650 results on '"FOXA2"'
Search Results
2. Chromatin confinement enhances target search of pioneer transcription factors in live cells
- Author
-
Wang, Zuhui, Niu, Di, Wang, Bo, Bi, Ying, Yin, Chao, Cattoglio, Claudia, Loh, Kyle M, Ge, Hao, and Deng, Wulan
- Subjects
single-molecule tracking ,pioneer transcription factor ,FOXA2 ,chromatin domain ,intrinsically disordered region ,transcription factor ,confined target search - Abstract
SMT trajectory data for “Chromatin confinement enhances target search of pioneer transcription factors in live cells” Zuhui Wang1,2†, Di Niu1,2,3†, Bo Wang1,2,3†, Ying Bi1,2, Chao Yin1,2,3, Claudia Cattoglio4,5, Kyle M Loh6, Hao Ge1, Wulan Deng1,2,3, * 1Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing 100871, China. 2Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China. 3Peking-Tsinghua Center for Life Sciences (CLS), Peking University, Beijing 100871, China. 4Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA. 5Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA. 6 Institute for Stem Cell Biology and Regenerative Medicine and Ludwig Center for Cancer Stem Cell Biology and Medicine, Departments of Pathology and Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA. † These authors contributed equally to this work. *Corresponding author: wdeng@pku.edu.cn Overview This repository contains all the SMT trajectory data associated with “Confined target search powers FOXA2 transcription activity”. In the repository, there are two main folders. The folder ‘slowSMT_trajectories_pooled’ contains pooled slowSMT trajectories that we used to calculate residence times. The folder ‘spaSMT_trajectories_single_cells’ contains spaSMT single-cell trajectories, with hESC/APS/DE, MEF and U2OS trajectories in the separate folders. We used the spaSMT data in the “hESC_APS_DE” and “MEF” folder for both Spot-On analysis and Anisotropy analysis, while for the spaSMT data in the “U2OS” folder, we used indicated subfolders for Spot-On analysis and/or Anisotropy analysis. For each pooled trajectory file in the folder ‘slowSMT_trajectories_pooled’, it contains two matlab variables. trackedPar: each element represents one trajectory containing one or more localizations xy: x- and y-coordinates in micrometers of the molecule localizations Frame: frame indexes of the molecule localizations. TimeStamp: timepoints in seconds of the molecule localizations. sampleinfo: names of individual cell samples. For each single cell trajectory file in the folder ‘spaSMT_trajectories_single_cells’, it contains two matlab variables. trackedPar: same as above. settings: contains file information, image acquisition settings, the algorithm settings used in MTT localization and tracking software. For detailed explanation, please see the MTT code at https://gitlab.com/tjian-darzacq-lab/SPT_LocAndTrack. The document does not contain information about how the data was analyzed. For details on how the data was analyzed and for codes to reproduce our figure, please go to our github repository. For the slowSMT analysis, we deposit the code under: https://github.com/denglabpku/slowSMT. For the anisotropy analysis, we deposit the code under: https://github.com/denglabpku/anisotropy. For the Spot-On analysis, please visit the Spot-On website: https://spoton.berkeley.edu/. Our trajectory file format is directly available as the input of the Spot-On software.  
- Published
- 2023
- Full Text
- View/download PDF
3. FOXA1/2 depletion drives global reprogramming of differentiation state and metabolism in a human liver cell line and inhibits differentiation of human stem cell-derived hepatic progenitor cells
- Author
-
Iyan Warren, Michael M. Moeller, Daniel Guiggey, Alexander Chiang, Mitchell Maloy, Ogechi Ogoke, Theodore Groth, Tala Mon, Saber Meamardoost, Xiaojun Liu, Sarah Thompson, Antoni Szeglowski, Ryan Thompson, Peter Chen, Ramasamy Paulmurugan, Martin L. Yarmush, Srivatsan Kidambi, and Natesh Parashurama
- Subjects
Homeobox protein NANOG ,Mesoderm ,Organogenesis ,Germ layer ,Biology ,Biochemistry ,Cell biology ,medicine.anatomical_structure ,Downregulation and upregulation ,embryonic structures ,medicine ,Genetics ,FOXA2 ,Stem cell ,Endoderm ,Molecular Biology ,Biotechnology - Abstract
FOXA factors are critical members of the developmental gene regulatory network (GRN) composed of master transcription factors (TF) which regulate murine cell fate and metabolism in the gut and liver. How FOXA dictates human liver cell fate, differentiation, and simultaneously regulate metabolic pathways is poorly understood. Here, we aimed to determine the role of FOXA2 (and FOXA1 which is believed to compensate for FOXA2) in hepatic differentiation and cell metabolism in a human hepatic cell line (HepG2). siRNA targeting of FOXA1 and FOXA2 in human hepatic (HepG2) cells and during hepatic differentiation significantly downregulated albumin (p < 0.05) and GRN TF gene expression (HNF4A, HEX, HNF1B, TBX3) (p < 0.05) and significantly upregulated endoderm/gut/hepatic endoderm markers (goosecoid (GSC), FOXA3, and GATA4), gut TF (CDX2), pluripotent TF (NANOG), and neuroectodermal TF (PAX6) (p < 0.05), all consistent with a partial/transient cell reprogramming. shFOXA1/2 targeting resulted in similar findings and demonstrated evidence of reversibility. RNA-seq followed by bioinformatic analysis of shFOXA1/2 knockdown HepG2 cells demonstrated 235 significant downregulated genes and 448 upregulated genes, including upregulation of markers for alternate germ layers lineages (cardiac, endothelial, muscle) and neurectoderm (eye, neural). We found widespread downregulation of glycolysis, citric acid cycle, mitochondrial genes, and alterations in lipid metabolism, pentose phosphate pathway, and ketogenesis. Functional metabolic analysis agreed with these findings, demonstrating significantly diminished glycolysis and mitochondrial respiration, and accumulation of lipid droplets. We hypothesized that FOXA1/2 inhibit the initiation of human liver differentiation in vitro. During hPSC-hepatic differentiation, siRNA knockdown demonstrated de-differentiation and unexpectedly, activation of pluripotency factors and neuroectoderm. shRNA knockdown demonstrated similar results and activation of SOX9 (hepatobiliary). These results demonstrate complex effects of FOXA1/2 on hepatic GRN effecting de-differentiation and metabolism with applications in studies of cancer, differentiation, and organogenesis.
- Published
- 2022
4. CD44 Promotes Breast Cancer Metastasis through AKT-Mediated Downregulation of Nuclear FOXA2
- Author
-
Anupama Vadhan, Ming-Feng Hou, Priya Vijayaraghavan, Yi-Chia Wu, Stephen Chu-Sung Hu, Yun-Ming Wang, Tian-Lu Cheng, Yen-Yun Wang, and Shyng-Shiou F. Yuan
- Subjects
Medicine (miscellaneous) ,General Biochemistry, Genetics and Molecular Biology ,breast cancer ,CD44 ,FOXA2 ,AKT ,metastasis - Abstract
The primary cause of breast cancer mortality is the metastatic invasion of cancerous stem cells (CSC). Cluster of differentiation 44 (CD44) is a well-known CSC marker in various cancers, as well as a key role player in metastasis and relapse of breast cancer. CD44 is a cell-membrane embedded protein, and it interacts with different proteins to regulate cancer cell behavior. Transcription factor forkhead box protein A2 (FOXA2) acts as an important regulator in multiple cancers, including breast cancer. However, the biological significance of CD44-FOXA2 association in breast cancer metastasis remains unclear. Herein, we observed that CD44 expression was higher in metastatic lymph nodes compared to primary tumors using a flow cytometric analysis. CD44 overexpression in breast cancer cell lines significantly promoted cell migration and invasion abilities, whereas the opposite effects occurred upon the knockdown of CD44. The stem cell array analysis revealed that FOXA2 expression was upregulated in CD44 knockdown cells. However, the knockdown of FOXA2 in CD44 knockdown cells reversed the effects on cell migration and invasion. Furthermore, we found that CD44 mediated FOXA2 localization in breast cancer cells through the AKT pathway. Moreover, the immunofluorescence assay demonstrated that AKT inhibitor wortmannin and AKT activator SC79 treatment in breast cancer cells impacted FOXA2 localization. Collectively, this study highlights that CD44 promotes breast cancer metastasis by downregulating nuclear FOXA2.
- Published
- 2022
- Full Text
- View/download PDF
5. FoXA2 promotes esophageal squamous cell carcinoma progression by ZEB2 activation
- Author
-
Haiyan Sun, Zheng Yan, Yanfang Chen, and Hanjing Gao
- Subjects
Esophageal Neoplasms ,RD1-811 ,Esophageal squamous cell carcinoma ,Surgical oncology ,Transcription (biology) ,Cell Movement ,Cell Line, Tumor ,Medicine ,Humans ,Neoplasm Invasiveness ,neoplasms ,reproductive and urinary physiology ,RC254-282 ,Cell Proliferation ,Zinc Finger E-box Binding Homeobox 2 ,ZEB2 ,Gene knockdown ,Progression ,Cell growth ,business.industry ,Research ,ESCC ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,respiratory system ,Prognosis ,digestive system diseases ,Blot ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,Oncology ,Head and Neck Neoplasms ,embryonic structures ,Cancer research ,Hepatocyte Nuclear Factor 3-beta ,Surgery ,FOXA2 ,Esophageal Squamous Cell Carcinoma ,business ,Chromatin immunoprecipitation - Abstract
Background It has been reported that Forkhead transcription family member (FOXA2) regulates esophageal squamous cell carcinoma (ESCC) progression. However, the specific mechanism, by which FOXA2 promotes ESCC malignant progression, remains unclear. Materials and methods QRT-PCR and western blotting were applied to measure FOXA2 expression in ESCC tissues, while CCK-8 assay and Transwell assays were used to investigate the effect of FOXA2 on ESCC. Luciferase reporter assay, followed by fast chromatin immunoprecipitation (ChIP) assay, was used to study the relationship between FOXA2 and ZEB2. Results FOXA2 was significantly increased in ESCC tissues, when compared to normal tissues. Moreover, high expression of FOXA2 was also found in ESCC cells. Knockdown of FOXA2 inhibited ESCC cell proliferation, invasion, and migration. Mechanically, FOXA2 was verified to regulate ZEB2 expression at transcription level. Moreover, ZEB2 reversed the inhibitory effect of FOXA2 on ESCC proliferation, invasion, and migration. The relationship between ZEB2 and FOXA2 in ESCC tissues was negative. Conclusions These results indicate that FOXA2 plays a critical role in ESCC progression and may become a potential candidate target for ESCC treatment.
- Published
- 2021
6. Fat mass and obesity‐associated protein regulates tumorigenesis of arecoline‐promoted human oral carcinoma
- Author
-
Xia Li, Jiang Song, Xiaoli Xie, Yilong Ai, Jianming Zhang, Yijun Gao, Xiufeng Cheng, and Yangcong Gu
- Subjects
Cancer Research ,Carcinogenesis ,medicine.disease_cause ,oral carcinoma ,Nuts ,RC254-282 ,Research Articles ,Cancer Biology ,Areca ,biology ,digestive, oral, and skin physiology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,Oncology ,Gene Knockdown Techniques ,Hepatocyte Nuclear Factor 3-beta ,Mouth Neoplasms ,FTO ,Research Article ,medicine.drug ,Arecoline ,Alpha-Ketoglutarate-Dependent Dioxygenase FTO ,Downregulation and upregulation ,Cell Line, Tumor ,medicine ,Carcinoma ,Humans ,Radiology, Nuclear Medicine and imaging ,Carcinogen ,Squamous Cell Carcinoma of Head and Neck ,business.industry ,Mouth Mucosa ,nutritional and metabolic diseases ,Cancer ,Methyltransferases ,biology.organism_classification ,medicine.disease ,tumorigenesis ,stomatognathic diseases ,Drug Resistance, Neoplasm ,Case-Control Studies ,Cancer research ,FOXA2 ,Cisplatin ,business - Abstract
Arecoline, a major alkaloid within areca nut extract, is recognized as the primary active carcinogen promoting oral squamous cell carcinoma (OSCC) pathological development. Dysregulation of N6‐methyladenosine (m6A) methyltransferase components (e.g., Fat mass and obesity‐associated protein [FTO] and methyltransferase‐like 3 [METTL3]) are closely associated with multiple cancer progression, including oral cancer. However, the biological function role of FTO in arecoline‐induced oral cancer is largely unknown. We identified that FTO was significantly upregulated in OSCC tissues from patients with areca nut chewing habits and chronic arecoline‐treated OSCC cell lines. Depletion of FTO attenuated the arecoline‐promoted stemness, chemoresistance, and oncogenicity of OSCC cells. Finally, we revealed that FTO was negatively regulated by a transcription factor forkhead box protein A2 (FOXA2) in OSCC cells. This study, for the first time, demonstrated that FTO plays an oncogenic role in arecoline‐induced OSCC progression. Thus, developing new therapeutic agents targeting FTO may serve as a promising method to treatment OSCC patients, especially those with areca nut chewing habits., We identified that FTO was significantly upregulated in OSCC tissues from patients with areca nut chewing habits and chronic arecoline‐treated OSCC cell lines. Depletion of FTO attenuated the arecoline‐promoted stemness, chemoresistance, and oncogenicity of OSCC cells. Therapeutic agents targeting FTO may serve as a promising method to treat OSCC patients, especially those with areca nut chewing habits.
- Published
- 2021
- Full Text
- View/download PDF
7. FOXA2 inhibits doxorubicin-induced apoptosis via transcriptionally activating HBP rate-limiting enzyme GFPT1 in HCC cells
- Author
-
Yuhan Wang, Ang Yu, Yangzhi Liu, Qiong Wu, Yubo Liu, Jianing Zhang, Tianmiao Huang, Meiyun Shi, Huang Huang, Lingyan Wang, Wenli Li, and Xiaoyu Wang
- Subjects
Carcinoma, Hepatocellular ,Physiology ,Apoptosis ,medicine.disease_cause ,Biochemistry ,Downregulation and upregulation ,medicine ,Humans ,Doxorubicin ,Transcription factor ,reproductive and urinary physiology ,Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing) ,Chemistry ,Liver Neoplasms ,Hexosamines ,General Medicine ,respiratory system ,Biosynthetic Pathways ,Cancer cell ,Hepatocyte Nuclear Factor 3-beta ,Cancer research ,FOXA2 ,Carcinogenesis ,Protein Processing, Post-Translational ,Intracellular ,medicine.drug - Abstract
Apoptosis plays an important role in both carcinogenesis and cancer treatment. Understanding the mechanisms through which resistance to apoptosis occurs in cancer cells has huge implications for cancer treatment. Although pieces of evidence have shown that elevated levels of global O-GlcNAcylation play an anti-apoptotic role in myriad cancers, the underlying mechanism is still ambiguous. In this study, we demonstrated that FOXA2, an essential transcription factor for liver homeostasis and hepatocellular carcinoma (HCC) development, inhibits doxorubicin (DOX)-induced apoptosis through elevating cellular O-GlcNAcylation in HCC cells. In response to DOX treatment, elevated FOXA2 and global O-GlcNAcylation level was observed in HCC cells, and higher FOXA2 levels indicated lower levels of DOX-induced apoptosis. Subsequently, we demonstrated that FOXA2 is a direct transcriptional activator of the hexosamine biosynthetic pathway (HBP) rate-limiting enzyme GFPT1. The upregulation of FOXA2 expression induced the synthesis of intracellular UDP-GlcNAc, which is the sugar substrate of O-GlcNAcylation produced by the HBP. The flux through the HBP elevated the global O-GlcNAcylation level and led to the activation of survival signaling pathways in HCC cells. Furthermore, GFPT1 was proved to be an important downstream regulator of FOXA2-mediated apoptotic suppression. These results provide insights into the molecular mechanism by which FOXA2 inhibits DOX-induced HCC cell apoptosis and suggest that targeting FOXA2 might offer a new strategy for HCC treatment.
