1
|
Kirchner S, Reuter S, Westphal A, Mrowka R. Decipher the complexity of cis-regulatory regions by a modified Cas9. PLoS One 2020; 15:e0235530. [PMID: 32614871 PMCID: PMC7332081 DOI: 10.1371/journal.pone.0235530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 06/18/2020] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Understanding complex mechanisms of human transcriptional regulation remains a major challenge. Classical reporter studies already enabled the discovery of cis-regulatory elements within the non-coding DNA; however, the influence of genomic context and potential interactions are still largely unknown. Using a modified Cas9 activation complex we explore the complexity of renin transcription in its native genomic context. METHODS With the help of genomic editing, we stably tagged the native renin on chromosome 1 with the firefly luciferase and stably integrated a programmable modified Cas9 based trans-activation complex (SAM-complex) by lentiviral transduction into human cells. By delivering five specific guide-RNA homologous to specific promoter regions of renin we were able to guide this SAM-complex to these regions of interest. We measured gene expression and generated and compared computational models. RESULTS SAM complexes induced activation of renin in our cells after renin specific guide-RNA had been provided. All possible combinations of the five guides were subjected to model analysis in linear models. Quantifying the prediction error and the calculation of an estimator of the relative quality of the statistical models for our given set of data revealed that a model incorporating interactions in the proximal promoter is the superior model for explanation of the data. CONCLUSION By applying our combined experimental and modelling approach we can show that interactions occur within the selected sequences of the proximal renin promoter region. This combined approach might potentially be useful to investigate other genomic regions. Our findings may help to better understand the transcriptional regulation of human renin.
Collapse
Affiliation(s)
- Steven Kirchner
- Experimental Nephrology Group, KIM III, Universitätsklinikum Jena, Jena, Germany
| | - Stefanie Reuter
- Experimental Nephrology Group, KIM III, Universitätsklinikum Jena, Jena, Germany
| | - Anika Westphal
- Experimental Nephrology Group, KIM III, Universitätsklinikum Jena, Jena, Germany
| | - Ralf Mrowka
- Experimental Nephrology Group, KIM III, Universitätsklinikum Jena, Jena, Germany
| |
Collapse
|
2
|
Lu KT, Keen HL, Weatherford ET, Sequeira-Lopez MLS, Gomez RA, Sigmund CD. Estrogen Receptor α Is Required for Maintaining Baseline Renin Expression. Hypertension 2016; 67:992-9. [PMID: 26928806 DOI: 10.1161/hypertensionaha.115.07082] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 02/08/2016] [Indexed: 01/08/2023]
Abstract
Enzymatic cleavage of angiotensinogen by renin represents the critical rate-limiting step in the production of angiotensin II, but the mechanisms regulating the initial expression of the renin gene remain incomplete. The purpose of this study is to unravel the molecular mechanism controlling renin expression. We identified a subset of nuclear receptors that exhibited an expression pattern similar to renin by reanalyzing a publicly available microarray data set. Expression of some of these nuclear receptors was similarly regulated as renin in response to physiological cues, which are known to regulate renin. Among these, only estrogen receptor α (ERα) and hepatic nuclear factor α have no known function in regulating renin expression. We determined that ERα is essential for the maintenance of renin expression by transfection of small interfering RNAs targeting Esr1, the gene encoding ERα, in renin-expressing As4.1 cells. We also observed that previously characterized negative regulators of renin expression, Nr2f2 and vitamin D receptor, exhibited elevated expression in response to ERα inhibition. Therefore, we tested whether ERα regulates renin expression through an interaction with Nr2f2 and vitamin D receptor. Renin expression did not return to baseline when we concurrently suppressed both Esr1 and Nr2f2 or Esr1 and vitamin D receptor mRNAs, strongly suggesting that Esr1 regulates renin expression independent of Nr2f2 and vitamin D receptor. ERα directly binds to the hormone response element within the renin enhancer region. We conclude that ERα is a previously unknown regulator of renin that directly binds to the renin enhancer hormone response element sequence and is critical in maintaining renin expression in renin-expressing As4.1 cells.
