151
|
Wang L, Shangguan S, Xin Y, Chang S, Wang Z, Lu X, Wu L, Niu B, Zhang T. Folate deficiency disturbs hsa-let-7 g level through methylation regulation in neural tube defects. J Cell Mol Med 2017. [PMID: 28631291 PMCID: PMC5706510 DOI: 10.1111/jcmm.13228] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Folic acid deficiency during pregnancy is believed to be a high‐risk factor for neural tube defects (NTDs). Disturbed epigenetic modifications, including miRNA regulation, have been linked to the pathogenesis of NTDs in those with folate deficiency. However, the mechanism by which folic acid‐regulated miRNA influences this pathogenesis remains unclear. It is believed that DNA methylation is associated with dysregulated miRNA expression. To clarify this issue, here we measured the methylation changes of 22 miRNAs in 57 human NTD cases to explore whether such changes are involved in miRNA regulation in NTD cases through folate metabolism. In total, eight of the 22 miRNAs tested reduced their methylation modifications in NTD cases, which provide direct evidence of the roles of interactions between DNA methylation and miRNA level in these defects. Among the findings, there was a significant association between folic acid concentration and hsa‐let‐7 g methylation level in NTD cases. Hypomethylation of hsa‐let‐7 g increased its own expression level in both NTD cases and cell models, which indicated that hsa‐let‐7 g methylation directly regulates its own expression. Overexpression of hsa‐let‐7 g, along with its target genes, disturbed the migration and proliferation of SK‐N‐SH cells, implying that hsa‐let‐7 g plays important roles in the prevention of NTDs by folic acid. In summary, our data suggest a relationship between aberrant methylation of hsa‐let‐7 g and disturbed folate metabolism in NTDs, implying that improvements in nutrition during early pregnancy may prevent such defects, possibly via the donation of methyl groups for miRNAs.
Collapse
Affiliation(s)
- Li Wang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Shaofang Shangguan
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Yu Xin
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Shaoyan Chang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Zhen Wang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Xiaolin Lu
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Lihua Wu
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Bo Niu
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| | - Ting Zhang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China
| |
Collapse
|
152
|
Motaung TE, Saitoh H, Tsilo TJ. Large-scale molecular genetic analysis in plant-pathogenic fungi: a decade of genome-wide functional analysis. MOLECULAR PLANT PATHOLOGY 2017; 18:754-764. [PMID: 27733021 PMCID: PMC6638310 DOI: 10.1111/mpp.12497] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 10/07/2016] [Accepted: 10/08/2016] [Indexed: 05/31/2023]
Abstract
Plant-pathogenic fungi cause diseases to all major crop plants world-wide and threaten global food security. Underpinning fungal diseases are virulence genes facilitating plant host colonization that often marks pathogenesis and crop failures, as well as an increase in staple food prices. Fungal molecular genetics is therefore the cornerstone to the sustainable prevention of disease outbreaks. Pathogenicity studies using mutant collections provide immense function-based information regarding virulence genes of economically relevant fungi. These collections are rich in potential targets for existing and new biological control agents. They contribute to host resistance breeding against fungal pathogens and are instrumental in searching for novel resistance genes through the identification of fungal effectors. Therefore, functional analyses of mutant collections propel gene discovery and characterization, and may be incorporated into disease management strategies. In the light of these attributes, mutant collections enhance the development of practical solutions to confront modern agricultural constraints. Here, a critical review of mutant collections constructed by various laboratories during the past decade is provided. We used Magnaporthe oryzae and Fusarium graminearum studies to show how mutant screens contribute to bridge existing knowledge gaps in pathogenicity and fungal-host interactions.
Collapse
Affiliation(s)
- Thabiso E. Motaung
- Agricultural Research Council ‐ Small Grain InstitutePrivate Bag X29Bethlehem9700South Africa
| | - Hiromasa Saitoh
- Iwate Biotechnology Research Center22‐174‐4 NaritaKitakamiIwate024‐0003Japan
| | - Toi J. Tsilo
- Agricultural Research Council ‐ Small Grain InstitutePrivate Bag X29Bethlehem9700South Africa
- Department of Life and Consumer SciencesUniversity of South AfricaPO Box 392Pretoria0003South Africa
| |
Collapse
|
153
|
Garon G, Bergeron F, Brousseau C, Robert NM, Tremblay JJ. FOXA3 Is Expressed in Multiple Cell Lineages in the Mouse Testis and Regulates Pdgfra Expression in Leydig Cells. Endocrinology 2017; 158:1886-1897. [PMID: 28379539 DOI: 10.1210/en.2016-1736] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 03/29/2017] [Indexed: 12/16/2022]
Abstract
The three FOXA transcription factors are mainly known for their roles in the liver. However, Foxa3-deficient mice become progressively sub/infertile due to germ cell loss. Because no data were available regarding the localization of the FOXA3 protein in the testis, immunohistochemistry was performed on mouse testis sections. In the fetal testis, a weak but consistent staining for FOXA3 is detected in the nucleus of Sertoli cells. In prepubertal and adult life, FOXA3 remains present in Sertoli cells of some but not all seminiferous tubules. FOXA3 is also detected in the nucleus of some peritubular cells. From postnatal day 20 onward, FOXA3 is strongly expressed in the nucleus of Leydig cells. To identify FOXA3 target genes in Leydig cells, MLTC-1 Leydig cells were transfected with a series of Leydig cell gene reporters in the presence of a FOXA3 expression vector. The platelet-derived growth factor receptor α (Pdgfra) promoter was significantly activated by FOXA3. The Pdgfra promoter contains three potential FOX elements and progressive 5' deletions and site-directed mutagenesis revealed that the most proximal element at -78 bp was sufficient to confer FOXA3 responsiveness. FOXA3 from Leydig cells could bind to this element in vitro (electrophoretic mobility shift assay) and was recruited to the proximal Pdgfra promoter in vivo (chromatin immunoprecipitation). Finally, endogenous Pdgfra messenger RNA levels were reduced in FOXA3-deficient MLTC-1 Leydig cells. Taken together, our data identify FOXA3 as a marker of the Sertoli cell lineage and of the adult Leydig cell population, and as a regulator of Pdgfra transcription in Leydig cells.
Collapse
Affiliation(s)
- Gabriel Garon
- Reproduction, Mother and Child Health, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, Québec G1V 4G2, Canada
| | - Francis Bergeron
- Reproduction, Mother and Child Health, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, Québec G1V 4G2, Canada
| | - Catherine Brousseau
- Reproduction, Mother and Child Health, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, Québec G1V 4G2, Canada
| | - Nicholas M Robert
- Reproduction, Mother and Child Health, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, Québec G1V 4G2, Canada
| | - Jacques J Tremblay
- Reproduction, Mother and Child Health, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec City, Québec G1V 4G2, Canada
- Centre for Research in Reproduction, Development, and Intergenerational Health, Department of Obstetrics, Gynecology, and Reproduction, Faculty of Medicine, Université Laval, Québec City, Québec G1V 0A6, Canada
| |
Collapse
|
154
|
Von Stetina SE, Liang J, Marnellos G, Mango SE. Temporal regulation of epithelium formation mediated by FoxA, MKLP1, MgcRacGAP, and PAR-6. Mol Biol Cell 2017; 28:2042-2065. [PMID: 28539408 PMCID: PMC5509419 DOI: 10.1091/mbc.e16-09-0644] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 05/18/2017] [Accepted: 05/18/2017] [Indexed: 12/15/2022] Open
Abstract
During embryo morphogenesis, minor epithelia are generated after, and then form bridges between, major epithelia (e.g., epidermis and gut). In Caenorhabditis elegans, this delay is regulated by four proteins that control production and localization of polarity proteins: the pioneer factor PHA-4/FoxA, kinesin ZEN-4/MKLP1, its partner CYK-4/MgcRacGAP, and PAR-6. To establish the animal body plan, embryos link the external epidermis to the internal digestive tract. In Caenorhabditis elegans, this linkage is achieved by the arcade cells, which form an epithelial bridge between the foregut and epidermis, but little is known about how development of these three epithelia is coordinated temporally. The arcade cell epithelium is generated after the epidermis and digestive tract epithelia have matured, ensuring that both organs can withstand the mechanical stress of embryo elongation; mistiming of epithelium formation leads to defects in morphogenesis. Using a combination of genetic, bioinformatic, and imaging approaches, we find that temporal regulation of the arcade cell epithelium is mediated by the pioneer transcription factor and master regulator PHA-4/FoxA, followed by the cytoskeletal regulator and kinesin ZEN-4/MKLP1 and the polarity protein PAR-6. We show that PHA-4 directly activates mRNA expression of a broad cohort of epithelial genes, including junctional factor dlg-1. Accumulation of DLG-1 protein is delayed by ZEN-4, acting in concert with its binding partner CYK-4/MgcRacGAP. Our structure–function analysis suggests that nuclear and kinesin functions are dispensable, whereas binding to CYK-4 is essential, for ZEN-4 function in polarity. Finally, PAR-6 is necessary to localize polarity proteins such as DLG-1 within adherens junctions and at the apical surface, thereby generating arcade cell polarity. Our results reveal that the timing of a landmark event during embryonic morphogenesis is mediated by the concerted action of four proteins that delay the formation of an epithelial bridge until the appropriate time. In addition, we find that mammalian FoxA associates with many epithelial genes, suggesting that direct regulation of epithelial identity may be a conserved feature of FoxA factors and a contributor to FoxA function in development and cancer.
Collapse
Affiliation(s)
- Stephen E Von Stetina
- Department of Molecular and Cellular Biology, Harvard University, Cambridge; MA 02138
| | - Jennifer Liang
- Department of Molecular and Cellular Biology, Harvard University, Cambridge; MA 02138
| | - Georgios Marnellos
- Informatics and Scientific Applications, Science Division, Faculty of Arts and Sciences, Harvard University, Cambridge; MA 02138
| | - Susan E Mango
- Department of Molecular and Cellular Biology, Harvard University, Cambridge; MA 02138
| |
Collapse
|
155
|
Ding B, Liang H, Gao M, Li Z, Xu C, Fan S, Chang N. Forkhead Box A2 (FOXA2) Inhibits Invasion and Tumorigenesis in Glioma Cells. Oncol Res 2017; 25:701-708. [PMID: 27983925 PMCID: PMC7841002 DOI: 10.3727/096504016x14772378087005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The forkhead box A2 (FOXA2) is the key transcriptional factor that plays an important role in tumorigenesis. However, until now the expression pattern and role of FOXA2 in glioma have yet to be elucidated. Therefore, the aim of this study was to evaluate the expression of FOXA2 in glioma and investigate its role in glioma cells. Our data showed that FOXA2 was significantly downregulated in human glioma cell lines. Forced expression of FOXA2 suppressed the ability of glioma cells to proliferate, migrate, and invade and influenced the expression level of EMT-associated proteins. In addition, forced expression of FOXA2 attenuated tumor growth of glioma in a nude mouse xenograft model. Mechanistically, we disclosed that forced expression of FOXA2 greatly downregulated the expression of β-catenin, cyclin D1, and c-Myc in glioma cells. Taken together, these results show that FOXA2 may play an important role in proliferation, invasion, and tumorigenesis in glioma cells. Thus, FOXA2 may be a potential therapeutic target for the treatment of glioma.
