Rasd1 Interacts with Ear2 (Nr2f6) to Regulate Renin Transcription Jen Jen Tan1,2, Shufen Angeline Ong1, Ken-Shiung Chen1*

Total Page:16

File Type:pdf, Size:1020Kb

Rasd1 Interacts with Ear2 (Nr2f6) to Regulate Renin Transcription Jen Jen Tan1,2, Shufen Angeline Ong1, Ken-Shiung Chen1* Tan et al. BMC Molecular Biology 2011, 12:4 http://www.biomedcentral.com/1471-2199/12/4 RESEARCHARTICLE Open Access Rasd1 interacts with Ear2 (Nr2f6) to regulate renin transcription Jen Jen Tan1,2, Shufen Angeline Ong1, Ken-Shiung Chen1* Abstract Background: The Rasd1 protein is a dexamethasone induced monomeric Ras-like G protein that oscillates in the suprachiasmatic nucleus (SCN). Previous studies have shown that Rasd1 modulates multiple signaling cascades. However, it is still unclear exactly how Rasd1 carries out its function. Studying protein-protein interactions involving Rasd1 may provide insights into its biological functions in different contexts. Results: To further explore the molecular function of Rasd1, we performed a yeast two-hybrid screen and identified Ear2, a negative regulator of renin transcription, as an interaction partner of Rasd1. We validated the interaction in vitro and in transfected COS-7 cells. We further confirmed the interaction of endogenous Rasd1 and Ear2 from HEK293T cell and mouse brain extract. Rasd1 inhibited transcriptional repression by Ear2 on a renin promoter-luciferase reporter construct both in the presence and absence of all-trans-retinoic acid. Moreover, real- time RT-PCR showed upregulation of endogenous renin transcription in As4.1 cells over-expressing Rasd1. We demonstrated that the ligand binding domain of Ear2 is required for physical and functional interaction between the two proteins. In addition, we demonstrated that shRNA-mediated knockdown of Rasd1 results in further repression of Ear2-mediated renin transcription, whereas induction of Rasd1 by dexamethasone counteracts the effects of shRNA-mediated Rasd1 knockdown. Finally, our study showed that Rasd1 missense mutations not only attenuate their physical interaction with Ear2 but also abolish their ability to counteract repression of renin transcription mediated by Ear2. Conclusions: Our study provides evidence for physical and functional interactions between Rasd1 and Ear2. The results suggest that their interactions are involved in renin transcriptional regulation. These findings not only reveal a novel role for Rasd1-medated signaling but also provide the basis for potential intervention of renin expression. Background enhancer was identified ~2.6 kb upstream of the mouse The renin-angiotensin system plays a major physiological renin gene, and this enhancer is homologous to a role in the control of blood pressure, fluid volume and sequence ~12 kb upstream of the human renin gene electrolyte balance. A functional renin-angiotensin sys- [6-8]. The transcriptional enhancer contains several tran- tem is also essential for the normal development of the scription factor binding sites that have both excitatory mammalian renal system [1]. Renin, an aspartyl protease, and inhibitory regulatory functions [9-12]. is the rate-limiting enzyme in the renin-angiotensin enzy- One protein that has been identified to bind to and matic cascade which leads to the production of angioten- regulate the renin enhancer is Ear2 [13]. It was deter- sin II (Ang II), a vasoactive peptide and major effector mined that Ear2 negatively regulates renin expression by molecule in the renin-angiotensin system [2,3]. Renin competing with retinoic acid receptor/ retinoid X recep- gene expression is largely regulated at the transcriptional tor (RAR/RXR) for binding to the retinoic acid response level, although post transcriptional regulation has also elements (RARE) on the renin enhancer [13]. Ear2 is an been reported [4,5]. A potent classical transcriptional orphan nuclear hormone receptor that belongs to the chicken ovalbumin upstream promoter-transcription * Correspondence: [email protected] factors (COUP-TF) gene family [14]. COUP-TFs have 1School of Biological Sciences, Department of Genomics and Genetics, been shown to bind to a number of variable direct and Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore indirect repeats with different spacings between the Full list of author information is available at the end of the article © 2011 Tan et al; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Tan