- Published
- 2021
- Full Text
- View/download PDF
8. The evolutionarily conserved long non‐coding RNA LINC00261 drives neuroendocrine prostate cancer proliferation and metastasis via distinct nuclear and cytoplasmic mechanisms
- Author
-
Noushin Nabavi, Yinglei Li, Mustapha Jaber, Yuzhuo Wang, Elena Jachetti, Hui Xue, Igor Ulitsky, Perla Pucci, Mario P. Colombo, Crystal N. Marconett, Abhijit Parolia, Rebecca L. Mather, David Roig-Carles, Wei Jiang, Ilaria Alborelli, Rebecca Wu, Dong Lin, Cheryl A. Hawkes, Xinpei Ci, Ignacio A. Romero, Erik Venalainen, Francesco Crea, Shih-Chun Chu, Sandra E. Carson, Luca Quagliata, and Hardev Pandha
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Cytoplasm ,Biology ,Metastasis ,Transcriptome ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Downregulation and upregulation ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,long noncoding RNA ,Gene ,Research Articles ,RC254-282 ,Cell Proliferation ,Gene knockdown ,neuroendocrine prostate cancer ,Prostate ,Prostatic Neoplasms ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,General Medicine ,medicine.disease ,Long non-coding RNA ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,CBX2 ,030220 oncology & carcinogenesis ,Cancer research ,Molecular Medicine ,RNA, Long Noncoding ,FOXA2 ,LINC00261 ,Research Article - Abstract
Metastatic neuroendocrine prostate cancer (NEPC) is a highly aggressive disease, whose incidence is rising. Long noncoding RNAs (lncRNAs) represent a large family of disease‐ and tissue‐specific transcripts, most of which are still functionally uncharacterized. Thus, we set out to identify the highly conserved lncRNAs that play a central role in NEPC pathogenesis. To this end, we performed transcriptomic analyses of donor‐matched patient‐derived xenograft models (PDXs) with immunohistologic features of prostate adenocarcinoma (AR+/PSA+) or NEPC (AR−/SYN+/CHGA+) and through differential expression analyses identified lncRNAs that were upregulated upon neuroendocrine transdifferentiation. These genes were prioritized for functional assessment based on the level of conservation in vertebrates. Here, LINC00261 emerged as the top gene with over 3229‐fold upregulation in NEPC. Consistently, LINC00261 expression was significantly upregulated in NEPC specimens in multiple patient cohorts. Knockdown of LINC00261 in PC‐3 cells dramatically attenuated its proliferative and metastatic abilities, which are explained by parallel downregulation of CBX2 and FOXA2 through distinct molecular mechanisms. In the cell cytoplasm, LINC00261 binds to and sequesters miR‐8485 from targeting the CBX2 mRNA, while inside the nucleus, LINC00261 functions as a transcriptional scaffold to induce SMAD‐driven expression of the FOXA2 gene. For the first time, these results demonstrate hyperactivation of the LINC00261‐CBX2‐FOXA2 axes in NEPC to drive proliferation and metastasis, and that LINC00261 may be utilized as a therapeutic target and a biomarker for this incurable disease., Neuroendocrine prostate cancer (NEPC) is a highly aggressive disease with high incidence of metastasis. Here, we identified LINC00261 among the top upregulated transcripts in NEPC. Inside the nucleus, LINC00261 promoted anchorage‐independent growth and metastatic gene programs by recruiting the SMAD2/3 transcriptional machinery to FOXA2 cis‐regulatory elements. Cytoplasmic LINC00261 blocked the binding of miR‐8485 to CBX2 and induced a CBX2‐driven proliferative gene program. Our study demonstrates the role of LINC00261‐CBX2‐FOXA2 axis in proliferation and metastasis of NEPC.
- Published
- 2021
9. Efficient generation of dopaminergic induced neuronal cells with midbrain characteristics
- Author
-
Soham Chanda, Marius Wernig, Nan Yang, Thomas C. Südhof, Yi Han Ng, Yuko Kokubu, and Justyna A. Janas
- Subjects
0301 basic medicine ,Cell type ,Tyrosine 3-Monooxygenase ,Dopamine ,Cell Culture Techniques ,Wnt1 Protein ,Biology ,Biochemistry ,Article ,iN cells ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Mesencephalon ,Basic Helix-Loop-Helix Transcription Factors ,Genetics ,Animals ,Humans ,mouse embyronic stem cells ,Glial Cell Line-Derived Neurotrophic Factor ,Induced pluripotent stem cell ,Transcription factor ,Embryonic Stem Cells ,transcription factor ,Brain-Derived Neurotrophic Factor ,Dopaminergic Neurons ,Dopaminergic ,reprogramming ,Cell Biology ,dopamine neurons ,human embryonic stem cells ,Embryonic stem cell ,Cell biology ,ASCL1 ,030104 developmental biology ,FOXA2 ,Reprogramming ,Biomarkers ,030217 neurology & neurosurgery ,Signal Transduction ,Transcription Factors ,Developmental Biology - Abstract
Summary The differentiation of pluripotent stem cells can be accomplished by sequential activation of signaling pathways or through transcription factor programming. Multistep differentiation imitates embryonic development to obtain authentic cell types, but it suffers from asynchronous differentiation with variable efficiency. Transcription factor programming induces synchronous and efficient differentiation with higher reproducibility but may not always yield authentic cell types. We systematically explored the generation of dopaminergic induced neuronal cells from mouse and human pluripotent stem cells. We found that the proneural factor Ascl1 in combination with mesencephalic factors Lmx1a and Nurr1 induce peripheral dopaminergic neurons. Co-delivery of additional midbrain transcription factors En1, FoxA2, and Pitx3 resulted in facile and robust generation of functional dopaminergic neurons of midbrain character. Our results suggest that more complex combinations of transcription factors may be needed for proper regional specification of induced neuronal cells generated by direct lineage induction., Graphical abstract, Highlights • Ascl1 alone can induce tyrosine hydroxylase (TH)-positive ES-iN cells • Ascl1 alone, or with Nurr1 and Lmx1a, induce peripheral TH-positive cells • WNT1 and neurotrophic factors increase TH-positive iN cells in culture • A 6-factor combination induces TH-positive dopamine iN cells of central identity, Dopaminergic neurons can be induced from pluripotent cells using overexpression of transcription factors. Existing transcription factor combinations result in cells of mixed regional identity. Here, Wernig and colleagues report that with extracellular signals, a six-factor combination delivered via piggyBac transposons is capable of inducing dopamine neurons of central identity from mouse and human embryonic stem cells.
- Published
- 2021
- Full Text
- View/download PDF
10. Inhibiting roles of FOXA2 in liver cancer cell migration and invasion by transcriptionally suppressing microRNA-103a-3p and activating the GREM2/LATS2/YAP axis
- Author
-
Jia Wang, Wenshan Chen, Jirong Chen, Tiantian Wei, and Guangzhen Ma
- Subjects
0301 basic medicine ,Chemistry ,Clinical Biochemistry ,Biomedical Engineering ,Bioengineering ,Cell migration ,Cell Biology ,respiratory system ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Downregulation and upregulation ,Cell culture ,030220 oncology & carcinogenesis ,embryonic structures ,microRNA ,Gene silencing ,Phosphorylation ,Original Article ,Luciferase ,FOXA2 ,reproductive and urinary physiology ,Biotechnology - Abstract
Forkhead box A2 (FOXA2) has emerged as a tumor inhibitor in several human malignancies. This work focused on the effect of FOXA2 on liver cancer (LC) cell invasion and migration and the involving molecules. FOXA2 expression in LC tissues and cell lines was determined. The potential target microRNA (miRNA) of FOXA2 was predicted via bioinformatic analysis and validated through a ChIP assay. The mRNA target of miRNA-103a-3p was predicted via bioinformatic analysis and confirmed via a luciferase assay. Altered expression of FOXA2, miR-103a-3p and GREM2 was introduced in cells to identify their roles in LC cell migration and invasion. Consequently, FOXA2 and GREM2 were poorly expressed while miR-103a-3p was highly expressed in LC samples. Overexpression of FOXA2 or GREM2 suppressed migration and invasion of LC cells, while up-regulation of miR-103a-3p led to inverse trends. FOXA2 transcriptionally suppressed miR-103a-3p to increase GREM2 expression. Silencing of GREM2 blocked the effects of FOXA2. GREM2 increased LATS2 activity and YAP phosphorylation and degradation. To conclude, this study demonstrated that FOXA2 suppressed miR-103a-3p transcription to induce GREM2 upregulation, which increased LATS2 activity and YAP phosphorylation to inhibit migration and invasion of LC cells.
- Published
- 2021
- Full Text
- View/download PDF
11. Dysgenesis and Dysfunction of the Pancreas and Pituitary Due to FOXA2 Gene Defects
- Author
-
Tulay Guran, Serap Turan, Sare Betul Kaygusuz, Bilgen Bilge Geçkinli, Maria Lillina Vignola, Esra Arslan Ates, Burcu Volkan, Carles Gaston-Massuet, and Abdullah Bereket
- Subjects
Male ,Transcriptional Activation ,Pituitary gland ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,Nonsense mutation ,Context (language use) ,Hypopituitarism ,Bioinformatics ,medicine.disease_cause ,Biochemistry ,Dysgenesis ,Endocrinology ,Internal medicine ,Diabetes Mellitus ,medicine ,Humans ,Pancreas ,Glucose Transporter Type 2 ,Mutation ,business.industry ,Biochemistry (medical) ,Infant ,Syndrome ,medicine.disease ,medicine.anatomical_structure ,Codon, Nonsense ,Pituitary Gland ,Hepatocyte Nuclear Factor 3-beta ,FOXA2 ,business ,Transcription Factors - Abstract
Context Developmental disorders of the pituitary gland leading to congenital hypopituitarism can either be isolated or associated with extrapituitary abnormalities (syndromic hypopituitarism). A large number of syndromic hypopituitarism cases are linked to mutations in transcription factors. The forkhead box A2 (FOXA2) is a transcription factor that plays a key role in the central nervous system, foregut, and pancreatic development. Objective This work aims to characterize 2 patients with syndromic hypopituitarism due to FOXA2 gene defects. Results We report a novel heterozygous nonsense c.616C > T(p.Q206X) variant that leads to a truncated protein that lacks part of the DNA-binding domain of FOXA2, resulting in impaired transcriptional activation of the glucose transporter type 2 (GLUT2)-luciferase reporter. The patient is the sixth patient described in the literature with a FOXA2 mutation, and the first patient exhibiting pancreatic hypoplasia. We also report a second patient with a novel de novo 8.53 Mb deletion of 20p11.2 that encompasses FOXA2, who developed diabetes mellitus that responded to sulfonylurea treatment. Conclusion Our 2 cases broaden the molecular and clinical spectrum of FOXA2-related disease, reporting the first nonsense mutation and the first case of pancreatic dysgenesis.
- Published
- 2021
- Full Text
- View/download PDF
12. O‐GlcNAcylation promotes the migratory ability of hepatocellular carcinoma cells via regulating FOXA2 stability and transcriptional activity
- Author
-
Lingyan Wang, Qiong Wu, Yangzhi Liu, Huang Huang, Xinyi Guo, Lin Shi, Xueqin Xie, Jianing Zhang, Yubo Liu, Tianmiao Huang, and Wenli Li
- Subjects
0301 basic medicine ,Carcinoma, Hepatocellular ,Glycosylation ,Transcription, Genetic ,Physiology ,Clinical Biochemistry ,Cell ,Regulator ,Biology ,N-Acetylglucosaminyltransferases ,03 medical and health sciences ,0302 clinical medicine ,Antigens, CD ,Cell Movement ,Transcription (biology) ,medicine ,Humans ,Neoplasm Invasiveness ,Transcription factor ,reproductive and urinary physiology ,Protein Stability ,Cadherin ,Liver Neoplasms ,Ubiquitination ,Cell migration ,Hep G2 Cells ,Cell Biology ,respiratory system ,Cadherins ,digestive system diseases ,Gene Expression Regulation, Neoplastic ,HEK293 Cells ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Proteolysis ,embryonic structures ,Hepatocyte Nuclear Factor 3-beta ,Cancer research ,FOXA2 ,Signal transduction ,Protein Processing, Post-Translational - Abstract
O-GlcNAcylation is a posttranslational modification that regulates numerous nuclear and cytoplasmic proteins and is emerging as a key regulator of various biological processes, such as transcription, signal transduction, and cell motility. Although increasing evidence has shown that elevated levels of global O-GlcNAcylation are linked to the metastasis in hepatocellular carcinoma (HCC) cells, the underlying mechanism is still ambiguous. In this study, we demonstrated that forkhead box protein A2 (FOXA2), an essential transcription factor for liver homeostasis and HCC developing, was O-GlcNAcylated by O-GlcNAc transferase (OGT) and regulates HCC cells migration and invasion. Opposite FOXA2 and OGT expression tendency were observed in HCC tissues, and lower FOXA2 levels predicted a poor prognosis in HCC patients. The reduction of FOXA2 in HCC cells was found to be inversely correlated with the cellular O-GlcNAcylation and cell migratory ability. Notably, we found that FOXA2 was modified by O-GlcNAcylation and that O-GlcNAcylation activated the ubiquitination degradation of FOXA2 in highly metastatic HCC cells. Although this modification did not affect FOXA2 nuclear localization capability, O-GlcNAcylation on FOXA2 was key for attenuating FOXA2-mediated transcription. O-GlcNAcylation decreased the transcription of FOXA2 downstream target gene E-cadherin and it ultimately promoted O-GlcNAcylation-mediated HCC cell migration and invasion. The results provide insights into the role of O-GlcNAcylation in regulating FOXA2 activity and suggest its important implications in HCC metastasis.