Collapse
Affiliation(s)
- Bingqian Ding
- *Department of Neurosurgery, Huaihe Hospital of Henan University, Kaifeng, P.R. China
| | - Huimin Liang
- †Department of Neurology, Huaihe Hospital of Henan University, Kaifeng, P.R. China
| | - Ming Gao
- *Department of Neurosurgery, Huaihe Hospital of Henan University, Kaifeng, P.R. China
| | - Zhenjiang Li
- *Department of Neurosurgery, Huaihe Hospital of Henan University, Kaifeng, P.R. China
| | - Chenyang Xu
- *Department of Neurosurgery, Huaihe Hospital of Henan University, Kaifeng, P.R. China
| | - Shaokang Fan
- *Department of Neurosurgery, Huaihe Hospital of Henan University, Kaifeng, P.R. China
| | - Na Chang
- †Department of Neurology, Huaihe Hospital of Henan University, Kaifeng, P.R. China
| |
Collapse
|
156
|
FOXA1 in HPV associated carcinomas: Its expression in carcinomas of the head and neck and of the uterine cervix. Exp Mol Pathol 2017; 102:230-236. [DOI: 10.1016/j.yexmp.2017.02.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 02/11/2017] [Indexed: 11/22/2022]
|
157
|
Dong R, Yang Y, Shen Z, Zheng C, Jin Z, Huang Y, Zhang Z, Zheng S, Chen G. Forkhead box A3 attenuated the progression of fibrosis in a rat model of biliary atresia. Cell Death Dis 2017; 8:e2719. [PMID: 28358366 PMCID: PMC5386589 DOI: 10.1038/cddis.2017.99] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 01/06/2017] [Accepted: 02/14/2017] [Indexed: 12/13/2022]
Abstract
Biliary atresia is a rare, devastating disease of infants where a fibroinflammatory process destroys the bile ducts, leading to fibrosis and biliary cirrhosis, and death if untreated. The cause and pathogenesis remain largely unknown. We tried to investigate factors involved in biliary atresia, especially forkhead box A3 (Foxa3), which might exert a role in the treatment of liver disease. We used RNA sequencing to sequence the whole transcriptomes of livers from six biliary atresia and six choledochal cysts patients. Then, we employed a rat disease model by bile duct ligation (BDL) and adenovirus transduction to address the function of Foxa3 in biliary atresia. We found that tight junction, adherence junction, cell cycle, apoptosis, chemokine singling, VEGF and MAPK signaling pathways were enriched in biliary atresia livers. We showed that Foxa3 expression was notably decreased in liver samples from biliary atresia patients. More importantly, we found that its lower expression predicted a poorer overall survival of biliary atresia patients. Rats that received BDL surgery and Foxa3 expression adenovirus resulted in a significant decrease in the deposition of collagen, and expression of profibrotic cytokines (transforming growth factor-β and connective tissue growth factor) and fibrosis markers (α-smooth muscle actin, collagen I and collagen III), as compared with rats that received BDL surgery and control adenovirus. Our data suggested a protection role for Foxa3 during the progression of liver fibrosis in biliary atresia, and thereby supported increasing Foxa3 as a targeted treatment strategy.
Collapse
Affiliation(s)
- Rui Dong
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai, China
| | - Yifan Yang
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai, China
| | - Zhen Shen
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai, China
| | - Chao Zheng
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai, China
| | - Zhu Jin
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai, China
| | - Yanlei Huang
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai, China
| | - Zhien Zhang
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai, China
| | - Shan Zheng
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai, China
| | - Gong Chen
- Department of Pediatric Surgery, Children's Hospital of Fudan University, and Key Laboratory of Neonatal Disease, Ministry of Health, Shanghai, China
| |
Collapse
|
158
|
Charney RM, Forouzmand E, Cho JS, Cheung J, Paraiso KD, Yasuoka Y, Takahashi S, Taira M, Blitz IL, Xie X, Cho KWY. Foxh1 Occupies cis-Regulatory Modules Prior to Dynamic Transcription Factor Interactions Controlling the Mesendoderm Gene Program. Dev Cell 2017; 40:595-607.e4. [PMID: 28325473 PMCID: PMC5434453 DOI: 10.1016/j.devcel.2017.02.017] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 12/24/2016] [Accepted: 02/16/2017] [Indexed: 12/14/2022]
Abstract
The interplay between transcription factors and chromatin dictates gene regulatory network activity. Germ layer specification is tightly coupled with zygotic gene activation and, in most metazoans, is dependent upon maternal factors. We explore the dynamic genome-wide interactions of Foxh1, a maternal transcription factor that mediates Nodal/TGF-β signaling, with cis-regulatory modules (CRMs) during mesendodermal specification. Foxh1 marks CRMs during cleavage stages and recruits the co-repressor Tle/Groucho in the early blastula. We highlight a population of CRMs that are continuously occupied by Foxh1 and show that they are marked by H3K4me1, Ep300, and Fox/Sox/Smad motifs, suggesting interplay between these factors in gene regulation. We also propose a molecular "hand-off" between maternal Foxh1 and zygotic Foxa at these CRMs to maintain enhancer activation. Our findings suggest that Foxh1 functions at the top of a hierarchy of interactions by marking developmental genes for activation, beginning with the onset of zygotic gene expression.
Collapse
Affiliation(s)
- Rebekah M Charney
- Department of Developmental and Cell Biology, Ayala School of Biological Sciences, University of California, Irvine, CA 92697, USA
| | - Elmira Forouzmand
- Department of Computer Science, Donald Bren School of Information & Computer Sciences, University of California, Irvine, CA 92697, USA
| | - Jin Sun Cho
- Department of Developmental and Cell Biology, Ayala School of Biological Sciences, University of California, Irvine, CA 92697, USA
| | - Jessica Cheung
- Department of Developmental and Cell Biology, Ayala School of Biological Sciences, University of California, Irvine, CA 92697, USA
| | - Kitt D Paraiso
- Department of Developmental and Cell Biology, Ayala School of Biological Sciences, University of California, Irvine, CA 92697, USA
| | - Yuuri Yasuoka
- Marine Genomics Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa 904-0495, Japan
| | - Shuji Takahashi
- Institute for Amphibian Biology, Graduate School of Science, Hiroshima University, 1-3-1 Kagamiyama, Higashihiroshima, Hiroshima 739-8526, Japan
| | - Masanori Taira
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Ira L Blitz
- Department of Developmental and Cell Biology, Ayala School of Biological Sciences, University of California, Irvine, CA 92697, USA
| | - Xiaohui Xie
- Department of Computer Science, Donald Bren School of Information & Computer Sciences, University of California, Irvine, CA 92697, USA
| | - Ken W Y Cho
- Department of Developmental and Cell Biology, Ayala School of Biological Sciences, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
159
|
Charney RM, Paraiso KD, Blitz IL, Cho KWY. A gene regulatory program controlling early Xenopus mesendoderm formation: Network conservation and motifs. Semin Cell Dev Biol 2017; 66:12-24. [PMID: 28341363 DOI: 10.1016/j.semcdb.2017.03.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 03/12/2017] [Accepted: 03/20/2017] [Indexed: 02/08/2023]
Abstract
Germ layer formation is among the earliest differentiation events in metazoan embryos. In triploblasts, three germ layers are formed, among which the endoderm gives rise to the epithelial lining of the gut tube and associated organs including the liver, pancreas and lungs. In frogs (Xenopus), where early germ layer formation has been studied extensively, the process of endoderm specification involves the interplay of dozens of transcription factors. Here, we review the interactions between these factors, summarized in a transcriptional gene regulatory network (GRN). We highlight regulatory connections conserved between frog, fish, mouse, and human endodermal lineages. Especially prominent is the conserved role and regulatory targets of the Nodal signaling pathway and the T-box transcription factors, Vegt and Eomes. Additionally, we highlight network topologies and motifs, and speculate on their possible roles in development.
Collapse
Affiliation(s)
- Rebekah M Charney
- Department of Developmental and Cell Biology, Ayala School of Biological Sciences, University of California, Irvine, CA 92697, USA
| | - Kitt D Paraiso
- Department of Developmental and Cell Biology, Ayala School of Biological Sciences, University of California, Irvine, CA 92697, USA
| | - Ira L Blitz
- Department of Developmental and Cell Biology, Ayala School of Biological Sciences, University of California, Irvine, CA 92697, USA
| | - Ken W Y Cho
- Department of Developmental and Cell Biology, Ayala School of Biological Sciences, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
160
|
Kanaki M, Kardassis D. Regulation of the human lipoprotein lipase gene by the forkhead box transcription factor FOXA2/HNF-3β in hepatic cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2017; 1860:327-336. [DOI: 10.1016/j.bbagrm.2017.01.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 01/13/2017] [Accepted: 01/13/2017] [Indexed: 12/11/2022]
|
161
|
FOXA1 is expressed in ovarian mucinous neoplasms. Pathology 2017; 49:271-276. [PMID: 28238418 DOI: 10.1016/j.pathol.2016.11.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 11/18/2016] [Accepted: 11/24/2016] [Indexed: 01/04/2023]
Abstract
FOXA1 is a transcription factor essential for the binding and action of other transcription factors on the chromatin. It is the major regulator of endoderm differentiation. It has important roles in breast, prostate and endometrial cancer. It has never been studied in ovarian tumours. The aim of this study was to investigate its expression in ovarian epithelial neoplasms. A total of 195 primary ovarian epithelial borderline or malignant tumours were immunohistochemically studied for the expression of FOXA1. Nineteen percent of the tumours strongly and diffusely expressed FOXA1. Of these, 75.7% belong to the mucinous category (p < 0.0001). Seventy-five per cent of mucinous borderline tumours and 46.7% of mucinous carcinomas overexpressed FOXA1. Brenner tumours also expressed FOXA1. FOXA1 was rarely expressed in serous (6/115) and endometrioid tumours (1/11). Clear cell tumours were completely negative (0/16). Of normal structures, ciliated tubal cells, Walthard nests and transitional metaplasias of the tubal-mesothelial junction, all strongly expressed FOXA1. In conclusion, FOXA1 is found in ovarian mucinous and Brenner tumours.
Collapse
|
162
|
Villasenor A, Stainier DYR. On the development of the hepatopancreatic ductal system. Semin Cell Dev Biol 2017; 66:69-80. [PMID: 28214561 DOI: 10.1016/j.semcdb.2017.02.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 02/03/2017] [Accepted: 02/13/2017] [Indexed: 12/13/2022]
Abstract
The hepatopancreatic ductal system is the collection of ducts that connect the liver and pancreas to the digestive tract. The formation of this system is necessary for the transport of exocrine secretions, for the correct assembly of the pancreatobiliary ductal system, and for the overall function of the digestive system. Studies on endoderm organ formation have significantly advanced our understanding of the molecular mechanisms that govern organ induction, organ specification and morphogenesis of the major foregut-derived organs. However, little is known about the mechanisms that control the development of the hepatopancreatic ductal system. Here, we provide a description of the different components of the system, summarize its development from the endoderm to a complex system of tubes, list the pathologies produced by anomalies in its development, as well as the molecules and signaling pathways that are known to be involved in its formation. Finally, we discuss its proposed potential as a multipotent cell reservoir and the unresolved questions in the field.
Collapse
Affiliation(s)
- Alethia Villasenor
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.
| |
Collapse
|
163
|
Li H, Chen L, Zhang M, Zhang B. Foxa1 gene and protein in developing rat eccrine sweat glands. J Mol Histol 2017; 48:1-7. [PMID: 27787633 DOI: 10.1007/s10735-016-9700-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 10/19/2016] [Indexed: 02/05/2023]
Abstract
To investigate the development of eccrine sweat glands and the expression of Foxa1 genes and proteins in the course of development, the footpads from E15.5 to E21.5, P1-P12, P14, P21, P28 and P56 rats were subjected to immunofluorescence staining of FoxA1 and double immunofluorescence staining of K14/α-SMA, FoxA1/K7 and FoxA1/α-SMA, and were processed for Foxa1 gene detection by RT-qPCR. The results showed that rat eccrine sweat gland germs was first observed emerging from the basal layer of epidermis at E19.5, and then elongated downward into the dermis, forming straight ducts by E21.5. Early development of the secretory segments appeared at P1. The Foxa1 gene was not expressed in rat footpads until P2, but from P2 to P5, its expression up-regulated sharply, and thereafter maintained at a high level until adulthood. FoxA1 protein was first observed at P6 in eccrine sweat glands, four days after initial detection of Foxa1 gene transcripts. In skin, FoxA1-positive cells were present exclusively in secretory coils, with 95% being K7-positive secretory cells and 5% being α-SMA-positive myoepithelial cells. We conclude that Foxa1 can be used as a marker of eccrine sweat glands in skin and also as a marker of secretory coils, and Foxa1 is related to the development of secretory coils.