et al. BMC Molecular Biology 2011, 12:4 Page 2 of 17 http://www.biomedcentral.com/1471-2199/12/4 repeats [15] to affect a large plethora of genes [16-19]. Table 1 Proteins identified from the yeast two-hybrid COUP-TFs have been proposed to inhibit transactivation screen of other nuclear receptors through multiple mechanisms, No Protein/ Gene Name Genbank including competitively binding to regulatory elements, Gene Accession No. competitively binding to RXR, mediating active repres- 1 Cenpb Mus musculus centromere protein B NM_007682.2 sion via direct binding to regulatory elements, and med- 2 Gnb1 Mus musculus guanine nucleotide NM_008142.3 iating transrepression of another nuclear receptor binding protein, beta 1 without binding to DNA itself [14,20-22]. Nuclear hor- 3 Nr2f6 Mus musculus nuclear receptor NM_010150.2 mone receptors have the ability to bind directly to DNA subfamily 2, group F, member 6 and regulate the expression of specific target genes; 4 Plscr1 Mus musculus phospholipid NM_011636.2 scramblase 1 therefore, they are extremely crucial to the development, 5 Sh3gl2 Mus musculus SH3-domain GRB2-like 2 NM_019535.2 homeostasis and metabolism of an organism [16,20,21, 23-25]. Ear2 is expressed in tissues of all major systems, 6 Supt16 h Mus musculus suppressor of Ty 16 NM_033618.2 homolog and its expression has been implicated in the regulation 7 Trp53bp2 Mus musculus transformation related NM_173378.2 of gene expression for normal embryo development protein 53 binding protein 2 [26,27]. Ear2 knockout mice are viable and fertile, but 8 Ywhah Mus musculus tyrosine 3- NM_011738.1 they possess circadian and nociception defects and monooxygenase/tryptophan 5- abnormal locus coeruleus (LC) development [25]. monooxygenase activation protein, eta Using a yeast two-hybrid system, we identified Ear2 as polypeptide an interacting protein of Rasd1. Rasd1 is a G-protein that belongs to the RAS superfamily of small GTPases. It was first discovered as a dexamethasone-inducible between Rasd1 and Ear2, we conducted an in vitro bind- gene in the AtT-20 pituitary cell line [28]. Rasd1 is an ing study utilizing transfected COS-7 cells. GST-Ear2 oscillating gene specific to the suprachiasmatic nucleus was immobilized on GSH-linked beads. The GST fusion (SCN) [29]. Rasd1 modulates several signaling cascades protein containing full-length Ear2 interacted with HA- and has several known functions. It interacts with tagged full-length Rasd1 from the lysate of transfected neuronal nitric oxide synthase via CAPON to enhance COS-7 cells (Figure 1A, lane 1), whereas GST itself physiological nitric oxide signaling [30]. Rasd1 is also showed no such interaction (Figure 1A, lane 2). crucial in the regulation of the responsiveness of the To test whether Rasd1 and Ear2 interact in intact circadian clock to external stimuli [31]. In this study, we mammalian cells, we performed co-transfection and demonstrate that the interaction between Rasd1 and co-precipitation experiments. pHisHA-Rasd1 was Ear2 regulates renin transcription. Our findings reveal a co-transfected together with pGST-Ear2 into COS-7 novel regulatory role of Rasd1 and a novel regulatory cells. As a control, pHisHA-Rasd1 was co-trans- mechanism for Ear2-mediated renin transcription. fected with pxJGST into COS-7 cells. GST-Ear2 or GST from transfected COS-7 cell lysate was Results immobilized on GSH-linked magnetic particles. Pre- Identification of Ear2 as a Rasd1 interacting protein-To cipitation of GST-Ear2 resulted in the co-precipita- identify the proteins that are associated with Rasd1, we tion of HisHA-Rasd1 (Figure 1B, lane 1), whereas conducted a yeast two-hybrid screen. A mouse brain precipitation of GST-tag alone did not co-precipitate cDNA library was screened against full length Rasd1. All HisHA-Rasd1 (Figure 1B, lane 2). This demonstrates surviving colonies after nutritional selection were that Rasd1 and Ear2 form a physiological complex in cul- screened for LacZ expression using b-galactosidase. tured cells. Additionally, a co-immunoprecipitation assay Table 1 lists the proteins that were identified. Ear2 is was carried out using lysate derived from COS-7 co-trans- the protein for which the highest number of indepen- fected with pHisHA-Rasd1 and pGST-Ear2. As a control, dent clones was obtained, with a total of three clones experiments were also conducted