- Published
- 2021
- Full Text
- View/download PDF
13. The Biological Effects of Forkhead Box Protein A2 (FOXA2) on Cervical Cancer Cells by Regulating Phosphatase and Tensin Homolog (PTEN)
- Author
-
Xiaochen Bi and Hangfang Lou
- Subjects
biology ,Phosphatase ,Biomedical Engineering ,Medicine (miscellaneous) ,Bioengineering ,respiratory system ,embryonic structures ,Cervical carcinoma ,biology.protein ,Cancer research ,Forkhead Box ,Tensin ,PTEN ,FOXA2 ,reproductive and urinary physiology ,Biotechnology - Abstract
The pathogenesis of cervical cancer is complex and FOX family is abnormally expressed in several diseases. FOXA2’s role in cervical cancer remains unclear. FOXA2 level in cervical cancer and adjacent normal tissues was detected. Cervical cancer Hela cells were divided into control group, FOXA2 group and FOXA2 siRNA group followed by analysis of FOXA2 level by Real time PCR and western blot, cell survival by MTT assay, cell migration and invasion, and PTEN expression by western blot. The cells were divided into NC group, FOXA2 group and FOXA2+PTEN inhibitor group followed by analysis of cell behaviors by flow cytometry and PTEN expression by western blot. FOXA2 was significantly downregulated in cancer tissues compared with adjacent tissues (PPPPP
- Published
- 2021
- Full Text
- View/download PDF
14. Trim46 contributes to the midbrain development via Sonic Hedgehog signaling pathway in zebrafish embryos
- Author
-
Tae-Lin Huh, Myungchull Rhee, Jaehun Kim, and Jangham Jung
- Subjects
0301 basic medicine ,animal structures ,Cyclopamine ,Protein family ,mhb (midbrain-hindbrain boundary) ,midbrain ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Midbrain ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine ,Ring finger ,Axon ,cyclopamine ,lcsh:QH301-705.5 ,lcsh:R5-920 ,foxa2 ,Sonic hedgehog signaling pathway ,Ubiquitin ligase ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,sonic hedgehog (shh) ,nervous system ,chemistry ,trim46a ,lcsh:Biology (General) ,030220 oncology & carcinogenesis ,embryonic structures ,biology.protein ,Animal Science and Zoology ,FOXA2 ,lcsh:Medicine (General) ,Developmental Biology ,Research Article - Abstract
TRIM46 is a RING finger E3 ligase which belongs to TRIM (tripartite motif-containing) protein family. TRIM46 is required for neuronal polarity and axon specification by driving the formation of parallel microtubule arrays, whereas its embryological functions remain to be determined yet. Expression patterns and biological functions of trim46a, a zebrafish homologue of TRIM46, were studied in zebrafish embryo. First, maternal transcripts of trim46a were present at 1 cell stage whereas zygotic messages were abundant in the eyes, MHB (Midbrain-Hindbrain Boundary) and hindbrain at 24 hpf (hours post fertilization). Second, transcriptional regulatory region of trim46a contains cis-acting elements binding a transcriptional factor Foxa2. Transcription of foxa2 is positively regulated by Sonic Hedgehog (SHH), and treatment of cyclopamine, an SHH inhibitor, represses transcription of foxa2 in 4 hpf through 24 hpf embryos. Third, the transcriptional repression of foxa2 inhibited transcription of trim46a to cause developmental defects in the midbrain and MHB. Finally, spatiotemporal expression patterns of a midbrain marker otx2b in the developmental defects confirmed inhibition of SHH by cyclopamine caused underdevelopment of the midbrain and MHB at 24 hpf. We propose a signaling network where trim46a contributes to development of the midbrain and MHB via Foxa2, a downstream element of SHH signaling in zebrafish embryogenesis.
- Published
- 2021
15. The pioneer and differentiation factor FOXA2 is a key driver of yolk‐sac tumour formation and a new biomarker for paediatric and adult yolk‐sac tumours
- Author
-
Wruck, Wasco, Bremmer, Felix, Kotthoff, Mara, Fichtner, Alexander, Skowron, Margaretha A., Schönberger, Stefan, Calaminus, Gabriele, Vokuhl, Christian, Pfister, David, Heidenreich, Axel, Albers, Peter, Adjaye, James, and Nettersheim, Daniel
- Subjects
Pluripotency ,Oncogene Proteins ,adult and paediatric germ cell tumours ,DNA methylation ,embryonal carcinomas ,microRNA ,Age Factors ,Endodermal Sinus Tumor ,Original Articles ,Immunohistochemistry ,Models, Biological ,Yolk‐sac tumours ,Cell Line, Tumor ,Biomarkers, Tumor ,Hepatocyte Nuclear Factor 3-beta ,biomarker ,Humans ,Original Article ,FOXA2 ,Disease Susceptibility ,SOX17 - Abstract
Yolk‐sac tumours (YSTs), a germ cell tumour subtype, occur in newborns and infants as well as in young adults of age 14‐44 years. In clinics, adult patients with YSTs face a poor prognosis, as these tumours are often therapy‐resistant and count for many germ cell tumour related deaths. So far, the molecular and (epi)genetic mechanisms that control development of YST are far from being understood. We deciphered the molecular and (epi)genetic mechanisms regulating YST formation by meta‐analysing high‐throughput data of gene and microRNA expression, DNA methylation and mutational burden. We validated our findings by qRT‐PCR and immunohistochemical analyses of paediatric and adult YSTs. On a molecular level, paediatric and adult YSTs were nearly indistinguishable, but were considerably different from embryonal carcinomas, the stem cell precursor of YSTs. We identified FOXA2 as a putative key driver of YST formation, subsequently inducing AFP, GPC3, APOA1/APOB, ALB and GATA3/4/6 expression. In YSTs, WNT‐, BMP‐ and MAPK signalling‐related genes were up‐regulated, while pluripotency‐ and (primordial) germ cell‐associated genes were down‐regulated. Expression of FOXA2 and related key factors seems to be regulated by DNA methylation, histone methylation / acetylation and microRNAs. Additionally, our results highlight FOXA2 as a promising new biomarker for paediatric and adult YSTs.
- Published
- 2021
16. 20p11.23-p11.21 deletion in a child with hyperinsulinemic hypoglycemia and GH deficiency: A case report
- Author
-
Hirofumi Ohashi, Ko Ichihashi, Hiroaki Sato, Daisuke Sugawara, and Misa Matsuura
- Subjects
medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Recurrent hypoglycemia ,Case Report ,Hypopituitarism ,Hypoglycemia ,medicine.disease_cause ,Endocrinology ,Internal medicine ,Diazoxide ,medicine ,Hyperinsulinemic hypoglycemia ,Pathological ,GH deficiency ,Pituitary stalk ,business.industry ,Insulin ,medicine.disease ,20p deletion ,Pediatrics, Perinatology and Child Health ,hyperinsulinemic hypoglycemia ,FOXA2 ,business ,medicine.drug - Abstract
Some neonatal hypoglycemias have genetic origins. For instance, mutation in forkhead box protein A2 (FOXA2), located on chromosome 20p11.21, has recently been reported to cause hyperinsulinemic hypoglycemia and hypopituitarism. Here, we report a case of hyperinsulinemic hypoglycemia and GH deficiency (GHD) with 20p11.23-p11.21 deletion, which included FOXA2. The boy was diagnosed with hyperinsulinemic hypoglycemia during the neonatal period and subsequently administered diazoxide for treatment. His blood glucose levels gradually stabilized, and the diazoxide dosage was slowly reduced and ultimately fully weaned. The patient was discharged at the age of 29 d. Unfortunately, the patient experienced recurrent hypoglycemia at 3 mo, and diazoxide administration was re-initiated. Further examination, including chromosomal microarray analysis, revealed a 2.48-Mb 20p11.23-p11.21 deletion that encompassed FOXA2. In addition, severe GHD was detected, and magnetic resonance imaging of the brain revealed pituitary stalk interruption. Accordingly, GH replacement therapy was started at 0.175 mg/kg/wk, and blood glucose levels were stabilized. Our report suggests that there are pathological conditions that can cause both hyperinsulinemic hypoglycemia and hypopituitarism and reaffirms the importance of evaluating not only insulin and congenital metabolic disorders but also pituitary function in patients with hypoglycemia.
- Published
- 2021
- Full Text
- View/download PDF
17. Confined target search powers FOXA2 transcription activity
- Author
-
Wang, Zuhui, Niu, Di, Cattoglio, Claudia, Bi, Ying, Lavis, Luke D, Loh, Kyle M, and Deng, Wulan
- Subjects
pioneer factor ,single-molecule tracking ,target search ,hESCs ,chromatin binding ,FOXA2 ,intrinsically disordered region ,transcription factor - Abstract
SMT trajectory data for "Confined target search powers FOXA2 transcription activity" Zuhui Wang1,2†, Di Niu1,2,3†, Claudia Cattoglio4,5, Ying Bi1,2, Luke D Lavis6, Kyle M Loh7, Wulan Deng1,2,3, * 1Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing 100871, China. 2Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China. 3Peking-Tsinghua Center for Life Sciences (CLS), Peking University, Beijing 100871, China. 4Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA. 5Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA. 6Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia. 7Institute for Stem Cell Biology and Regenerative Medicine and Ludwig Center for Cancer Stem Cell Biology and Medicine, Departments of Pathology and Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA. † These authors contributed equally to this work. *Corresponding author: wdeng@pku.edu.cn Overview This repository contains all the SMT trajectory data associated with "Confined target search powers FOXA2 transcription activity". In the repository, there are two main folders. The folder 'slowSMT_trajectories_pooled' contains pooled slowSMT trajectories that we used to calculate residence times. The folder 'spaSMT_trajectories_single_cells' contains spaSMT single-cell trajectories, with DE/APS and U2OS trajectories in the separate folders. We used the spaSMT data in the 'DE/APS'folder for both Spot-On analysis and Anisotropy analysis, while for the spaSMT data in the 'U2OS'folder, we used indicated subfolders for Spot-On analysis and/or Anisotropy analysis. For each pooled trajectory file in the folder 'slowSMT_trajectories_pooled', it contains two matlab variables. trackedPar: each element represents one trajectory containing one or more localizations xy: x- and y-coordinates in micrometers of the molecule localizations Frame: frame indexes of the molecule localizations. TimeStamp: timepoints in seconds of the molecule localizations. sampleinfo: names of individual cell samples. For each single cell trajectory file in the folder 'spaSMT_trajectories_single_cells', it contains two matlab variables. trackedPar: same as above. settings: contains file information, image acquisition settings, the algorithm settings used in MTT localization and tracking software. For detailed explanation, please see the MTT code at https://gitlab.com/tjian-darzacq-lab/SPT_LocAndTrack. The document does not contain information about how the data was analyzed. For details on how the data was analyzed and for codes to reproduce our figure, please go to our github repository. For the slowSMT analysis, we deposit the code under: https://github.com/denglabpku/slowSMT. For the anisotropy analysis, we deposit the code under: https://github.com/denglabpku/anisotropy. For the Spot-On analysis, please visit the Spot-On website: https://spoton.berkeley.edu/. Our trajectory file format is directly available as the input of the Spot-On software.  
- Published
- 2022
- Full Text
- View/download PDF
18. FoxA2 and p53 regulate the transcription of HSD17B1 in ovarian granulosa cells of pigs
- Author
-
Qi Wu, Zhe Zhang, Yao Jiang, Jiaqi Li, Hao Zhang, Wang Xilong, Xiaolong Yuan, Xiwu Qiao, Xiaofeng Zhou, and Zhixiang Yao
- Subjects
endocrine system ,17-Hydroxysteroid Dehydrogenases ,Transcription, Genetic ,Sus scrofa ,Biology ,Gene Expression Regulation, Enzymologic ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Endocrinology ,Transcription (biology) ,Animals ,Luciferase ,Promoter Regions, Genetic ,Gene ,Granulosa Cells ,030219 obstetrics & reproductive medicine ,0402 animal and dairy science ,Promoter ,04 agricultural and veterinary sciences ,040201 dairy & animal science ,Cell biology ,chemistry ,Hepatocyte Nuclear Factor 3-beta ,Female ,Animal Science and Zoology ,HSD17B1 ,FOXA2 ,Hydroxysteroid ,Tumor Suppressor Protein p53 ,Chromatin immunoprecipitation ,Biotechnology - Abstract
The oestrogens have been highly implicated in the fertility of female animals. It is widely known that the oestrogens are primarily synthetized by the ovarian granulosa cells (GCs), and the final and essential step of this process is to catalyse the oestrone to the more active oestradiol by the protein coded by hydroxysteroid 17-beta dehydrogenase 1 (HSD17B1) gene. However, the molecular mechanism regarding the transcription of HSD17B1 remains to be fully elucidated in ovarian GCs. In this study, the 5'-deletion, luciferase assay and chromatin immunoprecipitation (ChIP) were utilized to explore the molecular regulation of transcription of HSD17B1 with the porcine ovarian GCs as the cellular model. After the deletions with -2105 to -1754 bp, -1753 to -1429 bp, -1430 to -1081 bp and -1082 to -730 bp, the relative luciferase activity of HSD17B1 promoter did not change significantly, but the deletion of -731 to -332 bp significantly increased the relative luciferase activity of HSD17B1 promoter, and an insertion (GTTT) that might raise the transcription of HSD17B1 was identified at -401 bp of HSD17B1. These findings suggested the region from -731 to +38 bp was the core promoter of HSD17B1, and the region between -731 to -332 bp might be a silence element for HSD17B1. Furthermore, the forkhead box A2 (FoxA2) directly bound at -412 to -401 bp to negatively but p53 bound at -383 to -374 bp to positively regulate the transcription and translation of HSD17B1 in ovarian GCs. These findings will improve our understanding on HSD17B1-mediated oestrogens and provide useful information for further investigations into fertility of females.
- Published
- 2020
- Full Text
- View/download PDF
19. Generation of metabolically functional hepatocyte‐like cells from dedifferentiated fat cells by Foxa2, Hnf4a and Sall1 transduction
- Author
-
Takashi Matsumaru, Shiho Ibayashi, Yoshinao Oki, Reiko Hagiwara, and Koichiro Kano
- Subjects
Cell ,Adipose tissue ,Biology ,DFAT ,Mice ,03 medical and health sciences ,Transduction (genetics) ,zonation ,Transduction, Genetic ,Gene expression ,hepatocyte ,Genetics ,medicine ,Animals ,Cellular Reprogramming Techniques ,Transcription factor ,Cells, Cultured ,030304 developmental biology ,0303 health sciences ,gene expression analysis ,Mesenchymal stem cell ,Original Articles ,Cell Biology ,direct reprogramming ,Cell biology ,medicine.anatomical_structure ,Adipose Tissue ,Hepatocyte Nuclear Factor 4 ,Hepatocyte ,Cell Transdifferentiation ,Hepatocyte Nuclear Factor 3-beta ,Hepatocytes ,Original Article ,FOXA2 ,Transcription Factors - Abstract
Mature adipocyte‐derived dedifferentiated fat (DFAT) cells have been identified to possess similar multipotency to mesenchymal stem cells, but a method for converting DFAT cells into hepatocytes was previously unknown. Here, using comprehensive analysis of gene expression profiles, we have extracted three transcription factors, namely Foxa2, Hnf4a and Sall1 (FHS), that can convert DFAT cells into hepatocytes. Hepatogenic induction has converted FHS‐infected DFAT cells into an epithelial‐like morphological state and promoted the expression of hepatocyte‐specific features. Furthermore, the DFAT‐derived hepatocyte‐like (D‐Hep) cells catalyzed the detoxification of several compounds. These results indicate that the transduction of DFAT cells with three genes, which were extracted by comprehensive gene expression analysis, efficiently generated D‐Hep cells with detoxification abilities similar to those of primary hepatocytes. Thus, D‐Hep cells may be useful as a new cell source for surrogate hepatocytes and may be applied to drug discovery studies, such as hepatotoxicity screening and drug metabolism tests., In this study, we used a comprehensive analysis of the global expression profiles of hepatocytes, mature adipocyte‐derived dedifferentiated fat (DFAT) cells, hepatic stem cells and adipocytes to identify three transcription factors—Foxa2, Hnf4a and Sall1—that can specifically induce hepatocyte differentiation in DFAT cells.