Collapse
Affiliation(s)
- Haihong Li
- Burn and Plastic Surgery, The Second Affiliated Hospital, Shantou University Medical College, North Dongxia Road, Shantou, 515041, Guangdong Province, China.
- Research Center for Translational Medicine, Shantou University Medical College, North Dongxia Road, Shantou, 515041, Guangdong Province, China.
| | - Liyun Chen
- Burn and Plastic Surgery, The Second Affiliated Hospital, Shantou University Medical College, North Dongxia Road, Shantou, 515041, Guangdong Province, China
| | - Mingjun Zhang
- Burn and Plastic Surgery, The Second Affiliated Hospital, Shantou University Medical College, North Dongxia Road, Shantou, 515041, Guangdong Province, China
| | - Bingna Zhang
- Research Center for Translational Medicine, Shantou University Medical College, North Dongxia Road, Shantou, 515041, Guangdong Province, China
| |
Collapse
|
164
|
Casselli T, Qureshi H, Peterson E, Perley D, Blake E, Jokinen B, Abbas A, Nechaev S, Watt JA, Dhasarathy A, Brissette CA. MicroRNA and mRNA Transcriptome Profiling in Primary Human Astrocytes Infected with Borrelia burgdorferi. PLoS One 2017; 12:e0170961. [PMID: 28135303 PMCID: PMC5279786 DOI: 10.1371/journal.pone.0170961] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 01/14/2017] [Indexed: 02/07/2023] Open
Abstract
Lyme disease is caused by infection with the bacterium Borrelia burgdorferi (Bb), which is transmitted to humans by deer ticks. The infection manifests usually as a rash and minor systemic symptoms; however, the bacteria can spread to other tissues, causing joint pain, carditis, and neurological symptoms. Lyme neuroborreliosis presents itself in several ways, such as Bell's palsy, meningitis, and encephalitis. The molecular basis for neuroborreliosis is poorly understood. Analysis of the changes in the expression levels of messenger RNAs and non-coding RNAs, including microRNAs, following Bb infection could therefore provide vital information on the pathogenesis and clinical symptoms of neuroborreliosis. To this end, we used cultured primary human astrocytes, key responders to CNS infection and important components of the blood-brain barrier, as a model system to study RNA and microRNA changes in the CNS caused by Bb. Using whole transcriptome RNA-seq, we found significant changes in 38 microRNAs and 275 mRNAs at 24 and 48 hours following Bb infection. Several of the RNA changes affect pathways involved in immune response, development, chromatin assembly (including histones) and cell adhesion. Further, several of the microRNA predicted target mRNAs were also differentially regulated. Overall, our results indicate that exposure to Bb causes significant changes to the transcriptome and microRNA profile of astrocytes, which has implications in the pathogenesis, and hence potential treatment strategies to combat this disease.
Collapse
Affiliation(s)
- Timothy Casselli
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - Humaira Qureshi
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - Elizabeth Peterson
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - Danielle Perley
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - Emily Blake
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - Bradley Jokinen
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - Ata Abbas
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - Sergei Nechaev
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - John A. Watt
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - Archana Dhasarathy
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - Catherine A. Brissette
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| |
Collapse
|
165
|
|
166
|
Ahmed MS, Kang MH, Lee E, Park Y, Jeong Y, Bae YS. SH2 domain-containing adaptor protein B expressed in dendritic cells is involved in T-cell homeostasis by regulating dendritic cell-mediated Th2 immunity. Clin Exp Vaccine Res 2017; 6:50-60. [PMID: 28168174 PMCID: PMC5292358 DOI: 10.7774/cevr.2017.6.1.50] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 12/21/2016] [Accepted: 01/06/2017] [Indexed: 01/02/2023] Open
Abstract
Purpose The Src homology 2 domain–containing adaptor protein B (SHB) is widely expressed in immune cells and acts as an important regulator for hematopoietic cell function. SHB silencing induces Th2 immunity in mice. SHB is also involved in T-cell homeostasis in vivo. However, SHB has not yet been studied and addressed in association with dendritic cells (DCs). Materials and Methods The effects of SHB expression on the immunogenicity of DCs were assessed by Shb gene silencing in mouse bone marrow–derived DCs (BMDCs). After silencing, surface phenotype, cytokine expression profile, and T-cell stimulation capacity of BMDCs were examined. We investigated the signaling pathways involved in SHB expression during BMDC development. We also examined the immunogenicity of SHB-knockdown (SHBKD) BMDCs in a mouse atopic dermatitis model. Results SHB was steadily expressed in mouse splenic DCs and in in vitro–generated BMDCs in both immature and mature stages. SHB expression was contingent on activation of the mitogen- activated protein kinase/Foxa2 signaling pathway during DC development. SHBKD increased the expression of MHC class II and costimulatory molecules without affecting the cytokine expression of BMDCs. When co-cultured with T cells, SHBKD in BMDCs significantly induced CD4+ T-cell proliferation and the expression of Th2 cytokines, while the regulatory T cell (Treg) population was downregulated. In mouse atopic dermatitis model, mice inoculated with SHBKD DCs developed more severe symptoms of atopic dermatitis compared with mice injected with control DCs. Conclusion SHB expression in DCs plays an important role in T-cell homeostasis in vivo by regulating DC-mediated Th2 polarization.
Collapse
Affiliation(s)
- Md Selim Ahmed
- Department of Biological Science, Sungkyunkwan University, Suwon, Korea
| | - Myeong-Ho Kang
- Department of Biological Science, Sungkyunkwan University, Suwon, Korea
| | - Ezra Lee
- Department of Biological Science, Sungkyunkwan University, Suwon, Korea
| | - Yujin Park
- Department of Biological Science, Sungkyunkwan University, Suwon, Korea
| | - Yideul Jeong
- Department of Biological Science, Sungkyunkwan University, Suwon, Korea
| | - Yong-Soo Bae
- Department of Biological Science, Sungkyunkwan University, Suwon, Korea
| |
Collapse
|
167
|
Zinovyeva MV, Kuzmich AI, Monastyrskaya GS, Sverdlov ED. The role of FOXA subfamily factors in embryonic development and carcinogenesis of the pancreas. MOLECULAR GENETICS MICROBIOLOGY AND VIROLOGY 2017. [DOI: 10.3103/s0891416816030113] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
168
|
Forkhead box a2 (FOXA2) is essential for uterine function and fertility. Proc Natl Acad Sci U S A 2017; 114:E1018-E1026. [PMID: 28049832 DOI: 10.1073/pnas.1618433114] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Establishment of pregnancy is a critical event, and failure of embryo implantation and stromal decidualization in the uterus contribute to significant numbers of pregnancy losses in women. Glands of the uterus are essential for establishment of pregnancy in mice and likely in humans. Forkhead box a2 (FOXA2) is a transcription factor expressed specifically in the glands of the uterus and is a critical regulator of postnatal uterine gland differentiation in mice. In this study, we conditionally deleted FOXA2 in the adult mouse uterus using the lactotransferrin Cre (Ltf-Cre) model and in the neonatal mouse uterus using the progesterone receptor Cre (Pgr-Cre) model. The uteri of adult FOXA2-deleted mice were morphologically normal and contained glands, whereas the uteri of neonatal FOXA2-deleted mice were completely aglandular. Notably, adult FOXA2-deleted mice are completely infertile because of defects in blastocyst implantation and stromal cell decidualization. Leukemia inhibitory factor (LIF), a critical implantation factor of uterine gland origin, was not expressed during early pregnancy in adult FOXA2-deleted mice. Intriguingly, i.p. injections of LIF initiated blastocyst implantation in the uteri of both gland-containing and glandless adult FOXA2-deleted mice. Although pregnancy was rescued by LIF and was maintained to term in uterine gland-containing adult FOXA2-deleted mice, pregnancy failed by day 10 in neonatal FOXA2-deleted mice lacking uterine glands. These studies reveal a previously unrecognized role for FOXA2 in regulation of adult uterine function and fertility and provide original evidence that uterine glands and, by inference, their secretions play important roles in blastocyst implantation and stromal cell decidualization.
Collapse
|
169
|
|
170
|
Qian Y, Xia S, Feng Z. Sox9 mediated transcriptional activation of FOXK2 is critical for colorectal cancer cells proliferation. Biochem Biophys Res Commun 2016; 483:475-481. [PMID: 28007600 DOI: 10.1016/j.bbrc.2016.12.119] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 12/18/2016] [Indexed: 12/11/2022]
Abstract
FOXK2, which belongs to the fork head DNA binding protein family, has been shown to play a critical role in tumorigenesis. Here, we detected FOXK2 expression and its clinical significance in colorectal cancer, which has not been fully investigated before. Results from public database and our cohort indicated that FOXK2 was transcriptionally activated in colorectal cancer tissues compared to non-cancer tissues. High expression of FOXK2 was significantly correlated with poor survival. In vitro cell experiments suggested that FOXK2 promoted cell proliferation. Furthermore, we found that oncogene SOX9 was responsible for the up-regulation of FOXK2 by directly binding on its promoter. Depletion of FOXK2 attenuated SOX9 induced cell growth. In addition, we observed that the expression of FOXK2 was significantly associated with the expression of SOX9 both in the public database and our colorectal cancer tissues. The patients with SOX9+FOXK2+ had a poor overall survival (p = 0.0084). In conclusion, our data suggested that SOX9 transcriptionally activated FOXK2 was involved in the pathogenesis of colorectal cancer and might be a novel target for colorectal cancer therapy.
Collapse
Affiliation(s)
- Yu Qian
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan 030001, Shanxi, China.
| | - Suhua Xia
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Zhenyu Feng
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, Jiangsu, China
| |
Collapse
|
171
|
Abstract
Forkhead box (Fox) transcription factors are evolutionarily conserved in organisms ranging from yeast to humans. They regulate diverse biological processes both during development and throughout adult life. Mutations in many Fox genes are associated with human disease and, as such, various animal models have been generated to study the function of these transcription factors in mechanistic detail. In many cases, the absence of even a single Fox transcription factor is lethal. In this Primer, we provide an overview of the Fox family, highlighting several key Fox transcription factor families that are important for mammalian development.
Collapse
Affiliation(s)
- Maria L Golson
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
172
|
Wruck W, Schröter F, Adjaye J. Meta-Analysis of Transcriptome Data Related to Hippocampus Biopsies and iPSC-Derived Neuronal Cells from Alzheimer's Disease Patients Reveals an Association with FOXA1 and FOXA2 Gene Regulatory Networks. J Alzheimers Dis 2016; 50:1065-82. [PMID: 26890743 DOI: 10.3233/jad-150733] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Although the incidence of Alzheimer's disease (AD) is continuously increasing in the aging population worldwide, effective therapies are not available. The interplay between causative genetic and environmental factors is partially understood. Meta-analyses have been performed on aspects such as polymorphisms, cytokines, and cognitive training. Here, we propose a meta-analysis approach based on hierarchical clustering analysis of a reliable training set of hippocampus biopsies, which is condensed to a gene expression signature. This gene expression signature was applied to various test sets of brain biopsies and iPSC-derived neuronal cell models to demonstrate its ability to distinguish AD samples from control. Thus, our identified AD-gene signature may form the basis for determination of biomarkers that are urgently needed to overcome current diagnostic shortfalls. Intriguingly, the well-described AD-related genes APP and APOE are not within the signature because their gene expression profiles show a lower correlation to the disease phenotype than genes from the signature. This is in line with the differing characteristics of the disease as early-/late-onset or with/without genetic predisposition. To investigate the gene signature's systemic role(s), signaling pathways, gene ontologies, and transcription factors were analyzed which revealed over-representation of response to stress, regulation of cellular metabolic processes, and reactive oxygen species. Additionally, our results clearly point to an important role of FOXA1 and FOXA2 gene regulatory networks in the etiology of AD. This finding is in corroboration with the recently reported major role of the dopaminergic system in the development of AD and its regulation by FOXA1 and FOXA2.