using lysate derived identified. To confirm the specificity of interaction from COS-7 cells co-transfected with pHisHA-Rasd1 and between Rasd1 and Ear2, a separate yeast two-hybrid pxJGST. Immunoprecipitation of GST-Ear2 with analysis was performed by mating yeast containing the anti-GST antibody resulted in the co-precipitation of Ear2 cDNA plasmid with yeast that already carried HisHA-Rasd1 (Figure 1C, lane 1). There was no non-spe- Rasd1. This was followed by nutritional selection and cific interaction between GST tag and HisHA-Rasd1 b-galactosidase assay. Ear2 again showed positive inter- (Figure 1C, lane 2). actions with Rasd1 (not shown). Interaction of endogenous Rasd1 and Ear2 was identi- Rasd1 and Ear2 interact in vitro and in living cells- fied in HEK293T cells and the mouse brain-Theexis- To confirm the specificity of biochemical interaction tence of endogenous Rasd1-Ear2 complexes was further Tan et al. BMC Molecular Biology 2011, 12:4 Page 3 of 17 http://www.biomedcentral.com/1471-2199/12/4 Figure 1 Rasd1 and Ear2 interact in vitro and in living cells.(A)In vitro binding of GST-Ear2 and HisHA-Rasd1. COS-7 cells were transfected with plasmids expressing the indicated constructs. Cell lysates containing HisHA-Rasd1 was incubated with immobilized GST-Ear2 or GST, and specifically bound HisHA-Rasd1 was eluted by heating at 95°C for 10 minutes, and detected by Western blotting with Anti-HA antibody.
Recommended publications
  • Lipid Related Genes Altered in NASH Connect Inflammation in Liver Pathogenesis Progression to HCC: a Canonical Pathway
    Lipid related genes altered in NASH connect inflammation in liver pathogenesis progression to HCC: a canonical pathway Christophe Desterke1, Franck Chiappini2* 1 Inserm, U935, Villejuif, F-94800, France 2 Cell Growth and Tissue Repair (CRRET) Laboratory, Université Paris-Est Créteil (UPEC), EA 4397 / ERL CNRS 9215, F-94010, Créteil, France. Corresponding author: *Franck Chiappini. Laboratoire du CRRET (Croissance, Réparation et Régénération Tissulaires), Université Paris-Est Créteil, 61 avenue du Général de Gaulle F- 94010, Créteil Cedex, Val de Marne, France. Email address: [email protected]; Tel: +33(0)145177080; Fax: +33(0)145171816 Supplementary Information Supplementary Datasets Table S1: Text-mining list of genes associated in PubMed literature with lipid related keywords. Supplementary Datasets Table S2: Expression fold change of lipid related genes found differentially expressed between NASH and healthy obese liver samples. Supplementary Datasets Table S3: Liver as principal filter for prioritization of lipid related genes found differentially expressed in NASH. Supplementary Datasets Table S4: Gene prioritization secondary filters (immunological, inflammation, liver pathogenesis progression) table found with lipid related genes differentially expressed in NASH. Supplementary Datasets Table S5: Identification of protein partners of YWHAZ gene using InnateDB database. Supplementary Datasets Table S1: Text-mining list of genes associated in PubMed literature with lipid related keywords. Ranking of "lipidic" textmining Gene symbol
    [Show full text]
  • Pathogenesis of Coronary Artery Disease: Focus on Genetic Risk Factors and Identification of Genetic Variants
    The Application of Clinical Genetics Dovepress open access to scientific and medical research Open Access Full Text Article REVIEW Pathogenesis of coronary artery disease: focus on genetic risk factors and identification of genetic variants Sergi Sayols-Baixeras Abstract: Coronary artery disease (CAD) is the leading cause of death and disability worldwide, Carla Lluís-Ganella and its prevalence is expected to increase in the coming years. CAD events are caused by the Gavin Lucas interplay of genetic and environmental factors, the effects of which are mainly mediated through Roberto Elosua cardiovascular risk factors. The techniques used to study the genetic basis of these diseases have evolved from linkage studies to candidate gene studies and genome-wide association studies. Cardiovascular Epidemiology and Genetics Research Group, Institut Linkage studies have been able to identify genetic variants associated with monogenic diseases, Hospital del Mar d’Investigacions whereas genome-wide association studies have been more successful in determining genetic Mèdiques, Barcelona, Spain variants associated with complex