- Published
- 2020
- Full Text
- View/download PDF
20. Histone demethylase PHF8 drives neuroendocrine prostate cancer progression by epigenetically upregulating FOXA2
- Author
-
Luofu Wang, Qiubo Xie, Gaolei Liu, Jian Pang, Jun Qin, Zhuowei Huang, Lin-Ang Wang, Dianzheng Zhang, Jing Xu, Hualiang Xiao, Dali Tong, Weihua Lan, Tang Tang, Jun Jiang, Xingxia Yang, Qiuli Liu, Qiang Ma, and Yiqiang Huang
- Subjects
0301 basic medicine ,Male ,Transcription, Genetic ,PHF8 ,Epigenesis, Genetic ,Prostate cancer ,0302 clinical medicine ,Cell Movement ,Histone Demethylases ,Mice, Knockout ,Tissue microarray ,biology ,TRAMP mice ,prostate cancer ,Original Papers ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,Histone ,030220 oncology & carcinogenesis ,PC-3 Cells ,Hepatocyte Nuclear Factor 3-beta ,Adenocarcinoma ,epigenetic ,Tramp ,Mice, Nude ,histone ,demethylase ,Pathology and Forensic Medicine ,03 medical and health sciences ,medicine ,Biomarkers, Tumor ,neuroendocrine ,Animals ,Humans ,Epigenetics ,transcriptional factor ,Cell Proliferation ,Original Paper ,tissue microarray ,business.industry ,Prostatic Neoplasms ,medicine.disease ,Carcinoma, Neuroendocrine ,Mice, Inbred C57BL ,030104 developmental biology ,biology.protein ,Cancer research ,FOXA2 ,business ,Transcription Factors - Abstract
Neuroendocrine prostate cancer (NEPC) is a more aggressive subtype of castration‐resistant prostate cancer (CRPC). Although it is well established that PHF8 can enhance prostate cancer cell proliferation, whether PHF8 is involved in prostate cancer initiation and progression is relatively unclear. By comparing the transgenic adenocarcinoma of the mouse prostate (TRAMP) mice with or without Phf8 knockout, we systemically examined the role of PHF8 in prostate cancer development. We found that PHF8 plays a minimum role in initiation and progression of adenocarcinoma. However, PHF8 is essential for NEPC because not only is PHF8 highly expressed in NEPC but also animals without Phf8 failed to develop NEPC. Mechanistically, PHF8 transcriptionally upregulates FOXA2 by demethylating and removing the repressive histone markers on the promoter region of the FOXA2 gene, and the upregulated FOXA2 subsequently regulates the expression of genes involved in NEPC development. Since both PHF8 and FOXA2 are highly expressed in NEPC tissues from patients or patient‐derived xenografts, the levels of PHF8 and FOXA2 can either individually or in combination serve as NEPC biomarkers and targeting either PHF8 or FOXA2 could be potential therapeutic strategies for NEPC treatment. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
- Published
- 2020
21. X-box-binding protein 1 is required for pancreatic development in
- Author
-
Fang Yuan, Xuejun Wang, Deli Li, Jia Liu, Liyuan Wei, Xingjing Liu, Jing Yang, and Li Yuan
- Subjects
0303 health sciences ,Biophysics ,Activating transcription factor ,Xenopus ,General Medicine ,Biology ,CREB ,biology.organism_classification ,Biochemistry ,X-Box Binding Protein 1 ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,medicine ,biology.protein ,PDX1 ,FOXA2 ,Pancreas ,Transcription factor ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
X-box-binding protein 1 (XBP1) is a protein containing the basic leucine zipper structure. It belongs to the cAMP-response element binding protein (CREB)/activating transcription factor transcription factor family. As the main transcription factor, spliced XBP1 (XBP1s) participates in many physiological and pathological processes and plays an important role in embryonic development. Previous studies showed that XBP1-knockout mice died because of pancreatic exocrine function deficiency, indicating that XBP1 plays an important role in pancreatic development. However, the exact role of XBP1 in pancreatic development remains unclear. This study aimed to investigate the role of XBP1 in the pancreatic development of Xenopus laevis embryos. Whole-mount in situ hybridization and quantitative real-time PCR results revealed that the expression levels of pancreatic progenitor marker genes pdx1, p48, ngn3, and sox9 were downregulated in XBP1s morpholino oligonucleotide (MO)-injected embryos. The expression levels of pancreatic exocrine and endocrine marker genes insulin and amylase were also downregulated. Through the overexpression of XBP1s, the phenotype and gene expressions were opposite to those in XBP1s MO-injected embryos. Luciferase and chromatin immunoprecipitation assays showed that XBP1s could bind to the XBP1-binding site in the foxa2 promoter. These results revealed that XBP1 is required in the pancreatic development of Xenopus laevis and might function by regulating foxa2.
- Published
- 2020
- Full Text
- View/download PDF
22. Effects of Dimethyl Sulfoxide on the Pluripotency and Differentiation Capacity of Mouse Embryonic Stem Cells
- Author
-
Sung Hyun Kim, Zae-Young Ryoo, Song Park, Mee-Hyun Lee, Hai Huang, Jae Jung Ha, Junkoo Yi, Myoungok Kim, Si-Jun Park, and Dae-Hyun Kim
- Subjects
Pluripotent Stem Cells ,0301 basic medicine ,Cell Survival ,medicine.medical_treatment ,Biology ,Leukemia Inhibitory Factor ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,medicine ,Animals ,Dimethyl Sulfoxide ,Cells, Cultured ,030102 biochemistry & molecular biology ,Oncogene ,Dimethyl sulfoxide ,Cell Cycle ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Mouse Embryonic Stem Cells ,Cell Biology ,Methylation ,DNA Methylation ,Embryonic stem cell ,Cell biology ,030104 developmental biology ,Cytokine ,chemistry ,embryonic structures ,FOXA2 ,Stem cell ,Leukemia inhibitory factor ,Biomarkers ,Developmental Biology ,Biotechnology - Abstract
Mouse embryonic stem cells (mESCs) go through self-renewal in the existence of the cytokine leukemia inhibitory factor (LIF). LIF is added to the mouse stem cells culture medium, and its removal results in fast differentiation. Dimethyl sulfoxide (DMSO) is one of the most used solvents in drug test. We exposed 4-day mESC cultures to different concentrations of DMSO (0.1%, 0.5%, 1.0%, and 2.0%) to identify the safest dose exhibiting efficacy as a solvent. mESCs grown under general pluripotency conditions in the absence of LIF were treated with DMSO. In addition, as a control for differentiation, mESCs were grown in the absence of LIF. DMSO upregulated the mRNA expression level of pluripotency markers. Moreover, DMSO reduced the mRNA expression levels of ectodermal marker (β-tubulin3), mesodermal marker (Hand1), and endodermal markers (Foxa2 and Sox17) in mESCs. These results indicate that DMSO treatment enhances the pluripotency and disrupts the differentiation of mESCs. We also show that members of the Tet oncogene family are critical to inhibiting the differentiation and methylation of mESCs. DMSO is appropriate to sustain the pluripotency of mESCs in the absence of LIF, and that mESCs can be sustained in an undifferentiated state using DMSO. Therefore, DMSO may, in part, function as a substitute for LIF.
- Published
- 2020
- Full Text
- View/download PDF
23. Identification of determinants of differential chromatin accessibility through a massively parallel genome-integrated reporter assay
- Author
-
Jennifer Hammelman, Richard I. Sherwood, Konstantin Krismer, David K. Gifford, and Budhaditya Banerjee
- Subjects
Brachyury ,Oligonucleotides ,Method ,Computational biology ,Regulatory Sequences, Nucleic Acid ,Biology ,Genome ,DNA sequencing ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Genetics ,medicine ,Animals ,Nucleotide Motifs ,Transcription factor ,Embryonic Stem Cells ,Genetics (clinical) ,030304 developmental biology ,Base Composition ,0303 health sciences ,Endoderm ,Pioneer factor ,DNA ,Genomics ,Sequence Analysis, DNA ,Chromatin ,medicine.anatomical_structure ,chemistry ,FOXA2 ,Sequence motif ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
A key mechanism in cellular regulation is the ability of the transcriptional machinery to physically access DNA. Pioneer transcription factors interact with DNA to open chromatin, which subsequently enables changes to gene expression during development, disease, or as a response to environmental stimuli. However, the regulation of DNA accessibility via the recruitment of transcription factors is difficult to understand in the context of the native genome because every genomic site is distinct in multiple ways. Here we introduce the Multiplexed Integrated Accessibility Assay (MIAA), a multiplexed parallel reporter assay which measures changes to genome accessibility as a result of the integration of synthetic oligonucleotide phrase libraries into a controlled, natively inaccessible genomic context. We apply MIAA to measure the effects of sequence motifs on cell type-specific DNA accessibility between mouse embryonic stem cells and embryonic stem cell-derived definitive endoderm cells, screening a total of 7,905 distinct phrases. MIAA is able to recapitulate differential accessibility patterns of 100-nt sequences derived from natively differential genomic regions, identifying the presence of E-box motifs common to epithelial-mesenchymal transition driver transcription factors in stem cell-specific accessible regions that become repressed during differentiation to endoderm. We further present causal evidence that a single binding motif for a key regulatory transcription factor is sufficient to open chromatin, and classify sets of stem cell-specific, endoderm-specific, and shared pioneer factor motifs. We also demonstrate that over-expression of two definitive endoderm transcription factors, Brachyury and FoxA2, results in changes to accessibility in phrases containing their respective DNA-binding motifs. Finally, we use MIAA results to explore the order of motif interactions and identify preferential motif ordering arrangements that appear to have an effect on accessibility.
- Published
- 2020
- Full Text
- View/download PDF
24. Comprehensive genetic analysis of pediatric germ cell tumors identifies potential drug targets
- Author
-
Satoru Miyano, Keisuke Kataoka, Seishi Ogawa, Yukichi Tanaka, Yuichi Shiraishi, Kenichi Yoshida, Hiromichi Suzuki, Junko Takita, Hiroko Tanaka, Masahiro Sekiguchi, Mitsuteru Hiwatari, Tomoya Isobe, Shunsuke Kimura, Aiko Sato-Otsubo, Yasuo Kubota, Kenichi Chiba, Yoichi Fujii, Kenichiro Hata, Masafumi Seki, Yasuhide Hayashi, Kentaro Watanabe, Tomoko Kawai, Misa Yoshida, and Akira Oka
- Subjects
Male ,0301 basic medicine ,DNA Copy Number Variations ,Medicine (miscellaneous) ,Biology ,Polymorphism, Single Nucleotide ,Genetic analysis ,Article ,General Biochemistry, Genetics and Molecular Biology ,Paediatric cancer ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Carcinoma, Embryonal ,Exome Sequencing ,Cancer genomics ,medicine ,Humans ,Child ,Gene ,lcsh:QH301-705.5 ,Oligonucleotide Array Sequence Analysis ,GATA6 ,Germinoma ,Endodermal Sinus Tumor ,Teratoma ,Infant ,Germ cell tumours ,DNA Methylation ,Neoplasms, Germ Cell and Embryonal ,medicine.disease ,030104 developmental biology ,lcsh:Biology (General) ,Child, Preschool ,030220 oncology & carcinogenesis ,embryonic structures ,DNA methylation ,Cancer research ,Female ,FOXA2 ,Germ cell tumors ,General Agricultural and Biological Sciences - Abstract
To elucidate the molecular pathogenesis of pediatric germ cell tumors (GCTs), we performed DNA methylation array analysis, whole transcriptome sequencing, targeted capture sequencing, and single-nucleotide polymorphism array analysis using 51 GCT samples (25 female, 26 male), including 6 germinomas, 2 embryonal carcinomas, 4 immature teratomas, 3 mature teratomas, 30 yolk sac tumors, and 6 mixed germ cell tumors. Among the 51 samples, 11 were from infants, 23 were from young children, and 17 were from those aged ≥10 years. Sixteen of the 51 samples developed in the extragonadal regions. Germinomas showed upregulation of pluripotent genes and global hypomethylation. Pluripotent genes were also highly expressed in embryonal carcinomas. These genes may play essential roles in embryonal carcinomas given that their binding sites are hypomethylated. Yolk sac tumors exhibited overexpression of endodermal genes, such as GATA6 and FOXA2, the binding sites of which were hypomethylated. Interestingly, infant yolk sac tumors had different DNA methylation patterns from those observed in older children. Teratomas had higher expression of ectodermal genes, suggesting a tridermal nature. Based on our results, we suggest that KIT, TNFRSF8, and ERBB4 may be suitable targets for the treatment of germinoma, embryonal carcinomas, and yolk sac tumors, respectively., Yasuo Kubota et al. report a multi-omic analysis of pediatric germ cell tumors from 51 patients ranging in age from 2 months to 19 years. They identify unique methylation, expression, and mutational patterns for each of the main subtypes and propose potential target genes for treatments against the three main subtypes.
- Published
- 2020
- Full Text
- View/download PDF
25. Exendin-4 restores airway mucus homeostasis through the GLP1R-PKA-PPARγ-FOXA2-phosphatase signaling
- Author
-
Gee W. Lau, Woo-Suk Choi, Beata Kosmider, Robert Vassallo, Michael T. Borchers, Jingjun Lin, Shawn Choe, and Andrew H. Limper
- Subjects
0301 basic medicine ,Chronic bronchitis ,Cystic Fibrosis ,Gene Expression ,medicine.disease_cause ,Cystic fibrosis ,Pulmonary Disease, Chronic Obstructive ,chemistry.chemical_compound ,fluids and secretions ,0302 clinical medicine ,Homeostasis ,Immunology and Allergy ,STAT6 ,respiratory system ,ErbB Receptors ,Pseudomonas aeruginosa ,Hepatocyte Nuclear Factor 3-beta ,Disease Susceptibility ,Protein Binding ,Signal Transduction ,EGFR ,Immunology ,Respiratory Mucosa ,Models, Biological ,Article ,Glucagon-Like Peptide-1 Receptor ,Proinflammatory cytokine ,Incretin mimetics ,03 medical and health sciences ,Pyocyanin ,medicine ,COPD ,Humans ,business.industry ,Mucin ,Mucins ,medicine.disease ,Cyclic AMP-Dependent Protein Kinases ,Mucus ,PPAR gamma ,030104 developmental biology ,chemistry ,Cancer research ,Exenatide ,FOXA2 ,STAT6 Transcription Factor ,business ,Proto-Oncogene Proteins c-akt ,030215 immunology - Abstract
Goblet cell hyperplasia and metaplasia and excessive mucus are prominent pathologies of chronic airway diseases such as chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF), and chronic bronchitis. Chronic infection by respiratory pathogens, including Pseudomonas aeruginosa, exacerbates cyclical proinflammatory responses and mucus hypersecretion. P. aeruginosa and its virulence factor pyocyanin contribute to these pathologies by inhibiting FOXA2, a key transcriptional regulator of mucus homeostasis, through activation of antagonistic signaling pathways EGFR-AKT/ERK1/2 and IL-4/IL-13-STAT6-SPDEF. However, FOXA2-targeted therapy has not been previously explored. Here, we examined the feasibility of repurposing the incretin mimetic Exendin-4 to restore FOXA2-mediated airway mucus homeostasis. We have found that Exendin-4 restored FOXA2 expression, attenuated mucin production in COPD and CF-diseased airway cells, and reduced mucin and P. aeruginosa burden in mouse lungs. Mechanistically, Exendin-4 activated the GLP1R-PKA-PPAR-γ-dependent phosphatases PTEN and PTP1B phosphatases, which inhibited key kinases within both EGFR and STAT6 signaling cascades. Our results may lead to the repurposing of Exendin-4 and other incretin mimetics to restore FOXA2 function and ultimately regulate excessive mucus in diseased airways.