Collapse
Affiliation(s)
- Ko-Ting Lu
- From the Department of Pharmacology (K.-T.L., H.L.K., E.T.W., C.D.S.) and Center for Hypertension Research (C.D.S.), Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City; and Department of Pediatrics, University of Virginia, Charlottesville (M.L.S.S.-L., R.A.G.)
| | - Henry L Keen
- From the Department of Pharmacology (K.-T.L., H.L.K., E.T.W., C.D.S.) and Center for Hypertension Research (C.D.S.), Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City; and Department of Pediatrics, University of Virginia, Charlottesville (M.L.S.S.-L., R.A.G.)
| | - Eric T Weatherford
- From the Department of Pharmacology (K.-T.L., H.L.K., E.T.W., C.D.S.) and Center for Hypertension Research (C.D.S.), Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City; and Department of Pediatrics, University of Virginia, Charlottesville (M.L.S.S.-L., R.A.G.)
| | - Maria Luisa S Sequeira-Lopez
- From the Department of Pharmacology (K.-T.L., H.L.K., E.T.W., C.D.S.) and Center for Hypertension Research (C.D.S.), Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City; and Department of Pediatrics, University of Virginia, Charlottesville (M.L.S.S.-L., R.A.G.)
| | - R Ariel Gomez
- From the Department of Pharmacology (K.-T.L., H.L.K., E.T.W., C.D.S.) and Center for Hypertension Research (C.D.S.), Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City; and Department of Pediatrics, University of Virginia, Charlottesville (M.L.S.S.-L., R.A.G.)
| | - Curt D Sigmund
- From the Department of Pharmacology (K.-T.L., H.L.K., E.T.W., C.D.S.) and Center for Hypertension Research (C.D.S.), Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City; and Department of Pediatrics, University of Virginia, Charlottesville (M.L.S.S.-L., R.A.G.).
| |
Collapse
|
3
|
Herrmann S, Schnorr S, Ludwig A. HCN channels--modulators of cardiac and neuronal excitability. Int J Mol Sci 2015; 16:1429-47. [PMID: 25580535 PMCID: PMC4307311 DOI: 10.3390/ijms16011429] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 12/31/2014] [Indexed: 01/06/2023] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels comprise a family of cation channels activated by hyperpolarized membrane potentials and stimulated by intracellular cyclic nucleotides. The four members of this family, HCN1-4, show distinct biophysical properties which are most evident in the kinetics of activation and deactivation, the sensitivity towards cyclic nucleotides and the modulation by tyrosine phosphorylation. The four isoforms are differentially expressed in various excitable tissues. This review will mainly focus on recent insights into the functional role of the channels apart from their classic role as pacemakers. The importance of HCN channels in the cardiac ventricle and ventricular hypertrophy will be discussed. In addition, their functional significance in the peripheral nervous system and nociception will be examined. The data, which are mainly derived from studies using transgenic mice, suggest that HCN channels contribute significantly to cellular excitability in these tissues. Remarkably, the impact of the channels is clearly more pronounced in pathophysiological states including ventricular hypertrophy as well as neural inflammation and neuropathy suggesting that HCN channels may constitute promising drug targets in the treatment of these conditions. This perspective as well as the current therapeutic use of HCN blockers will also be addressed.
Collapse
Affiliation(s)
- Stefan Herrmann
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany.
| | - Sabine Schnorr
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany.
| | - Andreas Ludwig
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany.
| |
Collapse
|
4
|
Tanimoto K, Kanafusa S, Ushiki A, Matsuzaki H, Ishida J, Sugiyama F, Fukamizu A. A mouse renin distal enhancer is essential for blood pressure homeostasis in BAC-rescued renin-null mutant mice. J Recept Signal Transduct Res 2014; 34:401-9. [PMID: 24734888 DOI: 10.3109/10799893.2014.908917] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Renin is predominantly expressed in juxtaglomerular cells in the kidney and regulates blood pressure homeostasis. To examine possible in vivo functions of a mouse distal enhancer (mdE), we generated transgenic mice (TgM) carrying either wild-type or mdE-deficient renin BACs (bacterial artificial chromosome), integrated at the identical chromosomal site. In the kidneys of the TgM, the mdE contributed 80% to basal renin promoter activity. To test for possible physiological roles for the mdE, renin BAC transgenes were used to rescue the hypotensive renin-null mice. Interestingly, renal renin expression in the Tg(BAC):renin-null compound mice was indistinguishable between the wild-type and mutant BAC carriers. Surprisingly, however, the plasma renin activity and angiotensin I concentration in the mdE compound mutant mice were significantly lower than the same parameters in the control mice, and the mutants were consistently hypotensive, demonstrating that blood pressure homeostasis is regulated through transcriptional cis elements controlling renin activity.
