, and endothelium) contribute to or are involved in the development of HAPH. However, the principal action sites of HAPH are pulmonary artery smooth muscle cells (PASMCs), which interact with several extracellular and intracellular molecules and participate in mechanisms leading to proliferation, apoptosis, and fibrosis. This review summarizes the alterations in molecular pathways related to oxidative stress, inflammation, kinase activation, and other processes that occur in PASMCs during pulmonary hypertension under hypobaric hypoxia and proposes updates to pharmacological treatments to mitigate the pathological changes in PASMCs under such conditions. In general, PASMCs exposed to hypobaric hypoxia undergo oxidative stress mediated by Nox4, inflammation mediated by increases in interleukin-6 levels and inflammatory cell infiltration, and activation of the protein kinase ERK1/2, which lead to the proliferation of PASMCs and contribute to the development of hypobaric hypoxia-induced pulmonary hypertension.
Collapse
Affiliation(s)
- Patricia Siques
- Institute of Health Studies, Arturo Prat University, Iquique, Chile
| | - Eduardo Pena
- Institute of Health Studies, Arturo Prat University, Iquique, Chile
| | - Julio Brito
- Institute of Health Studies, Arturo Prat University, Iquique, Chile
| | - Samia El Alam
- Institute of Health Studies, Arturo Prat University, Iquique, Chile
| |
Collapse
|