diseases. Currently, genome-wide association studies have identified approximately 40 loci that explain 6% of the heritability of CAD. The application of this knowledge to clinical practice is challenging, but can be achieved using various strategies, such as genetic variants to identify new therapeutic targets, personal genetic information to improve disease risk prediction, and pharmacogenomics. The main aim of this narrative review is to provide a general overview of our current understanding of the genetics of coronary artery disease and its potential clinical utility. Keywords: coronary artery disease, pathogenesis, genetic risk factors, genetic variants Introduction Coronary artery disease (CAD) is the principal individual cause of mortality and morbidity worldwide.
    [Show full text]
  • Effects of in Utero Exposure to Di-N-Butyl Phthalate on Testicular
    Article Effects of In Utero Exposure to Di‐n‐Butyl Phthalate on Testicular Development in Rat Tan Ma 1,2,†, Xiaoqin Yin 1,2,†, Ruitong Han 1,2, Jie Ding 1,2, Huan Zhang 3,*, Xiaodong Han 1,2,* and Dongmei Li 1,2,* 1 Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing 210093, China; [email protected] (T.M.); [email protected] (X.Y.); [email protected] (R.H.); [email protected] (J.D.) 2 Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China 3 Department of Clinical and Experimental Medicine, Linköping University, SE‐581 83 Linköping, Sweden * Correspondence: [email protected] (H.Z.); [email protected] (X.H.); [email protected] (D.L.) † These authors contributed equally to this work. Received: 17 October 2017; Accepted: 20 October 2017; Published: 24 October 2017 Abstract: Humans are inevitably exposed to ubiquitous phthalate esters (PAEs). In utero exposure to di‐n‐butyl phthalate (DBP) induces abnormal development of the testis and reproductive tract in male offspring, which correspond closely with the human condition of testicular dysgenesis syndrome (TDS)‐like syndrome. However, the underlying mechanisms have not been elucidated in detail. In this study, pregnant rats were orally exposed to either corn oil (controls) or DBP at three different doses by gavage during Gestational Days 12.5–21.5. Pathological examinations were performed for toxicity evaluation. Proliferation and apoptosis related proteins (ras related dexamethasone induced 1 (Rasd1), mitogen‐activated protein kinase kinases1/2 (MEK1/2), Bcl‐2, and Bax) were measured for mechanisms exploration.
    [Show full text]
  • Role of Estrogen and RAS Signaling in Repeated Implantation Failure
    BMB Rep. 2018; 51(5): 225-229 BMB www.bmbreports.org Reports Invited Mini Review Role of estrogen and RAS signaling in repeated implantation failure Kwonho Hong1 & Youngsok Choi2,* 1Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, 2Department of Biomedical Science, CHA University, Seongnam 13488, Korea In humans, hormonal regulation is crucial for the preparation RAS signal as well as the implications for patient with repeated of uterine environment leading to either successful implantation failure. implantation or menstrual cycle. Estrogen is a pivotal female steroid hormone that regulates the uterine dynamics along ESTROGEN SIGNALING with progesterone in the estrous and menstrual cycles in humans. Estrogen signals act via nuclear estrogen receptor or Estrogen production from the ovary during the estrous or membrane-bound receptor. The membrane-bound estrogen menstrual cycle receptor plays a crucial role in the rapid response of estrogen Estrogen exists in three forms such as estrone, estradiol, and in the uterine epithelium. Recently, RASD1 has received estriol (1, 2). Estrogen is produced in various tissues including attention as a novel signal transducer of estrogen in various the ovary, placenta, fat, and the liver in females (3, 4). It is the systems including female reproductive organs. In this review, primary source of estrogen production. Two different somatic we discuss the regulation of estrogen and RASD1 signaling in cells known as theca and granulosa cells surround the oocyte the uterus and also provide insights into RAS as a novel in the ovarian follicles (3-5). Estrogen biosynthesis depends on signaling molecule in repeated implantation failure. [BMB P450 aromatase enzyme activity, which catalyzes the Reports 2018; 51(5): 225-229] hydroxylation of estrogen precursors, adrostenedione or testosterone (Fig.