- Published
- 2020
- Full Text
- View/download PDF
26. Inhibition of LSD1 promotes the differentiation of human induced pluripotent stem cells into insulin-producing cells
- Author
-
Xiaofei Yang, Fei He, Ning Li, Fu-Rong Li, Wei Huang, Ce Wang, and Shu-Yan Zhou
- Subjects
animal structures ,Induced Pluripotent Stem Cells ,Medicine (miscellaneous) ,LSD1 ,Mice, SCID ,Biology ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Diabetes Mellitus, Experimental ,Small hairpin RNA ,lcsh:Biochemistry ,Mice ,SOX2 ,Insulin-Secreting Cells ,Histone methylation ,Animals ,Humans ,Insulin ,lcsh:QD415-436 ,Histone Demethylases ,Gene knockdown ,lcsh:R5-920 ,Research ,Diabetes ,Cell Differentiation ,Human induced pluripotent stem cells ,Cell Biology ,Cell cycle ,Cell biology ,Apoptosis ,Differentiation ,embryonic structures ,Molecular Medicine ,Insulin-producing cells ,FOXA2 ,Stem cell ,lcsh:Medicine (General) - Abstract
Background Human induced pluripotent stem cells (hiPSCs) represent a potentially unlimited source of pancreatic endocrine lineage cells. Although insulin-producing β cells derived from hiPSCs have been successfully induced, much work remains to be done to achieve mature β cells. Lysine-specific demethylase 1 (LSD1) plays an important role in the regulation of hiPSC self-renewal and differentiation. We propose a new strategy to acquire insulin-producing cells (IPCs) from hiPSCs by knocking down LSD1. Methods Knockdown of LSD1 in hiPSCs with five shRNA. Assessment of the effects of shRNA on hiPSC proliferation, cell cycle, and apoptosis. Using knockdown hiPSCs with 31.33% LSD1 activity, we achieved a four-step differentiation into IPCs and test its differentiation efficiency, morphology, and marker genes and proteins. We implanted the IPCs into the renal subcapsular of SCID-Beige diabetic mice to evaluate the hypoglycemic effect in vivo. We tested LSD1 and HDAC1 whether they are present in the CoREST complex through IP-WB, and analyzed LSD1, CoREST, HDAC1, H3K4me2/me3, and H3K27me3 protein expression before and after knockdown of LSD1. Results Differentiated hiPSCs were 38.32% ± 3.54% insulin-positive cells and released insulin/C-peptide in response to glucose stimulus in a manner comparable to adult human islets. Most of the IPCs co-expressed mature β cell-specific markers. When transplanted under the left renal capsule of SCID-Beige diabetic mice, these IPCs reversed hyperglycemia, leading to a significant increase in the definitive endoderm cells. IP-WB results showed that LSD1, HDAC1, and CoREST formed a complex in hiPSCs. Chip-PCR results showed that LSD1, HDAC1, and CoREST were enriched in the same district during the SOX17 and FOXA2 promoter region. Inhibition of LSD1 would not affect the level of CoREST but decreased the HDAC1 expressions. The H3K4me2/me3 and H3K9act level of SOX17 and FOXA2 promoter region increased after inhibited of LSD1, and promoted transcriptional activation. The H3K4me2/me3 and H3K9act level of OCT4 and SOX2 promoter region decreased with the transcriptional repressed. Conclusions LSD1 regulated histone methylation and acetylation in promoter regions of pluripotent or endodermal genes. Our results suggest a highly efficient approach to producing IPCs from hiPSCs.
- Published
- 2020
- Full Text
- View/download PDF
27. LncRNA FTX activates FOXA2 expression to inhibit non–small‐cell lung cancer proliferation and metastasis
- Author
-
Jing He, Wen Gao, Shidai Jin, Deqin Wu, Jun Li, and Yue Zhou
- Subjects
Male ,0301 basic medicine ,non–small‐cell lung cancer ,FTX ,Biology ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,Carcinoma, Non-Small-Cell Lung ,medicine ,Humans ,Neoplasm Metastasis ,Lung cancer ,Transcription factor ,Cell Proliferation ,miR‐200a‐3p ,Competing endogenous RNA ,RNA ,Original Articles ,Cell Biology ,respiratory system ,medicine.disease ,respiratory tract diseases ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,030104 developmental biology ,A549 Cells ,030220 oncology & carcinogenesis ,Hepatocyte Nuclear Factor 3-beta ,Cancer research ,Molecular Medicine ,Original Article ,Female ,RNA, Long Noncoding ,XIST ,Ectopic expression ,FOXA2 - Abstract
Lung cancer leads to the highest mortality among all cancer types in the world, and non–small‐cell lung cancer (NSCLC) occupies over 80% of the lung cancer cases. Numerous studies have demonstrated that long non‐coding RNA (lncRNA) is involved in various human diseases including cancer. LncRNA FTX was firstly identified in Xist gene locus and was dysregulated in many human cancers. However, the function of FTX in NSCLC is still unclear. Here, we report that long non‐coding RNA FTX expression level is down‐regulated in NSCLC clinical tissue samples and cell lines. Ectopic expression of FTX inhibits proliferation and metastasis of lung cancer cells in vitro and in vivo. Furthermore, we find that FTX overexpression activates the expression of transcription factor FOXA2, an important regulator in lung cancer progression, and we reveal a novel FTX/miR‐200a‐3p/FOXA2 competing endogenous RNA regulatory axis in lung cancer cells. Our results provide new insights and directions for exploring the function of FTX in lung cancer progression.
- Published
- 2020
- Full Text
- View/download PDF
28. The key regulation of miR-124–3p during reprogramming of primary mouse hepatocytes into insulin-producing cells
- Author
-
Gui Pan, Quanwen Liu, Hongbo Xin, and Jianping Liu
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Cell ,Biophysics ,Biochemistry ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Insulin-Secreting Cells ,Insulin Secretion ,microRNA ,medicine ,Animals ,Molecular Biology ,Cells, Cultured ,NeuroD ,Chemistry ,Insulin ,Cell Differentiation ,Cell Biology ,Transfection ,Cellular Reprogramming ,Cell biology ,MicroRNAs ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,Hepatocyte Nuclear Factor 3-beta ,Hepatocytes ,PDX1 ,FOXA2 ,Biomarkers - Abstract
Based on the action of small molecule compounds, the efficiency of differentiation of mouse primary hepatocytes into insulin-producing cells (IPCs) was improved by changing the expression of miR-124–2p. Hepatocytes were transfected with microRNA-124–3p (miR-124–3p) mimic or inhibitor, followed by a chemical-defined culture system for maturation of IPCs. Then, detect the expression of insulin-related genes and protein and insulin secretion of each stage during differentiation. The expression of Foxa2, PDX1, NeuroD, insulin1, and insulin2 in IPCs in the miR-124–3p inhibition expression group was significantly upregulated, while the results were opposite in the miR-124–3p overexpression group. The results of cell immunofluorescence and glucose stimulation in vitro of the miR-124–3p inhibition expression group showed that the expression of insulin, PDX1, and C-peptide was increased, and the differentiation efficiency was higher than those of the control group and overexpression group. The primary mouse hepatocytes were successfully reprogrammed into IPCs by small-molecule compounds. We found that miR-124–3p plays a negative regulatory role in the differentiation of hepatocytes into IPCs in vitro. Inhibition of miR-124–3p expression significantly increased the expression of FOXA2 and PDX1, promoted the differentiation of hepatocytes into IPCs, and increased the induction efficiency.
- Published
- 2020
- Full Text
- View/download PDF
29. MicroRNA-1246 Promotes Melanoma Progression Through Targeting FOXA2
- Author
-
Yu Y, Yu F, and Sun P
- Subjects
cell metastasis ,melanoma ,foxa2 ,mir-1246 ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,cell viability - Abstract
Yanhua Yu,1 Fang Yu,1 Pijiang Sun2 1Department of Dermatology, Weihai Central Hospital Affiliated to Qingdao University, Weihai 264400, People’s Republic of China; 2Department of Hepatobiliary and Abdominal Hernias Surgery, Weihai Central Hospital Affiliated to Qingdao University, Weihai 264400, People’s Republic of ChinaCorrespondence: Pijiang SunDepartment of Hepatobiliary and Abdominal Hernias Surgery, Weihai Central Hospital Affiliated to Qingdao University, No. 3 Mishandong Road, Wendeng District, Weihai 264400, Shandong, People’s Republic of ChinaTel +86 152 6441 0981Email beciham@163.comIntroduction: Recently, the incidence of melanoma has been rising and there is a lack of effective targeted therapies. The regulatory mechanisms of microRNA-1246 (miR-1246) have been found in many cancers, except melanoma. This study focused on the regulatory mechanism of miR-1246 in melanoma development.Methods: The expression of miR-1246 was assessed using quantitative real-time polymerase chain reaction (RT-qPCR). Cell viability and metastasis were detected by Transwell and MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) assays. The protein expression of epithelial mesenchymal transition (EMT) makers was assessed by Western blot analysis. The target gene of miR-1246 was detected using luciferase reporter assay.Results: MiR-1246 expression was increased in melanoma tissues and cells. In addition, upregulation of miR-1246 promoted cell viability and metastasis in melanoma. Forkhead box protein A2 (FOXA2) was confirmed to be a direct target of miR-1246. And FOXA2 expression was decreased in melanoma and was suppressed by miR-1246. Importantly, upregulation of FOXA2 restored the carcinogenesis of miR-1246 in melanoma.Conclusion: MiR-1246 promoted cell viability and metastasis in melanoma by inhibiting FOXA2 expression.Keywords: miR-1246, melanoma, cell viability, cell metastasis, FOXA2
- Published
- 2020
30. Hepatic TET3 contributes to type-2 diabetes by inducing the HNF4α fetal isoform
- Author
-
Xiaoyong Yang, Gordon G. Carmichael, Sabrina Diano, Sungho Jin, Di Xie, Xinmei Huang, Hugh S. Taylor, Xiaoli Sun, Tiefeng Cao, Yingqun Huang, and Da Li
- Subjects
0301 basic medicine ,Male ,Metabolic disorders ,General Physics and Astronomy ,Mice ,0302 clinical medicine ,Glucose homeostasis ,Protein Isoforms ,Promoter Regions, Genetic ,lcsh:Science ,Regulation of gene expression ,Mice, Knockout ,Gene knockdown ,Multidisciplinary ,DNA methylation ,Fasting ,3. Good health ,Up-Regulation ,DNA Demethylation ,DNA-Binding Proteins ,Hepatocyte Nuclear Factor 4 ,Liver ,030220 oncology & carcinogenesis ,Hepatocyte Nuclear Factor 3-beta ,Gene isoform ,Transcriptional Activation ,medicine.medical_specialty ,Science ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Dioxygenases ,03 medical and health sciences ,Downregulation and upregulation ,Internal medicine ,medicine ,Animals ,Promoter ,General Chemistry ,Glucagon ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,DNA demethylation ,Glucose ,Diabetes Mellitus, Type 2 ,Gene Expression Regulation ,lcsh:Q ,FOXA2 ,Transcriptome - Abstract
Precise control of hepatic glucose production (HGP) is pivotal to maintain systemic glucose homeostasis. HNF4α functions to stimulate transcription of key gluconeogenic genes. HNF4α harbors two promoters (P2 and P1) thought to be primarily active in fetal and adult livers, respectively. Here we report that the fetal version of HNF4α is required for HGP in the adult liver. This isoform is acutely induced upon fasting and chronically increased in type-2 diabetes (T2D). P2 isoform induction occurs in response to glucagon-stimulated upregulation of TET3, not previously shown to be involved in HGP. TET3 is recruited to the P2 promoter by FOXA2, leading to promoter demethylation and increased transcription. While TET3 overexpression augments HGP, knockdown of either TET3 or the P2 isoform alone in the liver improves glucose homeostasis in dietary and genetic mouse models of T2D. These studies unmask an unanticipated, conserved regulatory mechanism in HGP and offer potential therapeutic targets for T2D., The HNF4α gene contains two promoters, which are thought to be active in the fetal and adult liver, the latter contributing to hepatic glucose production. Here the authors show that the fetal isoform of HNF4a is induced in mouse livers upon fasting and in type-2 diabetes in a manner regulated by TET3.
- Published
- 2020
31. Overexpression of microRNA-375 and microRNA-122 promotes the differentiation of human induced pluripotent stem cells into hepatocyte-like cells
- Author
-
Bita Geramizadeh, Parichehreh Yaghmaei, Mohammad Hossein Karimi, Zahra Jaafarpour, Naser Mobarra, Saman Hosseinkhani, and Masoud Soleimani
- Subjects
0301 basic medicine ,Induced Pluripotent Stem Cells ,Cell ,Gene Expression ,Bioengineering ,Biology ,Immunofluorescence ,Applied Microbiology and Biotechnology ,Cell Line ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Transduction, Genetic ,microRNA ,medicine ,Animals ,Humans ,Secretion ,030212 general & internal medicine ,Induced pluripotent stem cell ,Pharmacology ,Regulation of gene expression ,General Immunology and Microbiology ,medicine.diagnostic_test ,Lentivirus ,Cell Differentiation ,General Medicine ,Cell biology ,MicroRNAs ,030104 developmental biology ,medicine.anatomical_structure ,Hepatocyte ,Hepatocytes ,FOXA2 ,Biotechnology - Abstract
MicroRNAs (miRNAs) are involved in the regulation of gene expression. In this study, we evaluated the use of overexpression of microRNA-375 (miR-375) and miR-122 in differentiating the Human Induced Pluripotent Stem Cells (hiPSCs) into functional hepatocyte-like cells (HLCs) without growth factors. We also compared the differentiation by miRNAs versus growth factors. HiPSCs were divided into two main groups: 1- HiPSCs were induced using lentiviral overexpression of miR-375 to differentiate into definitive endoderm (DE) cells in seven days. Then lentiviral overexpression of miR-122 was applied to differentiate DE cells into HLCs in additional 14 days. 2- HiPSCs were differentiated into HLCs using growth factors in 21 days. DE and hepatocyte markers were investigated by qRT-PCR, immunofluorescence, secretion analysis and LDL uptake assay. In the produced cells of both groups: the expression levels of DE markers (FOXA2 and SOX17) and hepatocyte markers (albumin, CK18, and HNF4a) in comparison with the undifferentiated hiPSCs increased significantly in seven and 21 days respectively. The albumin and urea secretion and LDL uptake were also detected. These results weren't significantly different between two groups. Therefore, we demonstrated that the over expression of miR-375 and then miR-122 could differentiate hiPSCs into functional HLCs without growth factors for developing cell-based therapies.