Collapse
|
173
|
Foxa1 is essential for development and functional integrity of the subthalamic nucleus. Sci Rep 2016; 6:38611. [PMID: 27934886 PMCID: PMC5146925 DOI: 10.1038/srep38611] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 11/10/2016] [Indexed: 01/15/2023] Open
Abstract
Inactivation of transcription factor Foxa1 in mice results in neonatal mortality of unknown cause. Here, we report that ablation of Foxa1 causes impaired development and loss of the subthalamic nucleus (STN). Functional deficits in the STN have been implicated in the etiology of Huntington's and Parkinson's disease. We show that neuronal ablation by Synapsin1-Cre-mediated Foxa1 deletion is sufficient to induce hyperlocomotion in mice. Transcriptome profiling of STN neurons in conditional Foxa1 knockout mice revealed changes in gene expression reminiscent of those in neurodegenerative diseases. We identified Ppargc1a, a transcriptional co-activator that is implicated in neurodegeneration, as a Foxa1 target. These findings were substantiated by the observation of Foxa1-dependent demise of STN neurons in conditional models of Foxa1 mutant mice. Finally, we show that the spontaneous firing activity of Foxa1-deficient STN neurons is profoundly impaired. Our data reveal so far elusive roles of Foxa1 in the development and maintenance of STN function.
Collapse
|
174
|
Pavek P. Pregnane X Receptor (PXR)-Mediated Gene Repression and Cross-Talk of PXR with Other Nuclear Receptors via Coactivator Interactions. Front Pharmacol 2016; 7:456. [PMID: 27932985 PMCID: PMC5122737 DOI: 10.3389/fphar.2016.00456] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 11/14/2016] [Indexed: 12/13/2022] Open
Abstract
Pregnane X receptor is a ligand-activated nuclear receptor (NR) that mainly controls inducible expression of xenobiotics handling genes including biotransformation enzymes and drug transporters. Nowadays it is clear that PXR is also involved in regulation of intermediate metabolism through trans-activation and trans-repression of genes controlling glucose, lipid, cholesterol, bile acid, and bilirubin homeostasis. In these processes PXR cross-talks with other NRs. Accumulating evidence suggests that the cross-talk is often mediated by competing for common coactivators or by disruption of coactivation and activity of other transcription factors by the ligand-activated PXR. In this respect mainly PXR-CAR and PXR-HNF4α interference have been reported and several cytochrome P450 enzymes (such as CYP7A1 and CYP8B1), phase II enzymes (SULT1E1, Gsta2, Ugt1a1), drug and endobiotic transporters (OCT1, Mrp2, Mrp3, Oatp1a, and Oatp4) as well as intermediate metabolism enzymes (PEPCK1 and G6Pase) have been shown as down-regulated genes after PXR activation. In this review, I summarize our current knowledge of PXR-mediated repression and coactivation interference in PXR-controlled gene expression regulation.
Collapse
Affiliation(s)
- Petr Pavek
- Department of Pharmacology and Toxicology and Centre for Drug Development, Faculty of Pharmacy in Hradec Kralove, Charles University in Prague Hradec Kralove, Czechia
| |
Collapse
|
175
|
Dhasarathy A, Roemmich JN, Claycombe KJ. Influence of maternal obesity, diet and exercise on epigenetic regulation of adipocytes. Mol Aspects Med 2016; 54:37-49. [PMID: 27825817 DOI: 10.1016/j.mam.2016.10.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 10/25/2016] [Accepted: 10/25/2016] [Indexed: 12/11/2022]
Affiliation(s)
- Archana Dhasarathy
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - James N Roemmich
- USDA-ARS-PA, Grand Forks Human Nutrition Research Center, 2420 2nd Avenue North, Grand Forks, ND 58203, USA
| | - Kate J Claycombe
- USDA-ARS-PA, Grand Forks Human Nutrition Research Center, 2420 2nd Avenue North, Grand Forks, ND 58203, USA.
| |
Collapse
|
176
|
Chen Q, Yang X, Zhang H, Kong X, Yao L, Cui X, Zou Y, Fang F, Yang J, Chang Y. Metformin impairs systemic bile acid homeostasis through regulating SIRT1 protein levels. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1864:101-112. [PMID: 27816442 DOI: 10.1016/j.bbamcr.2016.10.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 10/24/2016] [Accepted: 10/30/2016] [Indexed: 01/04/2023]
Abstract
Metformin is widely used to treat hyperglycemia. However, metformin treatment may induce intrahepatic cholestasis and liver injury in a few patients with type II diabetes through an unknown mechanism. Here we show that metformin decreases SIRT1 protein levels in primary hepatocytes and liver. Both metformin-treated wild-type C57 mice and hepatic SIRT1-mutant mice had increased hepatic and serum bile acid levels. However, metformin failed to change systemic bile acid levels in hepatic SIRT1-mutant mice. Molecular mechanism study indicates that SIRT1 directly interacts with and deacetylates Foxa2 to inhibit its transcriptional activity on expression of genes involved in bile acids synthesis and transport. Hepatic SIRT1 mutation elevates Foxa2 acetylation levels, which promotes Foxa2 binding to and activating genes involved in bile acids metabolism, impairing hepatic and systemic bile acid homeostasis. Our data clearly suggest that hepatic SIRT1 mediates metformin effects on systemic bile acid metabolism and modulation of SIRT1 activity in liver may be an attractive approach for treatment of bile acid-related diseases such as cholestasis.
Collapse
Affiliation(s)
- Qi Chen
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China; Hangzhou Center for Disease Control and Prevention, Zhejiang, People's Republic of China
| | - Xiaoying Yang
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Huabing Zhang
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Xingxing Kong
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Lu Yao
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Xiaona Cui
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Yongkang Zou
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Fude Fang
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Jichun Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, People's Republic of China
| | - Yongsheng Chang
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China.
| |
Collapse
|
177
|
Guaraldo M, Santambrogio P, Rovelli E, Di Savino A, Saglio G, Cittaro D, Roetto A, Levi S. Characterization of human mitochondrial ferritin promoter: identification of transcription factors and evidences of epigenetic control. Sci Rep 2016; 6:33432. [PMID: 27625068 PMCID: PMC5022048 DOI: 10.1038/srep33432] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 08/02/2016] [Indexed: 02/07/2023] Open
Abstract
Mitochondrial ferritin (FtMt) is an iron storage protein belonging to the ferritin family but, unlike the cytosolic ferritin, it has an iron-unrelated restricted tissue expression. FtMt appears to be preferentially expressed in cell types characterized by high metabolic activity and oxygen consumption, suggesting a role in protecting mitochondria from iron-dependent oxidative damage. The human gene (FTMT) is intronless and its promoter region has not been described yet. To analyze the regulatory mechanisms controlling FTMT expression, we characterized the 5' flanking region upstream the transcriptional starting site of FTMT by in silico enquiry of sequences conservation, DNA deletion analysis, and ChIP assay. The data revealed a minimal promoter region and identified the presence of SP1, CREB and YY1 as positive regulators, and GATA2, FoxA1 and C/EBPβ as inhibitors of the transcriptional regulation. Furthermore, the FTMT transcription is increased by acetylating and de-methylating agent treatments in K562 and HeLa cells. These treatments up-regulate FtMt expression even in fibroblasts derived from a Friedreich ataxia patient, where it might exert a beneficial effect against mitochondrial oxidative damage. The expression of FTMT appears regulated by a complex mechanism involving epigenetic events and interplay between transcription factors.
Collapse
Affiliation(s)
- Michela Guaraldo
- San Raffaele Scientific Institute, Division of Neuroscience, 20132 Milano, Italy
| | - Paolo Santambrogio
- San Raffaele Scientific Institute, Division of Neuroscience, 20132 Milano, Italy
| | - Elisabetta Rovelli
- San Raffaele Scientific Institute, Division of Neuroscience, 20132 Milano, Italy
| | - Augusta Di Savino
- University of Torino, Department of Clinical and Biological Sciences, AOU San Luigi Gonzaga, 10043 Orbassano, Torino, Italy
| | - Giuseppe Saglio
- University of Torino, Department of Clinical and Biological Sciences, AOU San Luigi Gonzaga, 10043 Orbassano, Torino, Italy
| | - Davide Cittaro
- San Raffaele Scientific Institute, Center for Translational Genomics and Bioinformatics, 20132 Milano, Italy
| | - Antonella Roetto
- University of Torino, Department of Clinical and Biological Sciences, AOU San Luigi Gonzaga, 10043 Orbassano, Torino, Italy
| | - Sonia Levi
- San Raffaele Scientific Institute, Division of Neuroscience, 20132 Milano, Italy
- University Vita-Salute San Raffaele, 20132 Milano, Italy
| |
Collapse
|
178
|
Morris SA. Direct lineage reprogramming via pioneer factors; a detour through developmental gene regulatory networks. Development 2016; 143:2696-705. [PMID: 27486230 PMCID: PMC5004913 DOI: 10.1242/dev.138263] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Although many approaches have been employed to generate defined fate in vitro, the resultant cells often appear developmentally immature or incompletely specified, limiting their utility. Growing evidence suggests that current methods of direct lineage conversion may rely on the transition through a developmental intermediate. Here, I hypothesize that complete conversion between cell fates is more probable and feasible via reversion to a developmentally immature state. I posit that this is due to the role of pioneer transcription factors in engaging silent, unmarked chromatin and activating hierarchical gene regulatory networks responsible for embryonic patterning. Understanding these developmental contexts will be essential for the precise engineering of cell identity.
Collapse
Affiliation(s)
- Samantha A Morris
- Departments of Developmental Biology and Genetics, Center of Regenerative Medicine, Washington University School of Medicine in St. Louis, 660 S. Euclid Avenue, Campus Box 8103, St. Louis, MO 63110, USA
| |
Collapse
|
179
|
León S, Fernandois D, Sull A, Sull J, Calder M, Hayashi K, Bhattacharya M, Power S, Vilos GA, Vilos AG, Tena-Sempere M, Babwah AV. Beyond the brain-Peripheral kisspeptin signaling is essential for promoting endometrial gland development and function. Sci Rep 2016; 6:29073. [PMID: 27364226 PMCID: PMC4929565 DOI: 10.1038/srep29073] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/10/2016] [Indexed: 12/18/2022] Open
Abstract
Uterine growth and endometrial gland formation (adenogenesis) and function, are essential for fertility and are controlled by estrogens and other regulators, whose nature and physiological relevance are yet to be elucidated. Kisspeptin, which signals via Kiss1r, is essential for fertility, primarily through its central control of the hypothalamic-pituitary-ovarian axis, but also likely through peripheral actions. Using genetically modified mice, we addressed the contributions of central and peripheral kisspeptin signaling in regulating uterine growth and adenogenesis. Global ablation of Kiss1 or Kiss1r dramatically suppressed uterine growth and almost fully prevented adenogenesis. However, while uterine growth was fully rescued by E2 treatment of Kiss1−/− mice and by genetic restoration of kisspeptin signaling in GnRH neurons in Kiss1r−/− mice, functional adenogenesis was only marginally restored. Thus, while uterine growth is largely dependent on ovarian E2-output via central kisspeptin signaling, peripheral kisspeptin signaling is indispensable for endometrial adenogenesis and function, essential aspects of reproductive competence.