    [Show full text]
  • Single-Cell Transcriptome Profiling of the Kidney Glomerulus Identifies Key Cell Types and Reactions to Injury
    BASIC RESEARCH www.jasn.org Single-Cell Transcriptome Profiling of the Kidney Glomerulus Identifies Key Cell Types and Reactions to Injury Jun-Jae Chung ,1 Leonard Goldstein ,2 Ying-Jiun J. Chen,2 Jiyeon Lee ,1 Joshua D. Webster,3 Merone Roose-Girma,2 Sharad C. Paudyal,4 Zora Modrusan,2 Anwesha Dey,5 and Andrey S. Shaw1 Due to the number of contributing authors, the affiliations are listed at the end of this article. ABSTRACT Background The glomerulus is a specialized capillary bed that is involved in urine production and BP control. Glomerular injury is a major cause of CKD, which is epidemic and without therapeutic options. Single-cell transcriptomics has radically improved our ability to characterize complex organs, such as the kidney. Cells of the glomerulus, however, have been largely underrepresented in previous single-cell kidney studies due to their paucity and intractability. Methods Single-cell RNA sequencing comprehensively characterized the types of cells in the glomerulus from healthy mice and from four different disease models (nephrotoxic serum nephritis, diabetes, doxo- rubicin toxicity, and CD2AP deficiency). Results Allcelltypesintheglomeruluswereidentified using unsupervised clustering analysis. Novel marker genes and gene signatures of mesangial cells, vascular smooth muscle cells of the afferent and efferent arteri- oles, parietal epithelial cells, and three types of endothelial cells were identified. Analysis of the disease models revealed cell type–specific and injury type–specific responses in the glomerulus, including acute activation of the Hippo pathway in podocytes after nephrotoxic immune injury. Conditional deletion of YAP or TAZ resulted in more severe and prolonged proteinuria in response to injury, as well as worse glomerulosclerosis.
    [Show full text]
  • Up-Regulating of RASD1 and Apoptosis of DU-145 Human Prostate Cancer Cells Induced by Formononetin in Vitro
    DOI:http://dx.doi.org/10.7314/APJCP.2014.15.6.2835 Up-regulating of RASD1 and Apoptosis of DU-145 Human Prostate Cancer Cells Induced by Formononetin in Vitro RESEARCH ARTICLE Up-regulating of RASD1 and Apoptosis of DU-145 Human Prostate Cancer Cells Induced by Formononetin in Vitro Xiao-Jia Liu&, Yun-Qian Li&, Qiu-Yue Chen, Sheng-Jun Xiao, Si-En Zeng* Abstract Prostate cancer is one of the most prevalent malignant cancers in men. The isoflavone formononetin is a main active component of red clover plants. In the present study, we assessed the effect of formononetin on human prostate cancer DU-145 cells in vitro, and elucidated posssible mechanisms. DU-145 cells were treated with different concentrations of formononetin and cell proliferation was assessed by MTT assay, cell apoptosis by Hoechst 33258 and flow cytometry, and protein levels of RASD1, Bcl-2 and Bax by Western blotting. The results showed that formononetin inhibited the proliferation of DU-145 cells in a dose-dependent manner. DU-145 cells treated with different concentrations of formononetin displayed obvious morphological changes of apoptosis under fluorescence microscopy. In addition, formononetin increased the proportion of early apoptotic DU-145 cells, down-regulated the protein levels of Bcl-2 and up-regulated those of RASD1 and Bax. The level of RASD1 reached its maximum at 48h post-treatment, and rapidly decreased thereafter. Together, we present evidence that formononetin triggered cell apoptosis through the mitochondrial apoptotic pathway by up-regulating RASD1. Keywords: Formononetin - prostate cancer cells - apoptosis - RASD1 Asian Pac J Cancer Prev, 15 (6), 2835-2839 Introduction which has been wildly used for centuries.