Collapse
Affiliation(s)
- Keiji Tanimoto
- Faculty of Life and Environmental Sciences, University of Tsukuba , Tsukuba, Ibaraki , Japan
| | | | | | | | | | | | | |
Collapse
|
5
|
Renin and the IGFII/M6P receptor system in cardiac biology. ScientificWorldJournal 2013; 2013:260298. [PMID: 24288471 PMCID: PMC3826467 DOI: 10.1155/2013/260298] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 08/20/2013] [Indexed: 11/17/2022] Open
Abstract
Nonenzymatic cardiac activities of renin are well described during the last years and contribute either to cardiac-specific effects of the renin-angiotensin-aldosterone-system (RAAS) or to the pharmacological effects of RAAS inhibition. The interaction of renin with insulin-like growth factor II/mannose-6-phosphate (IGFII/M6P) receptors participates in nonclassical renin effects and contributes to cardiac remodelling caused by RAAS activation. The current findings suggest an important role for renin IGFII/M6P receptor interaction in cardiac adaptation to stress and support the idea that excessive accumulation of renin during inhibition of RAAS directly contributes to blood pressure-independent effects of these pharmacological interventions. It becomes a challenge for future studies focussing on chronic hypertension or myocardial infarction to comprise regulatory adaptations of the kidney, the main source of plasma renin and prorenin, because they directly contribute to key steps in regulation of cardiac (mal)adaptation via IGFII/M6P receptors. This receptor system is part of peptide/receptor interactions that modifies and possibly limits adverse remodelling effects caused by angiotensin II. Evaluation of interactions of renin with other pro-hypertrophic agonists is required to decide whether this receptor may become a target of pharmacological intervention.
Collapse
|
6
|
Glenn ST, Jones CA, Gross KW, Pan L. Control of renin [corrected] gene expression. Pflugers Arch 2012; 465:13-21. [PMID: 22576577 DOI: 10.1007/s00424-012-1110-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 04/17/2012] [Accepted: 04/19/2012] [Indexed: 10/28/2022]
Abstract
Renin, as part of the renin-angiotensin system, plays a critical role in the regulation of blood pressure, electrolyte homeostasis, mammalian renal development, and progression of fibrotic/hypertrophic diseases. Renin gene transcription is subject to complex developmental and tissue-specific regulation. Initial studies using the mouse As4.1 cell line, which has many characteristics of the renin-expressing juxtaglomerular cells of the kidney, have identified a proximal promoter region (-197 to -50 bp) and an enhancer (-2,866 to -2,625 bp) upstream of the Ren-1(c) gene, which are critical for renin gene expression. The proximal promoter region contains several transcription factor binding sites including a binding site for the products of the developmental control genes Hox. The enhancer consists of at least 11 transcription factor binding sites and is responsive to various signal transduction pathways including cAMP, retinoic acid, endothelin-1, and cytokines, all of which are known to alter renin mRNA levels. Furthermore, in vivo models have validated several of these key components found within the proximal promoter region and the enhancer as well as other key sites necessary for renin gene transcription.