Collapse
Affiliation(s)
- Silvia León
- Department of Cell Biology, Physiology &Immunology, Faculty of Medicine and Instituto Maimonides de Investigacion Biomedica de Córdoba (IMIBIC)/Hospital Reina Sofia, University of Córdoba, Avda. Menéndez Pidal s/n, Spain
| | - Daniela Fernandois
- Department of Cell Biology, Physiology &Immunology, Faculty of Medicine and Instituto Maimonides de Investigacion Biomedica de Córdoba (IMIBIC)/Hospital Reina Sofia, University of Córdoba, Avda. Menéndez Pidal s/n, Spain
| | - Alexandra Sull
- The Children's Health Research Institute, London, Ontario, Canada.,Lawson Health Research Institute, London, Ontario, Canada
| | - Judith Sull
- The Children's Health Research Institute, London, Ontario, Canada.,Lawson Health Research Institute, London, Ontario, Canada
| | - Michele Calder
- The Children's Health Research Institute, London, Ontario, Canada.,Lawson Health Research Institute, London, Ontario, Canada.,Department of Obstetrics and Gynaecology, Division of Reproductive Endocrinology and Infertility, London, Ontario, N6C 2V5, Canada
| | - Kanako Hayashi
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, IL 62901, USA
| | - Moshmi Bhattacharya
- Lawson Health Research Institute, London, Ontario, Canada.,Department of Physiology and Pharmacology, London, Ontario, N6C 2V5, Canada.,Department of Oncology, London, Ontario University of Western Ontario, London, Ontario, N6C 2V5, Canada
| | - Stephen Power
- Department of Obstetrics and Gynaecology, Division of Reproductive Endocrinology and Infertility, London, Ontario, N6C 2V5, Canada
| | - George A Vilos
- Department of Obstetrics and Gynaecology, Division of Reproductive Endocrinology and Infertility, London, Ontario, N6C 2V5, Canada
| | - Angelos G Vilos
- Department of Obstetrics and Gynaecology, Division of Reproductive Endocrinology and Infertility, London, Ontario, N6C 2V5, Canada
| | - Manuel Tena-Sempere
- Department of Cell Biology, Physiology &Immunology, Faculty of Medicine and Instituto Maimonides de Investigacion Biomedica de Córdoba (IMIBIC)/Hospital Reina Sofia, University of Córdoba, Avda. Menéndez Pidal s/n, Spain.,CIBEROBN, Instituto de Salud Carlos III, 14004 Córdoba, Spain.,FiDiPro Program, Department of Physiology, University of Turku, 20520 Turku, Finland
| | - Andy V Babwah
- The Children's Health Research Institute, London, Ontario, Canada.,Lawson Health Research Institute, London, Ontario, Canada.,Department of Obstetrics and Gynaecology, Division of Reproductive Endocrinology and Infertility, London, Ontario, N6C 2V5, Canada.,Department of Physiology and Pharmacology, London, Ontario, N6C 2V5, Canada
| |
Collapse
|
180
|
Vorvis C, Hatziapostolou M, Mahurkar-Joshi S, Koutsioumpa M, Williams J, Donahue TR, Poultsides GA, Eibl G, Iliopoulos D. Transcriptomic and CRISPR/Cas9 technologies reveal FOXA2 as a tumor suppressor gene in pancreatic cancer. Am J Physiol Gastrointest Liver Physiol 2016; 310:G1124-37. [PMID: 27151939 PMCID: PMC5005285 DOI: 10.1152/ajpgi.00035.2016] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 04/27/2016] [Indexed: 01/31/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer with low survival rates and limited therapeutic options. Thus elucidation of signaling pathways involved in PDAC pathogenesis is essential for identifying novel potential therapeutic gene targets. Here, we used a systems approach to elucidate those pathways by integrating gene and microRNA profiling analyses together with CRISPR/Cas9 technology to identify novel transcription factors involved in PDAC pathogenesis. FOXA2 transcription factor was found to be significantly downregulated in PDAC relative to control pancreatic tissues. Functional experiments revealed that FOXA2 has a tumor suppressor function through inhibition of pancreatic cancer cell growth, migration, invasion, and colony formation. In situ hybridization analysis revealed miR-199a to be significantly upregulated in pancreatic cancer. Bioinformatics and luciferase analyses showed that miR-199a negatively but directly regulates FOXA2 expression through binding in its 3'-untranslated region (UTR). Evaluation of the functional importance of miR-199a on pancreatic cancer revealed that miR-199a acts as an inhibitor of FOXA2 expression, inducing an increase in pancreatic cancer cell proliferation, migration, and invasion. Additionally, gene ontology and network analyses in PANC-1 cells treated with a small interfering RNA (siRNA) against FOXA2 revealed an enrichment for cell invasion mechanisms through PLAUR and ERK activation. FOXA2 deletion (FOXA2Δ) by using two CRISPR/Cas9 vectors in PANC-1 cells induced tumor growth in vivo resulting in upregulation of PLAUR and ERK pathways in FOXA2Δ xenograft tumors. We have identified FOXA2 as a novel tumor suppressor in pancreatic cancer and it is regulated directly by miR-199a, thereby enhancing our understanding of how microRNAs interplay with the transcription factors to affect pancreatic oncogenesis.
Collapse
Affiliation(s)
- Christina Vorvis
- 1Center for Systems Biomedicine, Division of Digestive Diseases, David Geffen School of Medicine, University of California, Los Angeles, California;
| | - Maria Hatziapostolou
- 2Centre for Biological Sciences, University of Southampton, Southampton, United Kingdom;
| | - Swapna Mahurkar-Joshi
- 1Center for Systems Biomedicine, Division of Digestive Diseases, David Geffen School of Medicine, University of California, Los Angeles, California;
| | - Marina Koutsioumpa
- 1Center for Systems Biomedicine, Division of Digestive Diseases, David Geffen School of Medicine, University of California, Los Angeles, California;
| | - Jennifer Williams
- 3Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, California; and
| | - Timothy R. Donahue
- 3Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, California; and
| | - George A. Poultsides
- 4Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Guido Eibl
- 3Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, California; and
| | - Dimitrios Iliopoulos
- Center for Systems Biomedicine, Division of Digestive Diseases, David Geffen School of Medicine, University of California, Los Angeles, California;
| |
Collapse
|
181
|
Ricci L, Cabrera F, Lotito S, Tiozzo S. Redeployment of germ layers related TFs shows regionalized expression during two non-embryonic developments. Dev Biol 2016; 416:235-248. [PMID: 27208394 DOI: 10.1016/j.ydbio.2016.05.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 05/11/2016] [Accepted: 05/12/2016] [Indexed: 11/26/2022]
Abstract
In all non-vertebrate metazoan phyla, species that evolved non-embryonic developmental pathways as means of propagation or regeneration can be found. In this context, new bodies arise through asexual reproduction processes (such as budding) or whole body regeneration, that lack the familiar temporal and spatial cues classically associated with embryogenesis, like maternal determinants, or gastrulation. The molecular mechanisms underlying those non-embryonic developments (i.e., regeneration and asexual reproduction), and their relationship to those deployed during embryogenesis are poorly understood. We have addressed this question in the colonial ascidian Botryllus schlosseri, which undergoes an asexual reproductive process via palleal budding (PB), as well as a whole body regeneration by vascular budding (VB). We identified early regenerative structures during VB and then followed the fate of differentiating tissues during both non-embryonic developments (PB and VB) by monitoring the expression of genes known to play key functions in germ layer specification with well conserved expression patterns in solitary ascidian embryogenesis. The expression patterns of FoxA1, GATAa, GATAb, Otx, Bra, Gsc and Tbx2/3 were analysed during both PB and VB. We found that the majority of these transcription factors were expressed during both non-embryonic developmental processes, revealing a regionalization of the palleal and vascular buds. Knockdown of GATAa by siRNA in palleal buds confirmed that preventing the correct development of one of these regions blocks further tissue specification. Our results indicate that during both normal and injury-induced budding, a similar alternative developmental program operates via early commitment of epithelial regions.
Collapse
Affiliation(s)
- Lorenzo Ricci
- Sorbonne Universités, UPMC Univ. Paris 06, CNRS, Laboratoire de Biologie du Développement de Villefranche-sur-mer (LBDV), 06230 Villefranche sur-mer, France
| | - Fabien Cabrera
- Sorbonne Universités, UPMC Univ. Paris 06, CNRS, Laboratoire de Biologie du Développement de Villefranche-sur-mer (LBDV), 06230 Villefranche sur-mer, France
| | - Sonia Lotito
- Sorbonne Universités, UPMC Univ. Paris 06, CNRS, Laboratoire de Biologie du Développement de Villefranche-sur-mer (LBDV), 06230 Villefranche sur-mer, France
| | - Stefano Tiozzo
- Sorbonne Universités, UPMC Univ. Paris 06, CNRS, Laboratoire de Biologie du Développement de Villefranche-sur-mer (LBDV), 06230 Villefranche sur-mer, France.
| |
Collapse
|
182
|
Liu ZY, Yu Q, Yang CH, Meng M, Ren CJ, Mu ZM, Cui WZ, Liu QX. Transcription factor SGF1 is critical for the neurodevelopment in the silkworm, Bombyx mori. Gene 2016; 587:70-5. [PMID: 27106119 DOI: 10.1016/j.gene.2016.04.034] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 04/06/2016] [Accepted: 04/18/2016] [Indexed: 10/21/2022]
Abstract
FoxA transcription factors play vital roles in regulating the expression of organ-specific genes. BmSGF1, the sole FoxA family member in Bombyx mori, is required for development of the silk gland. However, the function of BmSGF1 in development of the nervous system in the silkworm remains unknown. Here, we show that the amino acids sequence of BmSGF1 is evolutionarily conserved in its middle region from Trichoplax adhaerens to human and diverged from the homologues in most other species in its N-terminal region. BmSGF1 expresses in the nervous system at the embryonic stage. Knockdown of Bmsgf1 by RNA interference (RNAi) results in abnormal development of axons. Therefore, our results demonstrate that BmSGF1 is an indispensable regulator for neurodevelopment.
Collapse
Affiliation(s)
- Zhao-Yang Liu
- Laboratory of Developmental Genetics, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Qi Yu
- Laboratory of Developmental Genetics, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Chun-Hong Yang
- Laboratory of Developmental Genetics, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Miao Meng
- Laboratory of Developmental Genetics, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Chun-Jiu Ren
- Laboratory of Developmental Genetics, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Zhi-Mei Mu
- Laboratory of Developmental Genetics, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Wei-Zheng Cui
- Laboratory of Developmental Genetics, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Qing-Xin Liu
- Laboratory of Developmental Genetics, Shandong Agricultural University, Tai'an, Shandong 271018, China.
| |
Collapse
|
183
|
Li M, Zhang W, Liu C, Shi Y, Tang W, Chen S, Gu H, Yin J, Zhang Z, Jiang P. Forkhead box A1 (FOXA1) tagging polymorphisms and esophageal cancer risk in a Chinese population: a fine-mapping study. Biomarkers 2016; 21:523-9. [DOI: 10.3109/1354750x.2016.1160425] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
184
|
Slebe F, Rojo F, Vinaixa M, García-Rocha M, Testoni G, Guiu M, Planet E, Samino S, Arenas EJ, Beltran A, Rovira A, Lluch A, Salvatella X, Yanes O, Albanell J, Guinovart JJ, Gomis RR. FoxA and LIPG endothelial lipase control the uptake of extracellular lipids for breast cancer growth. Nat Commun 2016; 7:11199. [PMID: 27045898 PMCID: PMC4822041 DOI: 10.1038/ncomms11199] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 03/01/2016] [Indexed: 12/26/2022] Open
Abstract
The mechanisms that allow breast cancer (BCa) cells to metabolically sustain rapid growth are poorly understood. Here we report that BCa cells are dependent on a mechanism to supply precursors for intracellular lipid production derived from extracellular sources and that the endothelial lipase (LIPG) fulfils this function. LIPG expression allows the import of lipid precursors, thereby contributing to BCa proliferation. LIPG stands out as an essential component of the lipid metabolic adaptations that BCa cells, and not normal tissue, must undergo to support high proliferation rates. LIPG is ubiquitously and highly expressed under the control of FoxA1 or FoxA2 in all BCa subtypes. The downregulation of either LIPG or FoxA in transformed cells results in decreased proliferation and impaired synthesis of intracellular lipids. Deregulation of lipid metabolism in cancer cells is critical to the maintenance of certain malignant features. Here, the authors show that the proliferation of breast cancer cells depends upon the extracellular activity of the endothelial lipase enzyme LIPG whose expression is regulated by the FoxA family of transcription factors.