    [Show full text]
  • Biomarkers, Master Regulators and Genomic Fabric Remodeling in a Case of Papillary Thyroid Carcinoma
    G C A T T A C G G C A T genes Article Biomarkers, Master Regulators and Genomic Fabric Remodeling in a Case of Papillary Thyroid Carcinoma Dumitru A. Iacobas Personalized Genomics Laboratory, CRI Center for Computational Systems Biology, Roy G Perry College of Engineering, Prairie View A&M University, Prairie View, TX 77446, USA; [email protected]; Tel.: +1-936-261-9926 Received: 1 August 2020; Accepted: 1 September 2020; Published: 2 September 2020 Abstract: Publicly available (own) transcriptomic data have been analyzed to quantify the alteration in functional pathways in thyroid cancer, establish the gene hierarchy, identify potential gene targets and predict the effects of their manipulation. The expression data have been generated by profiling one case of papillary thyroid carcinoma (PTC) and genetically manipulated BCPAP (papillary) and 8505C (anaplastic) human thyroid cancer cell lines. The study used the genomic fabric paradigm that considers the transcriptome as a multi-dimensional mathematical object based on the three independent characteristics that can be derived for each gene from the expression data. We found remarkable remodeling of the thyroid hormone synthesis, cell cycle, oxidative phosphorylation and apoptosis pathways. Serine peptidase inhibitor, Kunitz type, 2 (SPINT2) was identified as the Gene Master Regulator of the investigated PTC. The substantial increase in the expression synergism of SPINT2 with apoptosis genes in the cancer nodule with respect to the surrounding normal tissue (NOR) suggests that SPINT2 experimental overexpression may force the PTC cells into apoptosis with a negligible effect on the NOR cells. The predictive value of the expression coordination for the expression regulation was validated with data from 8505C and BCPAP cell lines before and after lentiviral transfection with DDX19B.
    [Show full text]
  • An Important Region in Coronary Artery Disease: a Panel Approach to Investigation of the Coronary Artery Disease Etiology
    Int J Cardiovasc Pract Review Article April 2019, Volume 4, Issue 2 (21-35) 9P21.3 locus; An Important Region in Coronary Artery Disease: A Panel Approach to Investigation of the Coronary Artery Disease Etiology Soodeh Omidi 1, Fatemeh Ebrahimzadeh 2, Samira Kalayinia 3,* 1 Department of Genetic, Faculty of Advanced Medical Technologies, Golestan University of Medical Science (GUMS), Gorgan, Iran 2 Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences (ZUMS), Zanjan, Iran 3 Cardiogenetics Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran * Corresponding author: Samira Kalayinia, Ph.D. Cardiogenetics Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical DOI: 10.29252/ijcp-25001 Sciences, Tehran, Iran. Tel: +98-2123923033, Fax: +98-2122663213, E-mail: [email protected] Submitted: 07-04-2019 Abstract Accepted: 06-05-2019 Coronary artery disease (CAD) is a disease of major concern worldwide. It is the main Keywords: cause of mortality in many societies and improving the understanding about the CAD Etiology mechanism, progression and treatment, is necessary. Recent discovery of genetic factors Heart Disease underlying CAD has improved our knowledge of the disease in support of well-known Genome Wide Association traditional risk factors. Genotype-environment interaction is known as the main risk Study factor. Loci on many different chromosomes have been identified as a risk factors that © 2019. International Journal increase CAD susceptibility. Here we performed a comprehensive literature review of Cardiovascular Practice. pinpointing hotspot loci involved in CAD pathogenicity. The 9p21.3 locus is the most common region associated with CAD and its specific structure and function have been remarkable in many studies.