Collapse
Affiliation(s)
- Sean T Glenn
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263-0001, USA.
| | | | | | | |
Collapse
|
7
|
Weatherford ET, Liu X, Sigmund CD. Regulation of renin expression by the orphan nuclear receptors Nr2f2 and Nr2f6. Am J Physiol Renal Physiol 2012; 302:F1025-33. [PMID: 22278040 DOI: 10.1152/ajprenal.00362.2011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Understanding the transcriptional mechanisms of renin expression is key to understanding the regulation of the renin-angiotensin system. We previously identified the nuclear receptors RAR/RXR and Nr2f6 (EAR2) as positive and negative transcriptional regulators of renin expression, respectively (Liu X, Huang X, Sigmund CD. Circ Res 92: 1033-1040, 2003). Both mediate their effects through a hormone response element (HRE) within the renin enhancer. Here, we determined whether another nuclear receptor, Nr2f2 (Coup-TFII, Arp-1), identified in a screen of proteins that bind the HRE, also regulates renin expression. Luciferase assays indicate that Nr2f2 negatively regulates the renin promoter more potently than Nr2f6. Gel-shift and chromatin immunoprecipitation (ChIP) indicate that Nr2f2 and Nr2f6 can bind directly to the renin enhancer through the HRE. Surprisingly, baseline expression of endogenous renin was not effected when Nr2f2 was knocked down in As4.1 cells, whereas knockdown of Nr2f6 increased renin expression twofold. Interestingly, however, knockdown of Nr2f2 augmented the induction of renin expression caused by retinoic acid. These data indicate that both Nr2f6 and Nr2f2 can negatively regulate the renin promoter, under baseline conditions and in response to physiological queues, respectively. Therefore, Nr2f2 may require an initiating signal that results in a change at the chromatin level or activation of another transcription factor to exert its effects. We conclude that both Nr2f2 and Nr2f6 negatively regulate renin promoter activity, but may do so by divergent mechanisms.
Collapse
Affiliation(s)
- Eric T Weatherford
- Department of Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | | | | |
Collapse
|
8
|
Tan JJ, Ong SA, Chen KS. Rasd1 interacts with Ear2 (Nr2f6) to regulate renin transcription. BMC Mol Biol 2011; 12:4. [PMID: 21247419 PMCID: PMC3036621 DOI: 10.1186/1471-2199-12-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Accepted: 01/19/2011] [Indexed: 01/19/2023] Open
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.
Collapse
Affiliation(s)
- Jen Jen Tan
- School of Biological Sciences, Department of Genomics and Genetics, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore
| | | | | |
Collapse
|
9
|
Castrop H, Höcherl K, Kurtz A, Schweda F, Todorov V, Wagner C. Physiology of Kidney Renin. Physiol Rev 2010; 90:607-73. [PMID: 20393195 DOI: 10.1152/physrev.00011.2009] [Citation(s) in RCA: 189] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The protease renin is the key enzyme of the renin-angiotensin-aldosterone cascade, which is relevant under both physiological and pathophysiological settings. The kidney is the only organ capable of releasing enzymatically active renin. Although the characteristic juxtaglomerular position is the best known site of renin generation, renin-producing cells in the kidney can vary in number and localization. (Pro)renin gene transcription in these cells is controlled by a number of transcription factors, among which CREB is the best characterized. Pro-renin is stored in vesicles, activated to renin, and then released upon demand. The release of renin is under the control of the cAMP (stimulatory) and Ca2+(inhibitory) signaling pathways. Meanwhile, a great number of intrarenally generated or systemically acting factors have been identified that control the renin secretion directly at the level of renin-producing cells, by activating either of the signaling pathways mentioned above. The broad spectrum of biological actions of (pro)renin is mediated by receptors for (pro)renin, angiotensin II and angiotensin-( 1 – 7 ).