Collapse
Affiliation(s)
- Felipe Slebe
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | - Federico Rojo
- Cancer Research Programme, IMIM (Hospital del Mar Medical Research Institute), Barcelona 08003 Spain.,Pathology Department, IIS-Fundación Jimenez Diaz, Madrid 28040, Spain
| | - Maria Vinaixa
- Centre for Omic Sciences, Universitat Rovira i Virgili, Reus 43204, Spain.,Department of Electronic Engineering, Universitat Rovira i Virgili, Tarragona 43003, Spain.,Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid 28029, Spain
| | - Mar García-Rocha
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | - Giorgia Testoni
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | - Marc Guiu
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | - Evarist Planet
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | - Sara Samino
- Centre for Omic Sciences, Universitat Rovira i Virgili, Reus 43204, Spain.,Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid 28029, Spain
| | - Enrique J Arenas
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | - Antoni Beltran
- Centre for Omic Sciences, Universitat Rovira i Virgili, Reus 43204, Spain.,Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid 28029, Spain
| | - Ana Rovira
- Cancer Research Programme, IMIM (Hospital del Mar Medical Research Institute), Barcelona 08003 Spain.,Medical Oncology Service, Hospital del Mar, Barcelona 08003, Spain
| | - Ana Lluch
- Medical Oncology Service, Hospital Clinico, Valencia 46010, Spain
| | - Xavier Salvatella
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona 08010, Spain
| | - Oscar Yanes
- Centre for Omic Sciences, Universitat Rovira i Virgili, Reus 43204, Spain.,Department of Electronic Engineering, Universitat Rovira i Virgili, Tarragona 43003, Spain.,Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid 28029, Spain
| | - Joan Albanell
- Cancer Research Programme, IMIM (Hospital del Mar Medical Research Institute), Barcelona 08003 Spain.,Medical Oncology Service, Hospital del Mar, Barcelona 08003, Spain.,Universitat Pompeu Fabra, Barcelona 08003, Spain
| | - Joan J Guinovart
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain.,Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid 28029, Spain.,Department of Biochemistry and Molecular Biology, Universitat de Barcelona, Barcelona 08028, Spain
| | - Roger R Gomis
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona 08010, Spain
| |
Collapse
|
185
|
Balmer S, Nowotschin S, Hadjantonakis AK. Notochord morphogenesis in mice: Current understanding & open questions. Dev Dyn 2016; 245:547-57. [PMID: 26845388 DOI: 10.1002/dvdy.24392] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 01/26/2016] [Accepted: 01/26/2016] [Indexed: 12/25/2022] Open
Abstract
The notochord is a structure common to all chordates, and the feature that the phylum Chordata has been named after. It is a rod-like mesodermal structure that runs the anterior-posterior length of the embryo, adjacent to the ventral neural tube. The notochord plays a critical role in embryonic tissue patterning, for example the dorsal-ventral patterning of the neural tube. The cells that will come to form the notochord are specified at gastrulation. Axial mesodermal cells arising at the anterior primitive streak migrate anteriorly as the precursors of the notochord and populate the notochordal plate. Yet, even though a lot of interest has centered on investigating the functional and structural roles of the notochord, we still have a very rudimentary understanding of notochord morphogenesis. The events driving the formation of the notochord are rapid, taking place over the period of approximately a day in mice. In this commentary, we provide an overview of our current understanding of mouse notochord morphogenesis, from the initial specification of axial mesendodermal cells at the primitive streak, the emergence of these cells at the midline on the surface of the embryo, to their submergence and organization of the stereotypically positioned notochord. We will also discuss some key open questions. Developmental Dynamics 245:547-557, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Sophie Balmer
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sonja Nowotschin
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Anna-Katerina Hadjantonakis
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
186
|
Forkhead box A3 mediates glucocorticoid receptor function in adipose tissue. Proc Natl Acad Sci U S A 2016; 113:3377-82. [PMID: 26957608 DOI: 10.1073/pnas.1601281113] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Glucocorticoids (GCs) are widely prescribed anti-inflammatory agents, but their chronic use leads to undesirable side effects such as excessive expansion of adipose tissue. We have recently shown that the forkhead box protein A3 (Foxa3) is a calorie-hoarding factor that regulates the selective enlargement of epididymal fat depots and suppresses energy expenditure in a nutritional- and age-dependent manner. It has been demonstrated that Foxa3 levels are elevated in adipose depots in response to high-fat diet regimens and during the aging process; however no studies to date have elucidated the mechanisms that control Foxa3's expression in fat. Given the established effects of GCs in increasing visceral adiposity and in reducing thermogenesis, we assessed the existence of a possible link between GCs and Foxa3. Computational prediction analysis combined with molecular studies revealed that Foxa3 is regulated by the glucocorticoid receptor (GR) in preadipocytes, adipocytes, and adipose tissues and is required to facilitate the binding of the GR to its target gene promoters in fat depots. Analysis of the long-term effects of dexamethasone treatment in mice revealed that Foxa3 ablation protects mice specifically against fat accretion but not against other pathological side effects elicited by this synthetic GC in tissues such as liver, muscle, and spleen. In conclusion our studies provide the first demonstration, to our knowledge, that Foxa3 is a direct target of GC action in adipose tissues and point to a role of Foxa3 as a mediator of the side effects induced in fat tissues by chronic treatment with synthetic steroids.
Collapse
|
187
|
Yalley A, Schill D, Hatta M, Johnson N, Cirillo LA. Loss of Interdependent Binding by the FoxO1 and FoxA1/A2 Forkhead Transcription Factors Culminates in Perturbation of Active Chromatin Marks and Binding of Transcriptional Regulators at Insulin-sensitive Genes. J Biol Chem 2016; 291:8848-61. [PMID: 26929406 DOI: 10.1074/jbc.m115.677583] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Indexed: 01/04/2023] Open
Abstract
FoxO1 binds to insulin response elements located in the promoters of insulin-like growth factor-binding protein 1 (IGFBP1) and glucose-6-phosphatase (G6Pase), activating their expression. Insulin-mediated phosphorylation of FoxO1 promotes cytoplasmic translocation, inhibiting FoxO1-mediated transactivation. We have previously demonstrated that FoxO1 opens and remodels chromatin assembled from the IGFBP1 promoter via a highly conserved winged helix motif. This finding, which established FoxO1 as a "pioneer" factor, suggested a model whereby FoxO1 chromatin remodeling at regulatory targets facilitates binding and recruitment of additional regulatory factors. However, the impact of FoxO1 phosphorylation on its ability to bind chromatin and the effect of FoxO1 loss on recruitment of neighboring transcription factors at its regulatory targets in liver chromatin is unknown. In this study, we demonstrate that an amino acid substitution that mimics insulin-mediated phosphorylation of a serine in the winged helix DNA binding motif curtails FoxO1 nucleosome binding. We also demonstrate that shRNA-mediated loss of FoxO1 binding to the IGFBP1 and G6Pase promoters in HepG2 cells significantly reduces binding of RNA polymerase II and the pioneer factors FoxA1/A2. Knockdown of FoxA1 similarly reduced binding of RNA polymerase II and FoxO1. Reduction in acetylation of histone H3 Lys-27 accompanies loss of FoxO1 and FoxA1/A2 binding. Interdependent binding of FoxO1 and FoxA1/A2 possibly entails cooperative binding because FoxO1 and FoxA1/A2 facilitate one another's binding to IGFPB1 promoter DNA. These results illustrate how transcription factors can nucleate transcriptional events in chromatin in response to signaling events and suggest a model for regulation of hepatic glucose metabolism through interdependent FoxO/FoxA binding.
Collapse
Affiliation(s)
- Akua Yalley
- From the Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Daniel Schill
- From the Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Mitsutoki Hatta
- From the Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Nicole Johnson
- From the Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Lisa Ann Cirillo
- From the Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| |
Collapse
|
188
|
Holmfeldt P, Ganuza M, Marathe H, He B, Hall T, Kang G, Moen J, Pardieck J, Saulsberry AC, Cico A, Gaut L, McGoldrick D, Finkelstein D, Tan K, McKinney-Freeman S. Functional screen identifies regulators of murine hematopoietic stem cell repopulation. J Exp Med 2016; 213:433-49. [PMID: 26880577 PMCID: PMC4813668 DOI: 10.1084/jem.20150806] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 01/07/2016] [Indexed: 11/12/2022] Open
Abstract
Holmfeldt et al. perform a transplant-based screen to identify regulators of HSPC engraftment and report that Foxa3 is critical for optimal HSC function after transplant. Understanding the molecular regulation of hematopoietic stem and progenitor cell (HSPC) engraftment is paramount to improving transplant outcomes. To discover novel regulators of HSPC repopulation, we transplanted >1,300 mice with shRNA-transduced HSPCs within 24 h of isolation and transduction to focus on detecting genes regulating repopulation. We identified 17 regulators of HSPC repopulation: Arhgef5, Armcx1, Cadps2, Crispld1, Emcn, Foxa3, Fstl1, Glis2, Gprasp2, Gpr56, Myct1, Nbea, P2ry14, Smarca2, Sox4, Stat4, and Zfp521. Knockdown of each of these genes yielded a loss of function, except in the cases of Armcx1 and Gprasp2, whose loss enhanced hematopoietic stem cell (HSC) repopulation. The discovery of multiple genes regulating vesicular trafficking, cell surface receptor turnover, and secretion of extracellular matrix components suggests active cross talk between HSCs and the niche and that HSCs may actively condition the niche to promote engraftment. We validated that Foxa3 is required for HSC repopulating activity, as Foxa3−/− HSC fails to repopulate ablated hosts efficiently, implicating for the first time Foxa genes as regulators of HSPCs. We further show that Foxa3 likely regulates the HSC response to hematologic stress. Each gene discovered here offers a window into the novel processes that regulate stable HSPC engraftment into an ablated host.