    [Show full text]
  • Induction of IL-6 in Transcriptional Networks in Corneal Epithelial Cells After Herpes Simplex Virus Type 1 Infection
    Cornea Induction of IL-6 in Transcriptional Networks in Corneal Epithelial Cells after Herpes Simplex Virus Type 1 Infection Yuki Terasaka, Dai Miyazaki, Keiko Yakura, Tomoko Haruki, and Yoshitsugu Inoue 1–3 PURPOSE. To determine the transcriptional responses of human mimicry. Herpetic stromal keratitis (HSK) is exacerbated by corneal epithelial cells (HCECs) after herpes simplex virus type frequent reactivation of latent HSV-1 in the trigeminal ganglia, (HSV)-1 infection and to identify the critical inflammatory ele- which can eventually lead to severe corneal opacity that re- ment(s). quires corneal transplantation.4–6 METHOD. Immortalized HCECs were infected with HSV-1, and The CECs are the primary target of HSV infections, and they the global transcriptional profile determined. Molecular signal- serve as an innate barrier to deeper invasion before the devel- ing networks were constructed from the HSV-1-induced tran- opment of acquired immunity. The CECs respond immediately scriptomes. The relationships of the identified networks were to HSV exposure by releasing inflammatory mediators that confirmed by real-time-PCR and ELISA. Contributions of the recruit leukocytes, including neutrophils and macrophages. critical network nodes were further evaluated by protein array The primary responses are needed to establish T-lymphocyte- analyses as candidates for inflammatory element induction. based acquired immunity. Thus, understanding how CECs re- spond to HSV infection is important for understanding how the RESULTS. HSV-1 infection induced a global transcriptional re- eye reacts to a viral invasion. sponse, with 412 genes significantly activated or suppressed Ͻ Ͻ Ͼ CECs react to HSV-1 infection by expressing proinflamma- compared with mock-infected HCECs (P 0.05, 2 or 0.5 tory cytokines, including interferon (IFN)-␤, interleukin (IL)-6, threshold).
    [Show full text]
  • Dexras1 Links Glucocorticoids to Insulin-Like Growth Factor-1 Signaling in Adipogenesis Received: 16 February 2016 Hyo Jung Kim1, Jiyoung Y
    www.nature.com/scientificreports OPEN Dexras1 links glucocorticoids to insulin-like growth factor-1 signaling in adipogenesis Received: 16 February 2016 Hyo Jung Kim1, Jiyoung Y. Cha2, Jo Woon Seok1,3, Yoonjeong Choi1,3, Bo Kyung Yoon1,3, Accepted: 06 June 2016 Hyeonjin Choi1,†, Jung Hwan Yu1,3, Su Jin Song1,3, Ara Kim1,3, Hyemin Lee1,4, Daeun Kim1,3, Published: 27 June 2016 Ji Yoon Han1,3 & Jae-woo Kim1,3,4 Glucocorticoids are associated with obesity, but the underlying mechanism by which they function remains poorly understood. Previously, we showed that small G protein Dexras1 is expressed by glucocorticoids and leads to adipocyte differentiation. In this study, we explored the mechanism by which Dexras1 mediates adipogenesis and show a link to the insulin-like growth factor-1 (IGF-1) signaling pathway. Without Dexras1, the activation of MAPK and subsequent phosphorylation of CCAAT/enhancer binding protein β (C/EBPβ) is abolished, thereby inhibiting mitotic clonal expansion and further adipocyte differentiation. Dexras1 translocates to the plasma membrane upon insulin or IGF-1 treatment, for which the unique C-terminal domain (amino acids 223–276) is essential. Dexras1- dependent MAPK activation is selectively involved in the IGF-1 signaling, because another Ras protein, H-ras localized to the plasma membrane independently of insulin treatment. Moreover, neither epidermal growth factor nor other cell types shows Dexras1-dependent MAPK activation, indicating the importance of Dexras1 in IGF-1 signaling in adipogenesis. Dexras1 interacts with Shc and Raf, indicating that Dexras1-induced activation of MAPK is largely dependent on the Shc-Grb2-Raf complex.