- Published
- 2020
- Full Text
- View/download PDF
32. Exome-Sequencing Identifies Novel Genes Associated with Recurrent Pregnancy Loss in a Chinese Cohort
- Author
-
Huifen Xiang, Chunyan Wang, Hong Pan, Qian Hu, Ruyi Wang, Zuying Xu, Tengyan Li, Yezhou Su, Xu Ma, Yunxia Cao, and Binbin Wang
- Subjects
Nonsynonymous substitution ,Genetics ,Gene knockdown ,Pregnancy ,Candidate gene ,recurrent pregnancy loss ,QH426-470 ,Biology ,medicine.disease ,Miscarriage ,KHDC3L ,medicine ,Molecular Medicine ,whole-exome sequencing ,FOXA2 ,coagulation ,Gene ,Genetics (clinical) ,Exome sequencing ,Original Research - Abstract
Recurrent pregnancy loss (RPL) is a common reproductive problem affecting around 5% of couples worldwide. At present, about half of RPL cases remained unexplained. Previous studies have suggested an important role for genetic determinants in the etiology of RPL. Here, we performed whole-exome sequencing (WES) analysis on 100 unrelated Han Chinese women with a history of two or more spontaneous abortions. We identified 6736 rare deleterious nonsynonymous variants across all patients. To focus on possible candidate genes, we generated a list of 95 highly relevant genes that were functionally associated with miscarriage according to human and mouse model studies, and found 35 heterozygous variants of 28 RPL-associated genes in 32 patients. Four genes (FOXA2, FGA, F13A1, and KHDC3L) were identified as being strong candidates. The FOXA2 nonsense variant was for the first time reported here in women with RPL. FOXA2 knockdown in HEK-293T cells significantly diminished the mRNA and protein expression levels of LIF, a pivotal factor for maternal receptivity and blastocyst implantation. The other genes, with 29 variants, were involved in angiogenesis, the immune response and inflammation, cell growth and proliferation, which are functionally important processes for implantation and pregnancy. Our study identified several potential causal genetic variants in women with RPL by WES, highlighting the important role of genes controlling coagulation, confirming the pathogenic role of KHDC3L and identifying FOXA2 as a newly identified causal gene in women with RPL.
- Published
- 2021
- Full Text
- View/download PDF
33. Pioneer factor Foxa2 enables ligand-dependent activation of type II nuclear receptors FXR and LXRα
- Author
-
Jessica Kain, Claire Woods, Andrew J. Price, Irina M. Bochkis, Nihal A Reddy, and Xiaolong Wei
- Subjects
Male ,Receptors, Cytoplasmic and Nuclear ,Peroxisome proliferator-activated receptor ,Mice, Transgenic ,Foxa2 ,Ligands ,Mice ,Animals ,Pioneer factor ,Receptor ,Liver X receptor ,Molecular Biology ,Transcription factor ,Internal medicine ,reproductive and urinary physiology ,Liver X Receptors ,chemistry.chemical_classification ,Chemistry ,Cell Biology ,RC31-1245 ,Cell biology ,Chromatin ,Lipid metabolism ,Nuclear receptor ,Liver ,FXR ,Hepatocyte Nuclear Factor 3-beta ,Original Article ,LXR ,FOXA2 - Abstract
Objective Type II nuclear hormone receptors, including farnesoid X receptors (FXR), liver X receptors (LXR), and peroxisome proliferator-activated receptors (PPAR), which serve as drug targets for metabolic diseases, are permanently positioned in the nucleus and thought to be bound to DNA regardless of the ligand status. However, recent genome-wide location analysis showed that LXRα and PPARα binding in the liver is largely ligand-dependent. We hypothesized that pioneer factor Foxa2 evicts nucleosomes to enable ligand-dependent binding of type II nuclear receptors and performed genome-wide studies to test this hypothesis. Methods ATAC-Seq was used to profile chromatin accessibility; ChIP-Seq was performed to assess transcription factors (Foxa2, FXR, LXRα, and PPARα) binding; and RNA-Seq analysis determined differentially expressed genes in wildtype and Foxa2 mutants treated with a ligand (GW4064 for FXR, GW3965, and T09 for LXRα). Results We reveal that chromatin accessibility, FXR binding, LXRα occupancy, and ligand-responsive activation of gene expression by FXR and LXRα require Foxa2. Unexpectedly, Foxa2 occupancy is drastically increased when either receptor, FXR or LXRα, is bound by an agonist. In addition, co-immunoprecipitation experiments demonstrate that Foxa2 interacts with either receptor in a ligand-dependent manner, suggesting that Foxa2 and the receptor, bind DNA as an interdependent complex during ligand activation. Furthermore, PPARα binding is induced in Foxa2 mutants treated with FXR and LXR ligands, leading to the activation of PPARα targets. Conclusions Our model requires pioneering activity for ligand activation that challenges the existing ligand-independent binding mechanism. We also demonstrate that Foxa2 is required to achieve activation of the proper receptor – one that binds the added ligand – by repressing the activity of a competing receptor., Highlights • Foxa2 opens chromatin for FXR and LXRα binding during acute ligand activation. • Ligand-dependent activation of FXR & LXR-dependent gene expression requires Foxa2. • Foxa2 interacts with FXR and LXRα in a ligand-dependent manner. • Foxa2 restricts binding of competing receptor PPARα to ensure proper ligand-dependent activation of FXR and LXRα.
- Published
- 2021
34. Whole-exome sequencing identifies FOXL2, FOXA2 and FOXA3 as candidate genes for monogenic congenital anomalies of the kidneys and urinary tract
- Author
-
Nina Mann, Stuart B. Bauer, Chunyan Wang, Velibor Tasic, Shrikant Mane, Dervla M. Connaughton, Chen-Han Wilfred Wu, Luca Schierbaum, Natasa Stajic, Friedhelm Hildebrandt, Bixia Zheng, Makiko Nakayama, Sophia Schneider, Steve Seltzsam, Rufeng Dai, Hyun Joo Nam, and Shirlee Shril
- Subjects
Genetics ,Forkhead Box Protein L2 ,Vesico-Ureteral Reflux ,Transplantation ,Candidate gene ,business.industry ,Horseshoe kidney ,medicine.disease ,Kidney ,Nephrology ,Urogenital Abnormalities ,Exome Sequencing ,medicine ,Hepatocyte Nuclear Factor 3-beta ,Missense mutation ,Gene family ,Humans ,FOXA3 ,Original Article ,FOXA2 ,business ,Urinary Tract ,Gene ,Exome sequencing ,Hepatocyte Nuclear Factor 3-gamma - Abstract
BackgroundCongenital anomalies of the kidneys and urinary tract (CAKUT) constitute the most common cause of chronic kidney disease in the first three decades of life. Variants in four Forkhead box (FOX) transcription factors have been associated with CAKUT. We hypothesized that other FOX genes, if highly expressed in developing kidneys, may also represent monogenic causes of CAKUT.MethodsWe here performed whole-exome sequencing (WES) in 541 families with CAKUT and generated four lists of CAKUT candidate genes: (A) 36 FOX genes showing high expression during renal development, (B) 4 FOX genes known to cause CAKUT to validate list A, (C) 80 genes that we identified as unique potential novel CAKUT candidate genes when performing WES in 541 CAKUT families and (D) 175 genes identified from WES as multiple potential novel CAKUT candidate genes.ResultsTo prioritize potential novel CAKUT candidates in the FOX gene family, we overlapped 36 FOX genes (list A) with lists C and D of WES-derived CAKUT candidates. Intersection with list C identified a de novo FOXL2 in-frame deletion in a patient with eyelid abnormalities and ureteropelvic junction obstruction, and a homozygous FOXA2 missense variant in a patient with horseshoe kidney. Intersection with list D identified a heterozygous FOXA3 missense variant in a CAKUT family with multiple affected individuals.ConclusionsWe hereby identified FOXL2, FOXA2 and FOXA3 as novel monogenic candidate genes of CAKUT, supporting the utility of a paralog-based approach to discover mutated genes associated with human disease.
- Published
- 2021
35. MP33-18 LOSS OF FOXA2 IN NEUROENDOCRINE PROSTATE CANCER LEADS TO INDUCTION OF FUNCTIONAL ANDROGEN RECEPTOR
- Author
-
Xiuping Yu, Zachary M. Connelly, Siyuan Cheng, and Shu Yang
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Urology ,medicine.medical_treatment ,Cancer ,Gold standard (test) ,urologic and male genital diseases ,medicine.disease ,Androgen receptor ,Prostate cancer ,Internal medicine ,Medicine ,Hormone therapy ,FOXA2 ,business - Abstract
INTRODUCTION AND OBJECTIVE:Prostate cancer (PCa) is the leading diagnosed cancer in American men. Current treatments for PCa include surgery, radiation, and hormone therapy. The gold standard in th...
- Published
- 2021
- Full Text
- View/download PDF
36. Trophoblast glycoprotein is a marker for efficient sorting of ventral mesencephalic dopaminergic precursors derived from human pluripotent stem cells
- Author
-
Myung Soo Cho, Hye-Rim Shin, Dong-Youn Hwang, Jang-Hyeon Eom, Sanghyun Park, Kun Gu Lee, Dae-Sung Kim, Dongwook Kim, Mi-Young Jo, Jeong Eun Yoo, and Dongjin R. Lee
- Subjects
0301 basic medicine ,Chemistry ,Dopaminergic ,Trophoblast ,Cell sorting ,Embryonic stem cell ,Article ,TPBG ,Cell biology ,Cell therapy ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Pluripotent stem cells ,Neurology ,medicine ,Neurology. Diseases of the nervous system ,Neurology (clinical) ,FOXA2 ,RC346-429 ,Regeneration and repair in the nervous system ,Induced pluripotent stem cell ,030217 neurology & neurosurgery - Abstract
Successful cell therapy for Parkinson’s disease (PD) requires large numbers of homogeneous ventral mesencephalic dopaminergic (vmDA) precursors. Enrichment of vmDA precursors via cell sorting is required to ensure high safety and efficacy of the cell therapy. Here, using LMX1A-eGFP knock-in reporter human embryonic stem cells, we discovered a novel surface antigen, trophoblast glycoprotein (TPBG), which was preferentially expressed in vmDA precursors. TPBG-targeted cell sorting enriched FOXA2+LMX1A+ vmDA precursors and helped attain efficient behavioral recovery of rodent PD models with increased numbers of TH+, NURR1+, and PITX3+ vmDA neurons in the grafts. Additionally, fewer proliferating cells were detected in TPBG+ cell-derived grafts than in TPBG− cell-derived grafts. Our approach is an efficient way to obtain enriched bona fide vmDA precursors, which could open a new avenue for effective PD treatment.
- Published
- 2021
- Full Text
- View/download PDF
37. Tissue-specific transcription reprogramming promotes liver metastasis of colorectal cancer
- Author
-
Wei Guo, Yanmei Zhang, Ivan Garcia-Bassets, Zai Chang, Kequan Lin, Shuaishuai Teng, Pengyuan Wang, Zhi John Lu, Rui Qi, Ming Yang, Qunsheng Ji, Yiming Huang, Shasha Li, Yujing Cheng, Yang Eric Li, Dong Wang, Yang Cao, Qingyang Gu, Qianyu Wang, and Shanwen Chen
- Subjects
Transcriptional Activation ,Mice, Nude ,Biology ,Article ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,Transcription (biology) ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Hepatocyte Nuclear Factor 1-alpha ,Enhancer ,Molecular Biology ,Transcription factor ,030304 developmental biology ,Mice, Inbred BALB C ,0303 health sciences ,Liver Neoplasms ,Cell Biology ,Cellular Reprogramming ,medicine.disease ,HNF1A ,Gene Expression Regulation, Neoplastic ,Enhancer Elements, Genetic ,Liver ,Organ Specificity ,Cancer cell ,Hepatocyte Nuclear Factor 3-beta ,Cancer research ,Female ,FOXA2 ,Colorectal Neoplasms ,Transcriptome ,Reprogramming ,030217 neurology & neurosurgery - Abstract
Metastasis, the development of secondary malignant growths at a distance from a primary tumor, is the cause of death for 90% of cancer patients, but little is known about how metastatic cancer cells adapt to and colonize new tissue environments. Here, using clinical samples, patient-derived xenograft (PDX) samples, PDX cells, and primary/metastatic cell lines, we discovered that liver metastatic colorectal cancer (CRC) cells lose their colon-specific gene transcription program yet gain a liver-specific gene transcription program. We showed that this transcription reprogramming is driven by a reshaped epigenetic landscape of both typical enhancers and super-enhancers. Further, we identified that the liver-specific transcription factors FOXA2 and HNF1A can bind to the gained enhancers and activate the liver-specific gene transcription, thereby driving CRC liver metastasis. Importantly, similar transcription reprogramming can be observed in multiple cancer types. Our data suggest that reprogrammed tissue-specific transcription promotes metastasis and should be targeted therapeutically.
- Published
- 2019
- Full Text
- View/download PDF
38. Loss of FoxA2 accelerates neoplastic changes in the intrahepatic bile duct partly via the MAPK signaling pathway
- Author
-
Wei Peng, Xiaoyun Zhang, Junyi Shen, Chuan Li, Yongjie Zhou, Tianfu Wen, Qiang Zhou, Chihan Peng, and Yujun Shi
- Subjects
Adult ,Male ,Aging ,MAP Kinase Signaling System ,Cholangiocyte proliferation ,Intrahepatic bile ducts ,Gene Expression ,Thioacetamide ,Cholangiocarcinoma ,Mice ,Western blot ,intrahepatic cholangiocarcinoma ,Risk Factors ,Gene expression ,medicine ,Animals ,Humans ,Transcription factor ,Intrahepatic Cholangiocarcinoma ,reproductive and urinary physiology ,Aged ,Cell Proliferation ,Retrospective Studies ,Mice, Knockout ,biology ,medicine.diagnostic_test ,Cell Biology ,Neoplasms, Experimental ,respiratory system ,Middle Aged ,Prognosis ,TAA ,Bile Ducts, Intrahepatic ,Cell Transformation, Neoplastic ,Bile Duct Neoplasms ,embryonic structures ,biology.protein ,Cancer research ,MAPK signaling pathway ,Disease Progression ,Hepatocyte Nuclear Factor 3-beta ,Immunohistochemistry ,Female ,FOXA2 ,Neoplasm Recurrence, Local ,CREB1 ,Research Paper - Abstract
Background: Intrahepatic cholangiocarcinoma (ICC) is characterized by a highly aggressive nature and a dismal outcome. FOXA2 is an archetypal transcription factor involved in cholangiocyte proliferation. Results: FOXA2 expression was negatively correlated with tumor stage (p = 0.024). Univariate and multivariate analyses showed that low FoxA2 expression was associated with tumor relapse and survival. At 20 weeks after TAA administration, FoxA2-/- mice displayed significant manifestations of neoplasia, while WT mice did not. RNA sequencing analysis showed that the expression of genes in the MAPK signaling pathway was significantly higher in FoxA2-/- mice. IHC and Western blot results showed that p-ERK1/2, CREB1 and RAS were highly expressed in FoxA2-/- mice. Furthermore, using in vitro experiments with siRNA, we found that low expression of FoxA2 could exacerbate the metastatic potential of ICC. The expression of p-ERK1/2 and RAS, which are key mediators of the MAPK signaling pathway, was significantly increased. Conclusion: Low FOXA2 expression negatively affected the prognosis of patients with ICC. Loss of FoxA2 expression could promote intrahepatic bile duct neoplasia partly via activation of the MAPK signaling pathway. Materials and methods: In all, the data of 85 patients with ICC were retrospectively collected and analyzed. TAA was used to induce ICC in FoxA2-/- mice and WT mice. RNA-sequencing analysis was used to identify the expression of different genes.