Collapse
Affiliation(s)
- Per Holmfeldt
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Miguel Ganuza
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Himangi Marathe
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Bing He
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA 52242
| | - Trent Hall
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Guolian Kang
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Joseph Moen
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Jennifer Pardieck
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | | | - Alba Cico
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Ludovic Gaut
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Daniel McGoldrick
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - David Finkelstein
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Kai Tan
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242
| | | |
Collapse
|
189
|
von Wnuck Lipinski K, Sattler K, Peters S, Weske S, Keul P, Klump H, Heusch G, Göthert JR, Levkau B. Hepatocyte Nuclear Factor 1A Is a Cell-Intrinsic Transcription Factor Required for B Cell Differentiation and Development in Mice. THE JOURNAL OF IMMUNOLOGY 2016; 196:1655-65. [PMID: 26800876 DOI: 10.4049/jimmunol.1500897] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 12/07/2015] [Indexed: 12/22/2022]
Abstract
The hepatocyte NF (HNF) family of transcription factors regulates the complex gene networks involved in lipid, carbohydrate, and protein metabolism. In humans, HNF1A mutations cause maturity onset of diabetes in the young type 3, whereas murine HNF6 participates in fetal liver B lymphopoiesis. In this study, we have identified a crucial role for the prototypical member of the family HNF1A in adult bone marrow B lymphopoiesis. HNF1A(-/-) mice exhibited a clear reduction in total blood and splenic B cells and a further pronounced one in transitional B cells. In HNF1A(-/-) bone marrow, all B cell progenitors-from pre-pro-/early pro-B cells to immature B cells-were dramatically reduced and their proliferation rate suppressed. IL-7 administration in vivo failed to boost B cell development in HNF1A(-/-) mice, whereas IL-7 stimulation of HNF1A(-/-) B cell progenitors in vitro revealed a marked impairment in STAT5 phosphorylation. The B cell differentiation potential of HNF1A(-/-) common lymphoid progenitors was severely impaired in vitro, and the expression of the B lymphopoiesis-promoting transcription factors E2A, EBF1, Pax5, and Bach2 was reduced in B cell progenitors in vivo. HNF1A(-/-) bone marrow chimera featured a dramatic defect in B lymphopoiesis recapitulating that of global HNF1A deficiency. The HNF1A(-/-) lymphopoiesis defect was confined to B cells as T lymphopoiesis was unaffected, and bone marrow common lymphoid progenitors and hematopoietic stem cells were even increased. Our data demonstrate that HNF1A is an important cell-intrinsic transcription factor in adult B lymphopoiesis and suggest the IL-7R/STAT5 module to be causally involved in mediating its function.
Collapse
Affiliation(s)
- Karin von Wnuck Lipinski
- Institute for Pathophysiology, West German Heart and Vascular Center, University Hospital Essen, 45122 Essen, Germany
| | - Katherine Sattler
- Institute for Pathophysiology, West German Heart and Vascular Center, University Hospital Essen, 45122 Essen, Germany
| | - Susann Peters
- Institute for Pathophysiology, West German Heart and Vascular Center, University Hospital Essen, 45122 Essen, Germany
| | - Sarah Weske
- Institute for Pathophysiology, West German Heart and Vascular Center, University Hospital Essen, 45122 Essen, Germany
| | - Petra Keul
- Institute for Pathophysiology, West German Heart and Vascular Center, University Hospital Essen, 45122 Essen, Germany
| | - Hannes Klump
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, 45122 Essen, Germany; and
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University Hospital Essen, 45122 Essen, Germany
| | - Joachim R Göthert
- Department of Hematology, University Hospital Essen, University of Duisburg-Essen, 45122 Essen, Germany
| | - Bodo Levkau
- Institute for Pathophysiology, West German Heart and Vascular Center, University Hospital Essen, 45122 Essen, Germany;
| |
Collapse
|
190
|
Mueller E. Browning and Graying: Novel Transcriptional Regulators of Brown and Beige Fat Tissues and Aging. Front Endocrinol (Lausanne) 2016; 7:19. [PMID: 26973598 PMCID: PMC4773441 DOI: 10.3389/fendo.2016.00019] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 02/15/2016] [Indexed: 11/13/2022] Open
Abstract
Obesity represents a major risk factor for the development of a number of metabolic disorders, including cardiovascular disease and type 2 diabetes. Since the discovery that brown and beige fat cells exist in adult humans and contribute to energy expenditure, increasing interest has been devoted to the understanding of the molecular switches turning on calorie utilization. It has been reported that the ability of thermogenic tissues to burn energy declines during aging, possibly contributing to the development of metabolic dysfunction late in life. This review will focus on the recently identified transcriptional modulators of brown and beige cells and will discuss the potential impact of some of these thermogenic factors on age-associated metabolic disorders.
Collapse
Affiliation(s)
- Elisabetta Mueller
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
- *Correspondence: Elisabetta Mueller,
| |
Collapse
|
191
|
Sun L, Tang XJ, Luo FM. Forkhead box protein A2 and T helper type 2-mediated pulmonary inflammation. World J Methodol 2015; 5:223-229. [PMID: 26713283 PMCID: PMC4686420 DOI: 10.5662/wjm.v5.i4.223] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 03/24/2015] [Accepted: 09/30/2015] [Indexed: 02/06/2023] Open
Abstract
The transcription factor forkhead box protein A2 (FOXA2, also known as hepatocyte nuclear factor 3β or transcription factor 3β), has been found to play pivotal roles in multiple phases of mammalian life, from the early development to the organofaction, and subsequently in homeostasis and metabolism in the adult. In the embryonic development period, FOXA2 is require d for the formation of the primitive node and notochord, and its absence results in embryonic lethality. Moreover, FOXA2 plays an important role not only in lung development, but also in T helper type 2 (Th2)-mediated pulmonary inflammation and goblet cell hyperplasia. In this article, the role of FOXA2 in lung development and Th2-mediated pulmonary inflammation, as well as in goblet cell hyperplasia, is reviewed. FOXA2 deletion in airway epithelium results into Th2-mediated pulmonary inflammation and goblet cell hyperplasia in developing lung. Leukotriene pathway and signal transducers and activators of transcription 6 pathway may mediate this inflammation through recruitment and activation of denditric cell during lung developments. FOXA2 is a potential treatment target for lung diseases with Th2 inflammation and goblet cell hyperplasia, such as asthma and chronic obstructive pulmonary disease.
Collapse
|
192
|
Wang R, Shi Y, Chen L, Jiang Y, Mao C, Yan B, Liu S, Shan B, Tao Y, Wang X. The ratio of FoxA1 to FoxA2 in lung adenocarcinoma is regulated by LncRNA HOTAIR and chromatin remodeling factor LSH. Sci Rep 2015; 5:17826. [PMID: 26658322 PMCID: PMC4675985 DOI: 10.1038/srep17826] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 11/06/2015] [Indexed: 12/16/2022] Open
Abstract
The lncRNA HOTAIR is a critical regulator of cancer progression. Chromatin remodeling factor LSH is critical for normal development of plants and mammals. However, the underlying mechanisms causing this in cancer are not entirely clear. The functional diversification of the FOXA1 and FOXA2 contributes to the target genes during evolution and carcinogenesis. Little is known about the ratio of FOXA1 to FOXA2 in cancer. We here found that both HOTAIR and LSH overexpression was significantly correlated with poor survival in patients with lung adenocarcinoma cancer (ADC). Also, the ratio of FOXA1 and FOXA2 is linked with poor survival in patients with lung ADC. HOTAIR regulates the ratio of FOXA1 to FOXA2 and migration and invasion. HOTAIR and the ratio of FOXA1 to FOXA2 are negatively correlated. HOTAIR knockdown inhibits migration and invasion. HOTAIR is associated with LSH, and this association linked with the binding of LSH in the promoter of FOXA1, not FOXA2. Targeted inhibition of HOTAIR suppresses the migratory and invasive properties. These data suggest that HOTAIR is an important mediator of the ratio of FOXA1 and FOXA2 and LSH involves in, and suggest that HOTAIR inhibition may represent a promising therapeutic option for suppressing lung ADC progression.
Collapse
Affiliation(s)
- Ranran Wang
- Department of Thoracic and Cardiovascular Surgery, Second Xiangya Hospital of Central South University, Changsha, China
| | - Ying Shi
- Cancer Research Institute, Central South University, Changsha, Hunan, 410078 China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Hunan, 410078 China.,Key Laboratory of Carcinogenesis, Ministry of Health, Hunan, 410078 China
| | - Ling Chen
- Cancer Research Institute, Central South University, Changsha, Hunan, 410078 China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Hunan, 410078 China.,Key Laboratory of Carcinogenesis, Ministry of Health, Hunan, 410078 China
| | - Yiqun Jiang
- Cancer Research Institute, Central South University, Changsha, Hunan, 410078 China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Hunan, 410078 China.,Key Laboratory of Carcinogenesis, Ministry of Health, Hunan, 410078 China
| | - Chao Mao
- Cancer Research Institute, Central South University, Changsha, Hunan, 410078 China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Hunan, 410078 China.,Key Laboratory of Carcinogenesis, Ministry of Health, Hunan, 410078 China
| | - Bin Yan
- Cancer Research Institute, Central South University, Changsha, Hunan, 410078 China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Hunan, 410078 China.,Key Laboratory of Carcinogenesis, Ministry of Health, Hunan, 410078 China
| | - Shuang Liu
- Center for Medicine Research, Xiangya Hospital, Central South University, Changsha, Hunan, 410078 China
| | - Bin Shan
- College of Medical Sciences, Washington State University Spokane, 412 E. Spokane Falls Boulevard, Spokane, WA 99202, USA
| | - Yongguang Tao
- Cancer Research Institute, Central South University, Changsha, Hunan, 410078 China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Hunan, 410078 China.,Key Laboratory of Carcinogenesis, Ministry of Health, Hunan, 410078 China
| | - Xiang Wang
- Department of Thoracic and Cardiovascular Surgery, Second Xiangya Hospital of Central South University, Changsha, China.,Cancer Research Institute, Central South University, Changsha, Hunan, 410078 China
| |
Collapse
|
193
|
Tekola-Ayele F, Doumatey AP, Shriner D, Bentley AR, Chen G, Zhou J, Fasanmade O, Johnson T, Oli J, Okafor G, Eghan BA, Agyenim-Boateng K, Adebamowo C, Amoah A, Acheampong J, Adeyemo A, Rotimi CN. Genome-wide association study identifies African-ancestry specific variants for metabolic syndrome. Mol Genet Metab 2015; 116:305-13. [PMID: 26507551 PMCID: PMC5292212 DOI: 10.1016/j.ymgme.2015.10.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 10/21/2015] [Accepted: 10/21/2015] [Indexed: 12/21/2022]
Abstract
The metabolic syndrome (MetS) is a constellation of metabolic disorders that increase the risk of developing several diseases including type 2 diabetes and cardiovascular diseases. Although genome-wide association studies (GWAS) have successfully identified variants associated with individual traits comprising MetS, the genetic basis and pathophysiological mechanisms underlying the clustering of these traits remain unclear. We conducted GWAS of MetS in 1427 Africans from Ghana and Nigeria followed by replication testing and meta-analysis in another continental African sample from Kenya. Further replication testing was performed in an African American sample from the Atherosclerosis Risk in Communities (ARIC) study. We found two African-ancestry specific variants that were significantly associated with MetS: SNP rs73989312[A] near CA10 that conferred increased risk (P=3.86 × 10(-8), OR=6.80) and SNP rs77244975[C] in CTNNA3 that conferred protection against MetS (P=1.63 × 10(-8), OR=0.15). Given the exclusive expression of CA10 in the brain, our CA10 finding strengthens previously reported link between brain function and MetS. We also identified two variants that are not African specific: rs76822696[A] near RALYL associated with increased MetS risk (P=7.37 × 10(-9), OR=1.59) and rs7964157[T] near KSR2 associated with reduced MetS risk (P=4.52 × 10(-8), Pmeta=7.82 × 10(-9), OR=0.53). The KSR2 locus displayed pleiotropic associations with triglyceride and measures of blood pressure. Rare KSR2 mutations have been reported to be associated with early onset obesity and insulin resistance. Finally, we replicated the LPL and CETP loci previously found to be associated with MetS in Europeans. These findings provide novel insights into the genetics of MetS in Africans and demonstrate the utility of conducting trans-ethnic disease gene mapping studies for testing the cosmopolitan significance of GWAS signals of cardio-metabolic traits.