Collapse
Affiliation(s)
- Hayo Castrop
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Klaus Höcherl
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Armin Kurtz
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Frank Schweda
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Vladimir Todorov
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Charlotte Wagner
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
10
|
Sharma V, McNeill JH. To scale or not to scale: the principles of dose extrapolation. Br J Pharmacol 2009; 157:907-21. [PMID: 19508398 DOI: 10.1111/j.1476-5381.2009.00267.x] [Citation(s) in RCA: 377] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The principles of inter-species dose extrapolation are poorly understood and applied. We provide an overview of the principles underlying dose scaling for size and dose adjustment for size-independent differences. Scaling of a dose is required in three main situations: the anticipation of first-in-human doses for clinical trials, dose extrapolation in veterinary practice and dose extrapolation for experimental purposes. Each of these situations is discussed. Allometric scaling of drug doses is commonly used for practical reasons, but can be more accurate when one takes into account species differences in pharmacokinetic parameters (clearance, volume of distribution). Simple scaling of drug doses can be misleading for some drugs; correction for protein binding, physicochemical properties of the drug or species differences in physiological time can improve scaling. However, differences in drug transport and metabolism, and in the dose-response relationship, can override the effect of size alone. For this reason, a range of modelling approaches have been developed, which combine in silico simulations with data obtained in vitro and/or in vivo. Drugs that are unlikely to be amenable to simple allometric scaling of their clearance or dose include drugs that are highly protein-bound, drugs that undergo extensive metabolism and active transport, drugs that undergo significant biliary excretion (MW > 500, ampiphilic, conjugated), drugs whose targets are subject to inter-species differences in expression, affinity and distribution and drugs that undergo extensive renal secretion. In addition to inter-species dose extrapolation, we provide an overview of dose extrapolation within species, discussing drug dosing in paediatrics and in the elderly.
Collapse
Affiliation(s)
- Vijay Sharma
- Division of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | | |
Collapse
|
11
|
Glenn ST, Jones CA, Pan L, Gross KW. In vivo analysis of key elements within the renin regulatory region. Physiol Genomics 2008; 35:243-53. [PMID: 18780761 DOI: 10.1152/physiolgenomics.00017.2008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Renin is responsible for initiating the enzymatic cascade that results in the production of angiotensin II, the major effector molecule of the renin-angiotensin system (RAS). Extensive information on the regulatory region of the renin gene has been derived by transient transfection studies in vitro, particularly using the As4.1 cell line. To verify key factors within the regulatory region of renin in vivo, homologous recombination was used to introduce a green fluorescent protein (GFP) cassette into exon one of the renin gene contained within a 240 kb bacterial artificial chromosome (BAC) to create a construct that has GFP expression controlled by the renin regulatory region (RenGFP BAC). Within the regulatory region of the RenGFP BAC construct we independently deleted the enhancer, as well as mutated the HOX-PBX site within the proximal promoter element. Transgenic lines were generated for each of these BAC constructs and GFP expression was analyzed throughout a spectrum of tissues positive for renin expression including the kidney, adrenal gland, gonadal artery, and submandibular gland. The results described within this manuscript support the interpretation that the renin enhancer is critical for regulating baseline expression where as the Hox/Pbx site is important for the tissue specificity of renin expression.
Collapse
Affiliation(s)
- Sean T Glenn
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, New York 14263-0001, USA
| | | | | | | |
Collapse
|
12
|
Nakane M, Ma J, Ruan X, Kroeger PE, Wu-Wong R. Mechanistic analysis of VDR-mediated renin suppression. Nephron Clin Pract 2007; 107:p35-44. [PMID: 17687191 DOI: 10.1159/000106792] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2006] [Accepted: 05/08/2007] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The vitamin D receptor (VDR) is involved in the regulation of renin expression and vitamin D analogs down-regulated renin mRNA expression in As4.1 cells. METHODS Microarray analysis was used to assess the VDR-mediated gene expression profile in As4.1 cells treated with paricalcitol, followed by real-time RT-PCR. Mechanistic analyses were done using siRNA, electrophoretic mobility shift assay (EMSA) and Western blotting. RESULTS Microarray analysis shows that 709 target genes were affected by paricalcitol with 286 up- and 423 downregulated. A number of major pathways were impacted including transcription factors. Real-time RT-PCR confirmed the microarray results. Treatment of the cells with siRNA against nuclear receptor co-repressor (NCOR1) eliminated VDR-mediated renin suppression. Using EMSA, paricalcitol treatment reduced the level of protein complex binding to the cyclic AMP-responsive element (CRE)-like domain in the renin distal enhancer region. VDR, CRE-binding protein (CREB1) and NCOR1 were identified in the complex binding to the CRE-like domain by Western blot in the paricalcitol-treated cells. Paricalcitol treatment resulted in an increase in the VDR level, but no significant change in the CREB1 and NCOR1 levels. CONCLUSION These results suggest that VDR-mediated renin suppression likely acts through a transcriptional regulatory complex including CREB1, NCOR1 and VDR that binds to the CRE-like domain in the renin enhancer region.