- Published
- 2019
39. Long Noncoding RNA LL35/Falcor Regulates Expression of Transcription Factor Foxa2 in Hepatocytes in Normal and Fibrotic Mouse Liver
- Author
-
Ilia Kurochkin, S. A. Korinfskaya, Timofei S. Zatsepin, and Olga V. Sergeeva
- Subjects
0301 basic medicine ,Messenger RNA ,non-coding RNA ,transcription factor Foxa2 ,regulation ,respiratory system ,Biology ,liver ,Non-coding RNA ,Biochemistry ,Long non-coding RNA ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Downregulation and upregulation ,Transcription (biology) ,030220 oncology & carcinogenesis ,RNA splicing ,Molecular Medicine ,FOXA2 ,Molecular Biology ,Transcription factor ,Research Article ,Biotechnology - Abstract
Long noncoding RNAs (lncRNA) play important roles in the regulation of transcription, splicing, translation, and other processes in the cell. Human and mouse lncRNA (DEANR1 and LL35/Falcor, respectively) located in the genomic environment in close proximity to the Foxa2 transcription factor were discovered earlier. In this work, tissue-specific expression of LL35/Falcor lncRNA has been shown in mouse liver and lungs. The use of antisense oligonucleotides allowed us to achieve LL35/Falcor lncRNA downregulation by 90%. As a result, the level of Foxa2 mRNA and protein dropped, which confirms the involvement of LL35/Falcor lncRNA in the regulation of transcription factor Foxa2. We have shown a decrease in the expression of LL35 lncRNA in liver fibrosis, which correlates with the previously published data for mRNA Foxa2. Thus, lncRNA LL35 regulates Foxa2 expression in the liver not only in normal conditions, but also during development of fibrosis, which allows one to consider lncRNA a biomarker of this pathological process.
- Published
- 2019
- Full Text
- View/download PDF
40. The Expression of CNS-Specific PPARGC1A Transcripts Is Regulated by Hypoxia and a Variable GT Repeat Polymorphism
- Author
-
Soyal, Selma M., Bonova, Petra, Kwik, Markus, Zara, Greta, Auer, Simon, Scharler, Cornelia, Strunk, Dirk, Nofziger, Charity, Paulmichl, Markus, and Patsch, Wolfgang
- Subjects
Central Nervous System ,Male ,Polymorphism, Genetic ,Base Sequence ,PGC-1α ,Microsatellite ,CNS-specific isoforms ,ESRRA ,Ciclopirox ,Hypoxia-Inducible Factor 1, alpha Subunit ,Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha ,Article ,Brain Ischemia ,Cell Line ,Gene Expression Regulation ,Organ Specificity ,PPARGC1A ,Animals ,Humans ,FOXA2 ,RNA, Messenger ,Rats, Wistar ,Hypoxia ,Promoter Regions, Genetic ,Repetitive Sequences, Nucleic Acid - Abstract
PPARGC1A encodes a transcriptional co-activator also termed peroxisome proliferator-activated receptor (PPAR) gamma coactivator 1-alpha (PGC-1α) which orchestrates multiple transcriptional programs. We have recently identified CNS-specific transcripts that are initiated far upstream of the reference gene (RG) promoter. The regulation of these isoforms may be relevant, as experimental and genetic studies implicated the PPARGC1A locus in neurodegenerative diseases. We therefore studied cis- and trans-regulatory elements activating the CNS promoter in comparison to the RG promoter in human neuronal cell lines. A naturally occurring variable guanidine thymidine (GT) repeat polymorphism within a microsatellite region in the proximal CNS promoter increases promoter activity in neuronal cell lines. Both the RG and the CNS promoters are activated by ESRRA, and the PGC-1α isoforms co-activate ESRRA on their own promoters suggesting an autoregulatory feedback loop. The proximal CNS, but not the RG, promoter is induced by FOXA2 and co-activated by PGC-1α resulting in robust activation. Furthermore, the CNS, but not the RG, promoter is targeted by the canonical hypoxia response involving HIF1A. Importantly, the transactivation by HIF1A is modulated by the size of the GT polymorphism. Increased expression of CNS-specific transcripts in response to hypoxia was observed in an established rat model, while RG transcripts encoding the full-length reference protein were not increased. These results suggest a role of the CNS region of the PPARGC1A locus in ischemia and warrant further studies in humans as the activity of the CNS promoter as well as its induction by hypoxia is subject to inter-individual variability due to the GT polymorphism. Electronic supplementary material The online version of this article (10.1007/s12035-019-01731-5) contains supplementary material, which is available to authorized users.
- Published
- 2019
41. Pre-marked chromatin and transcription factor co-binding shape the pioneering activity of Foxa2
- Author
-
Zeyang Wang, Ida M. Evenroed, Filippo M. Cernilogar, Ingo Burtscher, Stefan Hasenöder, Gregor D. Gilfillan, Gunnar Schotta, Heiko Lickert, Michael Sterr, Sophia Groh, Pawel Smialowski, and Katharina Scheibner
- Subjects
Cell ,Biology ,Histones ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Genetics ,medicine ,Animals ,Histone code ,Binding site ,Transcription factor ,reproductive and urinary physiology ,030304 developmental biology ,Mice, Knockout ,Regulation of gene expression ,0303 health sciences ,Binding Sites ,Models, Genetic ,Endoderm ,Gene regulation, Chromatin and Epigenetics ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Mouse Embryonic Stem Cells ,Chromatin Assembly and Disassembly ,Embryonic stem cell ,Chromatin ,ddc ,GATA4 Transcription Factor ,Cell biology ,Histone Code ,medicine.anatomical_structure ,chemistry ,embryonic structures ,Hepatocyte Nuclear Factor 3-beta ,FOXA2 ,030217 neurology & neurosurgery ,DNA ,Signal Transduction - Abstract
Pioneer transcription factors (PTF) can recognize their binding sites on nucleosomal DNA and trigger chromatin opening for recruitment of other non-pioneer transcription factors. However, critical properties of PTFs are still poorly understood, such as how these transcription factors selectively recognize cell type-specific binding sites and under which conditions can they can initiate chromatin remodelling. Here we show that early endoderm binding sites of the paradigm PTF Foxa2 are epigenetically primed by low levels of active chromatin modifications in embryonic stem cells (ESC). Priming of these binding sites is supported by preferential recruitment of Foxa2 to endoderm binding sites compared to lineage-inappropriate binding sites, when ectopically expressed in ESCs. We further show that binding of Foxa2 is required for chromatin opening during endoderm differentiation. However, increased chromatin accessibility was only detected on binding sites which are synergistically bound with other endoderm transcription factors. Thus, our data suggest that binding site selection of PTFs is directed by the chromatin environment and that chromatin opening requires collaboration of PTFs with additional transcription factors.
- Published
- 2019
- Full Text
- View/download PDF
42. The Use of Foxa2-Overexpressing Adipose Tissue-Derived Stem Cells in a Scaffold System Attenuates Acute Liver Injury
- Author
-
Yeon Ji Chae, Jai Sun Lee, Hyeon Tae Kang, Dae Won Jun, Waqar Khalid Saeed, Kiseok Jang, and Jin Ho Lee
- Subjects
medicine.medical_treatment ,Adipose tissue ,Biocompatible Materials ,Thioacetamide ,Connexins ,Mice ,0302 clinical medicine ,Nude mouse ,Cytochrome P-450 Enzyme System ,Polylactic Acid-Polyglycolic Acid Copolymer ,Cells, Cultured ,Mesenchymal stem cell ,Liver injury ,education.field_of_study ,Tissue Scaffolds ,biology ,Gastroenterology ,acute ,Cell Differentiation ,Stem-cell therapy ,Electroporation ,Liver ,030220 oncology & carcinogenesis ,Hepatocyte Nuclear Factor 3-beta ,Connexin 32 ,Original Article ,030211 gastroenterology & hepatology ,alpha-Fetoproteins ,Chemical and Drug Induced Liver Injury ,Stem cell ,Glycogen ,Plasmids ,Dipeptidyl Peptidase 4 ,Mice, Nude ,Foxa2 ,Mesenchymal Stem Cell Transplantation ,Scaffold ,03 medical and health sciences ,Albumins ,medicine ,Animals ,education ,Keratin-18 ,Hepatology ,business.industry ,Liver failure ,Mesenchymal Stem Cells ,medicine.disease ,biology.organism_classification ,Hepatocytes ,Cancer research ,business ,Homing (hematopoietic) - Abstract
Background/Aims: For the clinical application of stem cell therapy, functional enhancement is needed to increase the survival rate and the engraftment rate. The purpose of this study was to investigate functional enhancement of the paracrine effect using stem cells and hepatocyte-like cells and to minimize stem cell homing by using a scaffold system in a liver disease model. Methods: A microporator was used to overexpress Foxa2 in adipose tissue-derived stem cells (ADSCs), which were cultured in a poly(lactic-co-glycolic acid) (PLGA) scaffold. Later, the ADSCs were cultured in hepatic differentiation medium for 2 weeks by a 3-step method. For in vivo experiments, Foxa2-overexpressing ADSCs were loaded in the scaffold, cultured in hepatic differentiation medium and later were implanted in the dorsa of nude mice subjected to acute liver injury (thioacetamide intraperitoneal injection). Results: Foxa2-overexpressing ADSCs showed greater increases in hepatocyte-specific gene markers (alpha fetoprotein [AFP], cytokeratin 18 [CK18], and albumin), cytoplasmic glycogen storage, and cytochrome P450 expression than cells that underwent the conventional differentiation method. In vivo experiments using the nude mouse model showed that 2 weeks after scaffold implantation, the mRNA expression of AFP, CK18, dipeptidyl peptidase 4 (CD26), and connexin 32 (CX32) was higher in the Foxa2-overexpressing ADSCs group than in the ADSCs group. The Foxa2-overexpressing ADSCs scaffold treatment group showed attenuated liver injury without stem cell homing in the thioacetamideinduced acute liver injury model. Conclusions: Foxa2-overexpressing ADSCs applied in a scaffold system enhanced hepatocyte-like differentiation and attenuated acute liver damage in an acute liver injury model without homing effects. (Gut Liver 2019;13:450-460)
- Published
- 2019
- Full Text
- View/download PDF
43. Integrative analysis of the forkhead box A2 (FOXA2) cistrome for the human endometrium
- Author
-
Thomas E. Spencer, Andrew M. Kelleher, Steven L. Young, Gregory W. Burns, Francesco J. DeMayo, and Susanta K. Behura
- Subjects
Adult ,0301 basic medicine ,Biology ,Endometrium ,Biochemistry ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Pregnancy ,Gene expression ,Genetics ,medicine ,Humans ,Embryo Implantation ,Molecular Biology ,Transcription factor ,reproductive and urinary physiology ,Research ,Uterus ,Decidualization ,respiratory system ,Cell biology ,DNA binding site ,Fertility ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Cistrome ,embryonic structures ,Hepatocyte Nuclear Factor 3-beta ,Female ,FOXA2 ,Stromal Cells ,Uterine gland ,030217 neurology & neurosurgery ,Transcription Factors ,Biotechnology - Abstract
The pioneer forkhead box (FOX)A2 transcription factor is specifically expressed in the glands of the uterus, which are central to endometrial function and fertility. In mice, FOXA2 is a critical regulator of uterine gland development in the neonate and gland function in the adult. An integrative approach was used here to define the FOXA2 cistrome in the human endometrium. Genome-wide mapping of FOXA2 binding intervals by chromatin immunoprecipitation sequencing was performed using proliferative (P)- and midsecretory (MS)-phase endometrium and integrated with the transcriptome determined by RNA sequencing. Distinctive FOXA2 binding intervals, enriched for different transcription factor binding site motifs, were detected in the P and MS endometrium. Pathway analysis revealed different biologic processes regulated by genes with FOXA2 binding intervals in the P and MS endometrium. Thus, FOXA2 is postulated to regulate gene expression in concert with other transcription factors and impact uterine gland development and function in a cycle phase–dependent manner. Analyses also identified potential FOXA2-regulated genes that influence uterine receptivity, blastocyst implantation, and stromal cell decidualization, which are key events in pregnancy establishment.—Kelleher, A. M., Behura, S. K., Burns, G. W., Young, S. L., DeMayo, F. J., Spencer, T. E. Integrative analysis of the forkhead box A2 (FOXA2) cistrome for the human endometrium.
- Published
- 2019
- Full Text
- View/download PDF
44. PGC-1β cooperating with FOXA2 inhibits proliferation and migration of breast cancer cells
- Author
-
Rong Ma, Jia Wang, Xi Wang, Jia Cao, Huimin Feng, Shihai Liu, Libin Wang, Danni Wang, and Xiaohan Li
- Subjects
Cancer Research ,Proliferation ,PGC-1β ,Biology ,medicine.disease_cause ,lcsh:RC254-282 ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Downregulation and upregulation ,Genetics ,medicine ,lcsh:QH573-671 ,Migration ,Gene knockdown ,Cell growth ,lcsh:Cytology ,Cancer ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Blot ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,FOXA2 ,Signal transduction ,Carcinogenesis ,Primary Research - Abstract
Background Breast cancer is one of the most common malignancy among females from the worldwide cancer incidence statistics. Peroxisome gamma coactivator-1β (PGC-1β) has long been identified to be involved in this type of tumorigenesis. However, the mechanisms of PGC-1β in human breast cancer have not been fully understood and the function requires to be further elucidated. Methods mRNA and protein expression of PGC-1β and FOXA2 in breast cancer tissues and cell lines were determined by qRT-PCR and Western Blotting, respectively. To further visualize the expression and localization of PGC-1β and FOXA2, immunochemistry and immunofluorescence staining methods were employed. The effect of PGC-1β and FOXA2 on cell proliferation and migration were evaluated by CCK8, clone formation, transwell and wound-healing assays, which has been done either with stable PGC-1β knockdown or FOXA2 overexpression in vitro. Xenografts model of nude mice were used to evaluate tumor growth in vivo. In addition, proteins expression of the PI3K-AKT-mTOR signaling pathway involved in the regulation of breast cancer were detected by Western Blotting. Results Our results showed that PGC-1β was upregulated and FOXA2 was downregulated in breast cancer tissues and cell lines. These two proteins can be interacted with each other to form the complex. Also, we found the combination of PGC-1β interference with FOXA2 overexpression significantly inhibited cell proliferation and migration in vitro as well as tumor growth in vivo. We further identified that PGC-1β and FOXA2 strongly correlated with the PI3K-AKT-mTOR signaling pathway, and they exerted their biological functions by activating this pathway. Conclusions We demonstrated that downregulation of PGC-1β combined with overexpression of FOXA2 obviously inhibited the function of breast cancer cells through regulating the PI3K-AKT-mTOR pathway.