Collapse
Affiliation(s)
- Fasil Tekola-Ayele
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Ayo P Doumatey
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Daniel Shriner
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Amy R Bentley
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Guanjie Chen
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jie Zhou
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | - Johnnie Oli
- University of Nigeria Teaching Hospital, Enugu, Nigeria
| | | | - Benjami A Eghan
- University of Science and Technology, Department of Medicine, Kumasi, Ghana
| | | | - Clement Adebamowo
- Department of Epidemiology and Public Health, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Albert Amoah
- University of Ghana Medical School, Department of Medicine, Accra, Ghana
| | - Joseph Acheampong
- University of Science and Technology, Department of Medicine, Kumasi, Ghana
| | - Adebowale Adeyemo
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Charles N Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
194
|
Garmhausen M, Hofmann F, Senderov V, Thomas M, Kandel BA, Habermann BH. Virtual pathway explorer (viPEr) and pathway enrichment analysis tool (PEANuT): creating and analyzing focus networks to identify cross-talk between molecules and pathways. BMC Genomics 2015; 16:790. [PMID: 26467653 PMCID: PMC4606501 DOI: 10.1186/s12864-015-2017-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 10/08/2015] [Indexed: 12/20/2022] Open
Abstract
Background Interpreting large-scale studies from microarrays or next-generation sequencing for further experimental testing remains one of the major challenges in quantitative biology. Combining expression with physical or genetic interaction data has already been successfully applied to enhance knowledge from all types of high-throughput studies. Yet, toolboxes for navigating and understanding even small gene or protein networks are poorly developed. Results We introduce two Cytoscape plug-ins, which support the generation and interpretation of experiment-based interaction networks. The virtual pathway explorer viPEr creates so-called focus networks by joining a list of experimentally determined genes with the interactome of a specific organism. viPEr calculates all paths between two or more user-selected nodes, or explores the neighborhood of a single selected node. Numerical values from expression studies assigned to the nodes serve to score identified paths. The pathway enrichment analysis tool PEANuT annotates networks with pathway information from various sources and calculates enriched pathways between a focus and a background network. Using time series expression data of atorvastatin treated primary hepatocytes from six patients, we demonstrate the handling and applicability of viPEr and PEANuT. Based on our investigations using viPEr and PEANuT, we suggest a role of the FoxA1/A2/A3 transcriptional network in the cellular response to atorvastatin treatment. Moreover, we find an enrichment of metabolic and cancer pathways in the Fox transcriptional network and demonstrate a patient-specific reaction to the drug. Conclusions The Cytoscape plug-in viPEr integrates –omics data with interactome data. It supports the interpretation and navigation of large-scale datasets by creating focus networks, facilitating mechanistic predictions from –omics studies. PEANuT provides an up-front method to identify underlying biological principles by calculating enriched pathways in focus networks. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-2017-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marius Garmhausen
- CECAD Research Center, Joseph-Stelzmann-Str. 26, 50931, Cologne, Germany.
| | - Falko Hofmann
- Gregor Mendel Institute of Molecular Plant Biology, Austrian Acacdemy of Sciences, Vienna Biocenter (VBC), Dr. Bohr-Gasse 3, 1030, Vienna, Austria.
| | - Viktor Senderov
- Research Group Computational Biology, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152, Martinsried, Germany. .,Present address: Pensoft Publisher, 1700, Sofia, Bulgaria.
| | - Maria Thomas
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tübingen, Auerbachstr. 112, 70376, Stuttgart, Germany.
| | - Benjamin A Kandel
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tübingen, Auerbachstr. 112, 70376, Stuttgart, Germany. .,Present address: Hain Lifescience GmbH, Hardwiesenstr. 1, 72147, Nehren, Germany.
| | - Bianca Hermine Habermann
- Research Group Computational Biology, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152, Martinsried, Germany.
| |
Collapse
|
195
|
Lempradl A, Pospisilik JA, Penninger JM. Exploring the emerging complexity in transcriptional regulation of energy homeostasis. Nat Rev Genet 2015; 16:665-81. [PMID: 26460345 DOI: 10.1038/nrg3941] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Obesity and its associated diseases are expected to affect more than 1 billion people by the year 2030. These figures have sparked intensive research into the molecular control of food intake, nutrient distribution, storage and metabolism--processes that are collectively termed energy homeostasis. Recent decades have also seen dramatic developments in our understanding of gene regulation at the signalling, chromatin and post-transcriptional levels. The seemingly exponential growth in this complexity now poses a major challenge for translational researchers in need of simplified but accurate paradigms for clinical use. In this Review, we consider the current understanding of transcriptional control of energy homeostasis, including both transcriptional and epigenetic regulators, and crosstalk between pathways. We also provide insights into emerging developments and challenges in this field.
Collapse
Affiliation(s)
- Adelheid Lempradl
- Max Planck Institute of Immunobiology and Epigenetics, Stuebeweg 51, 79108 Freiburg, Germany
| | - J Andrew Pospisilik
- Max Planck Institute of Immunobiology and Epigenetics, Stuebeweg 51, 79108 Freiburg, Germany
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr Bohr-Gasse 3, 1030 Vienna, Austria
| |
Collapse
|
196
|
Hepatic Long Intergenic Noncoding RNAs: High Promoter Conservation and Dynamic, Sex-Dependent Transcriptional Regulation by Growth Hormone. Mol Cell Biol 2015; 36:50-69. [PMID: 26459762 DOI: 10.1128/mcb.00861-15] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 10/02/2015] [Indexed: 01/04/2023] Open
Abstract
Long intergenic noncoding RNAs (lincRNAs) are increasingly recognized as key chromatin regulators, yet few studies have characterized lincRNAs in a single tissue under diverse conditions. Here, we analyzed 45 mouse liver RNA sequencing (RNA-Seq) data sets collected under diverse conditions to systematically characterize 4,961 liver lincRNAs, 59% of them novel, with regard to gene structures, species conservation, chromatin accessibility, transcription factor binding, and epigenetic states. To investigate the potential for functionality, we focused on the responses of the liver lincRNAs to growth hormone stimulation, which imparts clinically relevant sex differences to hepatic metabolism and liver disease susceptibility. Sex-biased expression characterized 247 liver lincRNAs, with many being nuclear RNA enriched and regulated by growth hormone. The sex-biased lincRNA genes are enriched for nearby and correspondingly sex-biased accessible chromatin regions, as well as sex-biased binding sites for growth hormone-regulated transcriptional activators (STAT5, hepatocyte nuclear factor 6 [HNF6], FOXA1, and FOXA2) and transcriptional repressors (CUX2 and BCL6). Repression of female-specific lincRNAs in male liver, but not that of male-specific lincRNAs in female liver, was associated with enrichment of H3K27me3-associated inactive states and poised (bivalent) enhancer states. Strikingly, we found that liver-specific lincRNA gene promoters are more highly species conserved and have a significantly higher frequency of proximal binding by liver transcription factors than liver-specific protein-coding gene promoters. Orthologs for many liver lincRNAs were identified in one or more supraprimates, including two rat lincRNAs showing the same growth hormone-regulated, sex-biased expression as their mouse counterparts. This integrative analysis of liver lincRNA chromatin states, transcription factor occupancy, and growth hormone regulation provides novel insights into the expression of sex-specific lincRNAs and their potential for regulation of sex differences in liver physiology and disease.
Collapse
|
197
|
Endometrial gene expression profile of pregnant sows with extreme phenotypes for reproductive efficiency. Sci Rep 2015; 5:14416. [PMID: 26435523 PMCID: PMC5155628 DOI: 10.1038/srep14416] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 08/19/2015] [Indexed: 12/22/2022] Open
Abstract
Prolificacy can directly impact porcine profitability, but large genetic variation and low heritability have been found regarding litter size among porcine breeds. To identify key differences in gene expression associated to swine reproductive efficiency, we performed a transcriptome analysis of sows' endometrium from an Iberian x Meishan F2 population at day 30-32 of gestation, classified according to their estimated breeding value (EBV) as high (H, EBV > 0) and low (L, EBV < 0) prolificacy phenotypes. For each sample, mRNA and small RNA libraries were RNA-sequenced, identifying 141 genes and 10 miRNAs differentially expressed between H and L groups. We selected four miRNAs based on their role in reproduction, and five genes displaying the highest differences and a positive mapping into known reproductive QTLs for RT-qPCR validation on the whole extreme population. Significant differences were validated for genes: PTGS2 (p = 0.03; H/L ratio = 3.50), PTHLH (p = 0.03; H/L ratio = 3.69), MMP8 (p = 0.01; H/L ratio =4.41) and SCNN1G (p = 0.04; H/L ratio = 3.42). Although selected miRNAs showed similar expression levels between H and L groups, significant correlation was found between the expression level of ssc-miR-133a (p < 0.01) and ssc-miR-92a (p < 0.01) and validated genes. These results provide a better understanding of the genetic architecture of prolificacy-related traits and embryo implantation failure in pigs.
Collapse
|
198
|
Ma X, Xu L, Mueller E. Calorie hoarding and thrifting: Foxa3 finds a way. Adipocyte 2015; 4:325-8. [PMID: 26451291 DOI: 10.1080/21623945.2015.1028700] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 03/04/2015] [Accepted: 03/06/2015] [Indexed: 12/11/2022] Open
Abstract
Obesity and diabetes are major health concerns worldwide. Western diets, often calorically rich, paired with sedentary habits are driving the current worldwide epidemic of pediatric and adult obesity. In addition, age related energy imbalances lead to increased adiposity and metabolic disorders later in life, making the middle aged population particularly susceptible. Here we discuss how Forkhead box A3 (Foxa3), a family member of the forkhead box binding proteins, can potentially contribute to pathology by playing a double role in metabolism. Recent data revealed that Foxa3 favors the selective expansion of visceral depots under high caloric conditions (e.g., high fat diet) and suppresses subcutaneous fat tissue energy expenditure during aging. This evidence suggests that Foxa3 acts to both preserve and conserve calories, by accumulating fat and by reducing metabolic burn. In other words, Foxa3 appears to function to enable energy "hoarding," which may be critical for survival of organisms with intermittent exposure to external caloric sources, but pathologic in circumstances where calories are abundant. Understanding how this "calorie hoarder gene" functions may suggest approaches to combat obesity and associated metabolic disorders.
Collapse
|
199
|
Transcription factors FOXA1 and FOXA2 maintain dopaminergic neuronal properties and control feeding behavior in adult mice. Proc Natl Acad Sci U S A 2015; 112:E4929-38. [PMID: 26283356 DOI: 10.1073/pnas.1503911112] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Midbrain dopaminergic (mDA) neurons are implicated in cognitive functions, neuropsychiatric disorders, and pathological conditions; hence understanding genes regulating their homeostasis has medical relevance. Transcription factors FOXA1 and FOXA2 (FOXA1/2) are key determinants of mDA neuronal identity during development, but their roles in adult mDA neurons are unknown. We used a conditional knockout strategy to specifically ablate FOXA1/2 in mDA neurons of adult mice. We show that deletion of Foxa1/2 results in down-regulation of tyrosine hydroxylase, the rate-limiting enzyme of dopamine (DA) biosynthesis, specifically in dopaminergic neurons of the substantia nigra pars compacta (SNc). In addition, DA synthesis and striatal DA transmission were reduced after Foxa1/2 deletion. Furthermore, the burst-firing activity characteristic of SNc mDA neurons was drastically reduced in the absence of FOXA1/2. These molecular and functional alterations lead to a severe feeding deficit in adult Foxa1/2 mutant mice, independently of motor control, which could be rescued by L-DOPA treatment. FOXA1/2 therefore control the maintenance of molecular and physiological properties of SNc mDA neurons and impact on feeding behavior in adult mice.
Collapse
|
200
|
Transcription factor FOXA2-centered transcriptional regulation network in non-small cell lung cancer. Biochem Biophys Res Commun 2015; 463:961-7. [DOI: 10.1016/j.bbrc.2015.06.042] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 06/05/2015] [Indexed: 11/21/2022]
|