Collapse
|
13
|
Mrowka R, Steege A, Kaps C, Herzel H, Thiele BJ, Persson PB, Blüthgen N. Dissecting the action of an evolutionary conserved non-coding region on renin promoter activity. Nucleic Acids Res 2007; 35:5120-9. [PMID: 17660193 PMCID: PMC1976450 DOI: 10.1093/nar/gkm535] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Elucidating the mechanisms of the human transcriptional regulatory network is a major challenge of the post-genomic era. One important aspect is the identification and functional analysis of regulatory elements in non-coding DNA. Genomic sequence comparisons between related species can guide the discovery of cis-regulatory sequences. Using this technique, we identify a conserved region CNSmd of ∼775 bp in size, ∼14 kb upstream of the renin gene. Renin plays a pivotal role for mammalian blood pressure regulation and electrolyte balance. To analyse the cis-regulatory role of this region in detail, we perform 132 combinatorial reporter gene assays in an in vitro Calu-6 cell line model. To dissect the role of individual subregions, we fit several mathematical models to the experimental data. We show that a multiplicative switch model fits best the experimental data and that one subregion has a dominant effect on promoter activity. Mapping of the sub-sequences on phylogenetic conservation data reveals that the dominant regulatory region is the one with the highest multi-species conservation score.
Collapse
Affiliation(s)
- Ralf Mrowka
- Institute for Physiology, Systems Biology-Computational Physiology, Charité Universitätsmedizin, Berlin.
| | | | | | | | | | | | | |
Collapse
|
14
|
Itani HA, Liu X, Pratt JH, Sigmund CD. Functional characterization of polymorphisms in the kidney enhancer of the human renin gene. Endocrinology 2007; 148:1424-30. [PMID: 17158202 DOI: 10.1210/en.2006-1381] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The renin gene is regulated by an enhancer located 2.6 kb upstream of the transcription start site in the mouse and 11 kb upstream in humans. Despite extensive sequence conservation, the mouse renin enhancer is transcriptionally more active than the human renin enhancer. We report that the mechanism accounting for this is a result of sequence variation in the promoter proximal half-site of a retinoic-acid response element present in the enhancer. This sequence difference also prompted us to search for naturally occurring polymorphisms in the renin enhancer among normal and hypertensive human subjects. We sequenced the kidney enhancer from 90 samples derived from the Coriell Polymorphism Discovery Resource and 95 severely hypertensive Caucasian and African-American individuals. A single relatively frequent polymorphism (7, 2, and 7%, respectively in the Coriell, African-American, and Caucasian) was identified in the enhancer, one nucleotide downstream of the promoter distal half-site of the retinoic-acid response element. This variant was transcriptionally silent in transfection assays performed in renin-expressing As4.1 cells, a model of renal juxtaglomerular cells. A singleton polymorphism in the promoter was also identified in a single African-American individual. This polymorphism was located between binding sites for CBF1 and homeobox D10 but was also transcriptionally silent either in the presence or absence of the enhancer. Our study demonstrates the presence of silent polymorphisms in the renin promoter and enhancer, thus underscoring the critical importance of performing functional analyses before initiating expensive clinical studies seeking association between polymorphisms and complex diseases such as hypertension.