    [Show full text]
  • Rasd1, a Small G Protein with a Big Role in the Hypothalamic Response to Neuronal Activation Michael P
    Greenwood et al. Molecular Brain (2016) 9:1 DOI 10.1186/s13041-015-0182-2 RESEARCH Open Access Rasd1, a small G protein with a big role in the hypothalamic response to neuronal activation Michael P. Greenwood1*, Mingkwan Greenwood1, Andre S. Mecawi2,3,4, José Antunes-Rodrigues2, Julian F. R. Paton5 and David Murphy1,3 Abstract Background: Rasd1 is a member of the Ras family of monomeric G proteins that was first identified as a dexamethasone inducible gene in the pituitary corticotroph cell line AtT20. Using microarrays we previously identified increased Rasd1 mRNA expression in the rat supraoptic nucleus (SON) and paraventricular nucleus (PVN) of the hypothalamus in response to increased plasma osmolality provoked by fluid deprivation and salt loading. RASD1 has been shown to inhibit adenylyl cyclase activity in vitro resulting in the inhibition of the cAMP-PKA-CREB signaling pathway. Therefore, we tested the hypothesis that RASD1 may inhibit cAMP stimulated gene expression in the brain. Results: We show that Rasd1 is expressed in vasopressin neurons of the PVN and SON, within which mRNA levels are induced by hyperosmotic cues. Dexamethasone treatment of AtT20 cells decreased forskolin stimulation of c-Fos, Nr4a1 and phosphorylated CREB expression, effects that were mimicked by overexpression of Rasd1, and inhibited by knockdown of Rasd1. These effects were dependent upon isoprenylation, as both farnesyltransferase inhibitor FTI-277 and CAAX box deletion prevented Rasd1 inhibition of cAMP-induced gene expression. Injection of lentiviral vector into rat SON expressing Rasd1 diminished, whereas CAAX mutant increased, cAMP inducible genes in response to osmotic stress. Conclusions: We have identified two mechanisms of Rasd1 induction in the hypothalamus, one by elevated glucocorticoids in response to stress, and one in response to increased plasma osmolality resulting from osmotic stress.
    [Show full text]
  • Tubular P53 Regulates Multiple Genes to Mediate AKI
    BASIC RESEARCH www.jasn.org Tubular p53 Regulates Multiple Genes to Mediate AKI † † † † † Dongshan Zhang,* Yu Liu,* Qingqing Wei, Yuqing Huo, Kebin Liu, Fuyou Liu,* and † Zheng Dong* *Departments of Emergency Medicine and Nephrology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China; and †Department of Cellular Biology and Anatomy, Vascular Biology Center and Department of Biochemistry and Molecular Biology, Georgia Regents University and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia ABSTRACT A pathogenic role of p53 in AKI was suggested a decade ago but remains controversial. Indeed, recent work indicates that inhibition of p53 protects against ischemic AKI in rats but exacerbates AKI in mice. One intriguing possibility is that p53 has cell type-specific roles in AKI. To determine the role of tubular p53, we generated two conditional gene knockout mouse models, in which p53 is specifically ablated from proximal tubules or other tubular segments, including distal tubules, loops of Henle, and medullary collecting ducts. Proximal tubule p53 knockout (PT-p53-KO) mice were resistant to ischemic and cisplatin nephrotoxic AKI, which was indicated by the analysis of renal function, histology, apoptosis, and inflammation. However, other tubular p53 knockout (OT-p53-KO) mice were sensitive to AKI. Mechanis- tically, AKI associated with the upregulation of several known p53 target genes, including Bax, p53- upregulated modulator of apoptosis-a, p21, and Siva, and this association was attenuated in PT-p53-KO mice. In global expression analysis, ischemic AKI induced 371 genes in wild-type kidney cortical tissues, but the induction of 31 of these genes was abrogated in PT-p53-KO tissues.
    [Show full text]