- Published
- 2019
- Full Text
- View/download PDF
45. Reduced Immunohistochemical Expression of Hnf1β and FoxA2 in Liver Tissue Can Discriminate Between Biliary Atresia and Other Causes of Neonatal Cholestasis
- Author
-
Noha L Ibrahim, Nermine A. Ehsan, Mohamed O Abd El-Fatah, Ghada M Naser, Mervat M Sultan, and Usama F Shaalan
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Histology ,Gastroenterology ,Pathology and Forensic Medicine ,03 medical and health sciences ,0302 clinical medicine ,Biliary Atresia ,Biliary atresia ,Fibrosis ,Internal medicine ,Humans ,Medicine ,Neonatal cholestasis ,Stage (cooking) ,Hepatocyte Nuclear Factor 1-beta ,Retrospective Studies ,Cholestasis ,business.industry ,Infant ,Retrospective cohort study ,medicine.disease ,Immunohistochemistry ,Medical Laboratory Technology ,Hepatocyte nuclear factors ,030104 developmental biology ,Gene Expression Regulation ,Liver ,030220 oncology & carcinogenesis ,Hepatocyte Nuclear Factor 3-beta ,Female ,FOXA2 ,business - Abstract
Biliary atresia (BA) is a necroinflammatory occlusive cholangiopathy that affects infants. Genetic and environmental factors has been proposed for its occurrence. The objectives of this study was to investigate the protein expression of 2 important genes regulating ductal plate remodeling, hepatocyte nuclear factor 1-beta (Hnf1β) and the fork head box protein A2 (FoxA2) in liver tissue from patients with BA and to compare their expression with other causes of neonatal cholestasis (NC). This retrospective study included 60 pediatric patients, 30 with BA and 30 with NC. Immunohistochemistry of Hnf1β and FoxA2 was performed on liver tissues from studied patients as well as 20 healthy subjects. Statistical analysis between immunohistochemistry results and other parameters was performed. Liver tissue from patients with BA revealed reduced Hnf1β and FoxA2 immunoexpression. A strong significant statistical difference between BA and NC group (P
- Published
- 2019
- Full Text
- View/download PDF
46. Exome sequencing identifies a de novo FOXA2 variant in a patient with syndromic diabetes
- Author
-
Michel Guipponi, Caroline Stekelenburg, Mariarosaria Lang-Muritano, Karine Gerster, Jean-Louis Blouin, Federico Santoni, Valerie M. Schwitzgebel, University of Zurich, and Schwitzgebel, Valerie M
- Subjects
Models, Molecular ,Male ,Endocrinology, Diabetes and Metabolism ,DNA Mutational Analysis ,Bioinformatics ,Pediatrics ,Whole Exome Sequencing ,Diabetes mellitus genetics ,Endocrinology ,0302 clinical medicine ,Models ,Medicine ,Missense mutation ,ddc:576.5 ,030212 general & internal medicine ,Child ,Exome ,Index case ,Exome sequencing ,ddc:618 ,Single Nucleotide ,Syndrome ,Perinatology ,3. Good health ,and Child Health ,Diabetes and Metabolism ,2712 Endocrinology, Diabetes and Metabolism ,Hepatocyte Nuclear Factor 3-beta ,Proline ,Mutation, Missense ,DNA Mutational Analysis/methods ,610 Medicine & health ,030209 endocrinology & metabolism ,Polymorphism, Single Nucleotide ,03 medical and health sciences ,Leucine ,Diabetes mellitus ,Exome Sequencing ,Diabetes Mellitus ,Internal Medicine ,Humans ,2735 Pediatrics, Perinatology and Child Health ,Polymorphism ,Hepatocyte Nuclear Factor 3-beta/chemistry/genetics ,business.industry ,Neonatal hypoglycemia ,Molecular ,Diabetes Mellitus/congenital/genetics ,Leucine/genetics ,medicine.disease ,Amino Acid Substitution ,10036 Medical Clinic ,2724 Internal Medicine ,Mutation ,Pediatrics, Perinatology and Child Health ,Proline/genetics ,FOXA2 ,Missense ,business - Abstract
When diabetes is associated with congenital malformations, without autoimmune antibodies, a genetic cause is suspected. Here, we aimed to identify a defective gene that led to diabetes. We performed an exome analysis of an index case and his healthy parents. The child presented with childhood-onset diabetes, congenital hypopituitarism, cardiac malformation, and anal atresia. A DNA analysis revealed a heterozygous de novo pathogenic variant in the developmental transcription factor, forkhead box A2 (FOXA2). The mutation resided in the DNA-binding domain, which is highly conserved among species. Tridimensional molecular dynamics simulation modeling predicted an altered interaction between the mutated protein and DNA. A defect in the FOXA2 DNA-binding domain was associated with childhood-onset diabetes and multiple congenital anomalies, which reflected the pleiotropic nature of the gene. This report extends the recently described phenotype of neonatal hypoglycemia to later-onset diabetes. We suggest to include FOXA2 analysis for neonatal hypoglycemia and to implement a long-term follow-up, particularly for the risk of diabetes.
- Published
- 2019
- Full Text
- View/download PDF
47. Expression and prognosis analyses of forkhead box A (FOXA) family in human lung cancer
- Author
-
Chao-Qun Huang, Jiuyang Liu, Yutaka Yonemura, Xiao-Jun Yang, and Bin Xiong
- Subjects
Hepatocyte Nuclear Factor 3-alpha ,0301 basic medicine ,Lung Neoplasms ,Transcription, Genetic ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Databases, Genetic ,Genetics ,medicine ,Forkhead Box ,Humans ,RNA, Messenger ,Lung cancer ,Gene ,Human lung cancer ,Gene Expression Profiling ,Computational Biology ,Cancer ,General Medicine ,respiratory system ,Prognosis ,medicine.disease ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Multigene Family ,030220 oncology & carcinogenesis ,Cancer research ,FOXA3 ,FOXA2 ,FOXA1 ,Hepatocyte Nuclear Factor 3-gamma - Abstract
Despite advances in early diagnosis and treatment, cancer still remains the major reason of mortality worldwide. The forkhead box A (FOXA) family is reported to participate in diverse human diseases. However, little is known about their expression and prognostic values in human lung cancer. Herein, we conducted a detailed cancer vs. normal analysis. The mRNA expression levels of FOXA family in numerous kind of cancers, including lung cancer, were analyzed using the Oncomine and GEPIA database. We observed that the mRNA expression levels of FOXA1, and FOXA3 were all increased while FOXA2 were decreased in most cancers compared with normal tissues, especially in lung cancer. Moreover, the expression levels of FOXA1, and FOXA3 are also highly expressed, while FOXA2 were decreased in almost all cancer cell lines, particularly in lung cancer cell lines, analyzing by Cancer Cell Line Encyclopedia (CCLE) and EMBL-EBI databases. Furthermore, the LinkedOmics database was used to evaluate the prognostic values, indicating that higher expression of FOXA1, FOXA3 indicated a poor overall survival (OS), while increased FOXA2 revealed a better OS in lung cancer. To conclusion, FOXA family showed significant expression differences between cancer and normal tissues, especially lung cancer, and FOXA1, FOXA3 could be promising prognostic biomarkers for lung cancer.
- Published
- 2019
- Full Text
- View/download PDF
48. Regulation of the Pancreatic Exocrine Differentiation Program and Morphogenesis by Onecut 1/Hnf6Summary
- Author
-
Maureen Gannon, Peter A. Kropp, and Xiaodong Zhu
- Subjects
0301 basic medicine ,PDAC, pancreatic ductal adenocarcinoma ,Acinar Cells ,Epithelium ,Nr5a2, nuclear receptor subfamily 5, group A, member 2 ,Transcriptome ,Mice ,0302 clinical medicine ,Transcriptional regulation ,Morphogenesis ,MPC, multipotent pancreatic progenitor cell ,Ptf1a, Pancreas transcription factor, 1a ,Original Research ,Gastroenterology ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Pancreas, Exocrine ,Pancreas Development ,Cell biology ,Hepatocyte Nuclear Factor 6 ,medicine.anatomical_structure ,Cym, Chymosin ,030211 gastroenterology & hepatology ,Pancreas ,Inhba, Inhibin, Beta A ,Sox9, SRY-related HMG-box 9 ,ChIP-Seq, chromatin immunoprecipitation followed by high-throughput sequencing ,RNA-Seq, RNA-sequencing ,Hnf6, hepatocyte nuclear factor 6 ,Biology ,TUNEL, terminal deoxynucleotidyl transferase dUTP nick end labeling ,ADM, acinar-to-ductal metaplasia ,03 medical and health sciences ,CK19, Cytokeratin 19 ,medicine ,Acinar cell ,Animals ,Ihh, Indian hedgehog ,Mist1, muscle, intestine, stomach transcription factor 1 ,lcsh:RC799-869 ,Transcription factor ,Hnf1β, hepatocyte nuclear factor 1β ,Cell Proliferation ,Pancreatic duct ,Oc1, Onecut1 ,Hepatology ,Base Sequence ,Ptch2, Patched 2 ,Smo, Smoothened ,HH, Hedgehog ,Embryo, Mammalian ,Spink 1, serine protease inhibitor Kazal type 1 ,030104 developmental biology ,Animals, Newborn ,lcsh:Diseases of the digestive system. Gastroenterology ,FOXA2 ,Pdx1, pancreatic and duodenal homeobox 1 ,Exocrine - Abstract
Background & Aims The Onecut 1 transcription factor (Oc1, a.k.a. HNF6) promotes differentiation of endocrine and duct cells of the pancreas; however, it has no known role in acinar cell differentiation. We sought to better understand the role of Oc1 in exocrine pancreas development and to identify its direct transcriptional targets. Methods Pancreata from Oc1Δpanc (Oc1fl/fl;Pdx1-Cre) mouse embryos and neonates were analyzed morphologically. High-throughput RNA-sequencing was performed on control and Oc1-deficient pancreas; chromatin immunoprecipitation sequencing was performed on wild-type embryonic mouse pancreata to identify direct Oc1 transcriptional targets. Immunofluorescence labeling was used to confirm the RNA-sequencing /chromatin immunoprecipitation sequencing results and to further investigate the effects of Oc1 loss on acinar cells. Results Loss of Oc1 from the developing pancreatic epithelium resulted in disrupted duct and acinar cell development. RNA-sequencing revealed decreased expression of acinar cell regulatory factors (Nr5a2, Ptf1a, Gata4, Mist1) and functional genes (Amylase, Cpa1, Prss1, Spink1) at embryonic day (e) 18.5 in Oc1Δpanc samples. Approximately 1000 of the altered genes were also identified as direct Oc1 targets by chromatin immunoprecipitation sequencing, including most of the previously noted genes. By immunolabeling, we confirmed that Amylase, Mist1, and GATA4 protein levels are significantly decreased by P2, and Spink1 protein levels were significantly reduced and mislocalized. The pancreatic duct regulatory factors Hnf1β and FoxA2 were also identified as direct Oc1 targets. Conclusions These findings confirm that Oc1 is an important regulator of both duct and acinar cell development in the embryonic pancreas. Novel transcriptional targets of Oc1 have now been identified and provide clarity into the mechanisms of Oc1 transcriptional regulation in the developing exocrine pancreas. Oc1 can now be included in the gene-regulatory network of acinar cell regulatory genes. Oc1 regulates other acinar cell regulatory factors and acinar cell functional genes directly, and it can also regulate some acinar cell regulatory factors (eg, Mist1) indirectly. Oc1 therefore plays an important role in acinar cell development., Graphical abstract
- Published
- 2019
49. FoxA1 and FoxA2 regulate growth and cellular identity in NKX2-1-positive lung adenocarcinoma
- Author
-
Grace Orstad, Gabriela Fort, Timothy J Parnell, Alex Jones, Chris Stubben, Brian Lohman, Katherine L. Gillis, Walter Orellana, Rushmeen Tariq, Olaf Klingbeil, Klaus Kaestner, Christopher R. Vakoc, Benjamin T. Spike, and Eric L. Snyder
- Subjects
Lung ,medicine.anatomical_structure ,Cancer cell ,Gene expression ,medicine ,Cancer research ,Adenocarcinoma ,Cancer ,FOXA2 ,Biology ,FOXA1 ,medicine.disease ,Gene - Abstract
Change in cancer cell identity is well characterized as a mechanism of cancer progression and acquired resistance to targeted therapies. Lung adenocarcinoma (LUAD) exhibits significant heterogeneity in cell identity and differentiation state; these characteristics correlate directly with prognosis, response to available therapies, and acquisition of drug resistance. In previous work, we have shown that FoxA1 and FoxA2 (FoxA1/2) activate a gastric differentiation program in NKX2-1-negative LUAD. Here we investigate the role of FoxA1/2 in NKX2-1-positive LUAD. We find that FoxA1/2 are consistently expressed in NKX2-1-positive human LUAD. Foxa1/2 deletion severely impairs proliferation and significantly prolongs overall survival in a genetically engineered mouse model of KRAS-driven LUAD. FoxA1/2 activate expression of a mixed-lineage transcriptional program characterized by co-expression of pulmonary (Alveolar Type II) and gastrointestinal marker genes. Loss of FoxA1/2 causes a lineage switch, activating gene expression programs associated with Alveolar Type I cells and maturing squamous epithelial cells. Inhibition of NKX2-1 partially rescues the antiproliferative impact of FoxA1/2 loss, showing that NKX2-1 retains some degree of activity in FoxA1/2-null cells, despite its inability to activate canonical target genes. In summary, this study identifies FoxA1/2 expression as a novel vulnerability in NKX2-1 positive LUAD and shows that FoxA1/2 actively regulate cellular identity in this disease.
- Published
- 2021
- Full Text
- View/download PDF
50. Onset of taste bud cell renewal starts at birth and coincides with a shift in SHH function
- Author
-
Jennifer K. Scott, Linda A. Barlow, Eric D. Larson, Erin Golden, Timothy J Fellin, Kenneth L. Jones, Dany Gaillard, G Devon Trahan, and Lauren A Shechtman
- Subjects
0301 basic medicine ,Male ,Mouse ,SOX2 ,Hedgehog signaling ,taste ,0302 clinical medicine ,Taste receptor ,Cell Movement ,Biology (General) ,Sonic hedgehog ,Cell Self Renewal ,General Neuroscience ,Stem Cells ,Gene Expression Regulation, Developmental ,Cell Differentiation ,General Medicine ,RNAseq ,Taste Buds ,Stem Cells and Regenerative Medicine ,Hedgehog signaling pathway ,Cell biology ,medicine.anatomical_structure ,embryonic structures ,Hepatocyte Nuclear Factor 3-beta ,Medicine ,Female ,taste bud ,Stem cell ,Research Article ,Signal Transduction ,Hepatocyte Nuclear Factor 3-alpha ,QH301-705.5 ,Science ,Biology ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,stomatognathic system ,Taste bud ,medicine ,Cell Adhesion ,Animals ,Cell Lineage ,Hedgehog Proteins ,Progenitor cell ,Cell adhesion ,General Immunology and Microbiology ,Keratin-14 ,Epithelial Cells ,Embryonic stem cell ,030104 developmental biology ,Animals, Newborn ,regeneration ,biology.protein ,FOXA2 ,FOXA1 ,Transcriptome ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Embryonic taste bud primordia are specified as taste placodes on the tongue surface and differentiate into the first taste receptor cells (TRCs) at birth. Throughout adult life, TRCs are continually regenerated from epithelial progenitors. Sonic hedgehog (SHH) signaling regulates TRC development and renewal, repressing taste fate embryonically, but promoting TRC differentiation in adults. Here, using mouse models, we show TRC renewal initiates at birth and coincides with onset of SHHs pro-taste function. Using transcriptional profiling to explore molecular regulators of renewal, we identified Foxa1 and Foxa2 as potential SHH target genes in lingual progenitors at birth and show that SHH overexpression in vivo alters FoxA1 and FoxA2 expression relevant to taste buds. We further bioinformatically identify genes relevant to cell adhesion and cell locomotion likely regulated by FOXA1;FOXA2 and show that expression of these candidates is also altered by forced SHH expression. We present a new model where SHH promotes TRC differentiation by regulating changes in epithelial cell adhesion and migration.
- Published
- 2021
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.