Collapse
Affiliation(s)
- Hana A Itani
- Molecular and Cellular Biology Interdisciplinary Graduate Program, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | |
Collapse
|
15
|
Adams DJ, Head GA, Markus MA, Lovicu FJ, van der Weyden L, Ko¨ntgen F, Arends MJ, Thiru S, Mayorov DN, Morris BJ. Renin Enhancer Is Critical for Control of Renin Gene Expression and Cardiovascular Function. J Biol Chem 2006. [DOI: 10.1016/s0021-9258(19)84090-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
16
|
Zhou X, Davis DR, Sigmund CD. The human renin kidney enhancer is required to maintain base-line renin expression but is dispensable for tissue-specific, cell-specific, and regulated expression. J Biol Chem 2006; 281:35296-304. [PMID: 16990260 DOI: 10.1074/jbc.m608055200] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Renin is the rate-limiting enzyme in the renin-angiotensin system and thus dictates the level of the pressor hormone angiotensin-II. The classical site of renin expression and secretion is the renal juxtaglomerular cell, where its expression is tightly regulated by physiological cues. An evolutionarily conserved transcriptional enhancer located 11 kb upstream of the human RENIN gene has been reported to markedly enhance transcription in renin expressing cells in vitro. However, its importance in vivo remains unclear. We tested whether this enhancer is required for appropriate tissue- and cell-specific expression, or for physiological regulation of the human RENIN gene. To accomplish this, we used a retrofitting technique employing homologous recombination in bacteria to delete the enhancer from a 160-kb P1-artificial chromosome containing human RENIN, two upstream genes and one downstream gene, and then generated two lines of transgenic mice. We previously showed that human renin expression in transgenic mice containing the wild type construct is tightly regulated as is expression of the linked genes. Deletion of the enhancer had no effect on tissue-specific expression of human RENIN, but using the downstream gene as an internal control, found that human RENIN mRNA levels were 3-10-fold decreased compared with constructs containing the enhancer. Despite this decrease in expression, renin protein remained localized to renal juxtaglomerular cells and was appropriately regulated by cues that either increase or decrease expression of renin. Our results suggest that sequences other than the enhancer may be necessary for tissue-specific, cell-specific, and regulated expression of human RENIN.
Collapse
Affiliation(s)
- Xiyou Zhou
- Molecular and Cellular Biology Graduate Program, Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, USA
| | | | | |
Collapse
|
17
|
Adams DJ, Head GA, Markus MA, Lovicu FJ, van der Weyden L, Köntgen F, Arends MJ, Thiru S, Mayorov DN, Morris BJ. Renin enhancer is critical for control of renin gene expression and cardiovascular function. J Biol Chem 2006; 281:31753-61. [PMID: 16895910 DOI: 10.1074/jbc.m605720200] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The important cardiovascular regulator renin contains a strong in vitro enhancer 2.7 kb upstream of its gene. Here we tested the in vivo role of the mouse Ren-1c enhancer. In renin-expressing As4.1 cells stably transfected with Ren-1c promoter with or without enhancer, expression of linked beta-geo reporter, stable expression, and colony formation were dependent on the presence of the enhancer. We then generated mice carrying a targeted deletion of the enhancer (REKO mice) and found marked depletion of renin in renal juxtaglomerular and submandibular ductal cells, as well as hyperplasia of macula densa cells. Plasma creatinine was increased, but electrolytes were normal. Male REKO mice implanted with telemetry devices had 9 +/- 1 mm Hg lower mean arterial pressure (p < 0.001), which was partly normalized by a high NaCl diet. Locomotor activity was lower, and baroreflex sensitivity was normal. Markedly reduced mean arterial pressure variability in the midfrequency band indicated a contribution of reduced sympathetic vasomotor tone to the hypotension. In conclusion, the renin enhancer is critical for renin gene expression and physiological sequelae, including response to alteration in salt intake. The REKO mouse may be useful as a low renin expression model.
Collapse
Affiliation(s)
- David J Adams
- School of Medical Sciences and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Todorov VT, Völkl S, Friedrich J, Kunz-Schughart LA, Hehlgans T, Vermeulen L, Haegeman G, Schmitz ML, Kurtz A. Role of CREB1 and NFκB-p65 in the Down-regulation of Renin Gene Expression by Tumor Necrosis Factor α. J Biol Chem 2005; 280:24356-62. [PMID: 15857826 DOI: 10.1074/jbc.m502968200] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Tumor necrosis factor-alpha (TNFalpha) is a potent inhibitor of renin gene expression in renal juxtaglomerular cells. We have found that TNFalpha suppresses renin transcription via transcription factor NFkappaB, which targets a cAMP responsive element (CRE) in the renin promoter. Here we aimed to further clarify the role of NFkappaB and the canonical CRE-binding proteins of the CRE-binding protein/activating transcription factor (CREB/ATF) family in the inhibition of renin gene expression by TNFalpha in the juxtaglomerular cell line As4.1. TNFalpha caused a moderate decrease in the binding of CREB1 to its cognate CRE DNA binding site. On the other hand, NFkappaB-p65 transcriptional activity was substantially reduced by TNFalpha, which targeted a trans-activation domain at the very C terminus of the p65 molecule. Our results suggest that TNFalpha inhibits renin gene expression by decreasing the transactivating capacity of NFkappaB-p65 and partially by attenuating CREB1 binding to CRE.
Collapse
Affiliation(s)
- Vladimir T Todorov
- Institute of Physiology, Institute of Pathology, and Department of Immunology, Regensburg University, D-93040 Regensburg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Prandini MH, Dreher I, Bouillot S, Benkerri S, Moll T, Huber P. The human VE-cadherin promoter is subjected to organ-specific regulation and is activated in tumour angiogenesis. Oncogene 2005; 24:2992-3001. [PMID: 15735710 PMCID: PMC2800996 DOI: 10.1038/sj.onc.1208483] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Vascular endothelial (VE)-cadherin is exclusively expressed at interendothelial junctions of normal and tumour vessels. In this report, we characterized the transcriptional activity of the human VE-cadherin promoter. Transient transfection assays revealed that sequences at positions --1135/-744 and -166/-5 base pairs are critical for promoter activity in endothelial cells. We show that specific sequences in the proximal region interact with Ets and Sp1 family members. Transgenic mice were created and the human VE-cadherin promoter was able to confer correct temporal and spatial expression on the LacZ gene in embryos. In adults, the transgene was specifically and strongly expressed in the lung, heart, ovary, spleen and kidney glomeruli, whereas expression was weak or absent in the vasculature of other organs, including the brain, thymus, liver and skeletal muscle. Neovessels in tumour grafts and Matrigel implants harboured strong stainings, indicating that promoter activity is enhanced in angiogenic situations. Furthermore, Matrigel and transfection assays showed that VE-cadherin promoter is subjected to bFGF induction. Transgene expression was also noticed in extravascular sites of the central nervous system, suggesting that silencer elements may be located elsewhere in the gene. These results are a first step towards addressing the organ- and tumour-specific regulation of the VE-cadherin gene.
Collapse
Affiliation(s)
- Marie-Hélène Prandini
- Laboratoire de développement et vieillissement de l'endothélium
INSERM : EMI0219CEA : DSV/IRTSVUniversité Joseph Fourier - Grenoble IFR
| | | | - Stéphanie Bouillot
- Laboratoire de développement et vieillissement de l'endothélium
INSERM : EMI0219CEA : DSV/IRTSVUniversité Joseph Fourier - Grenoble IFR
| | - Souhila Benkerri
- Laboratoire de développement et vieillissement de l'endothélium
INSERM : EMI0219CEA : DSV/IRTSVUniversité Joseph Fourier - Grenoble IFR
| | | | - Philippe Huber
- Laboratoire de développement et vieillissement de l'endothélium
INSERM : EMI0219CEA : DSV/IRTSVUniversité Joseph Fourier - Grenoble IFR
- * Correspondence should be adressed to: Philippe Huber
| |
Collapse
|
20
|
Tamura K, Chen YE, Tanaka Y, Sakai M, Tsurumi Y, Koide Y, Kihara M, Pratt RE, Horiuchi M, Umemura S, Dzau VJ. Nuclear receptor LXRalpha is involved in cAMP-mediated human renin gene expression. Mol Cell Endocrinol 2004; 224:11-20. [PMID: 15353176 DOI: 10.1016/j.mce.2004.07.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2004] [Revised: 07/01/2004] [Accepted: 07/14/2004] [Indexed: 01/17/2023]
Abstract
The cAMP-signaling pathway plays a crucial role in the regulation of the renin gene, but the mechanism involved remains poorly understood. We have focused our studies of renin gene regulation on the unique cAMP responsive element (huREN/CNRE, -135 to -107) in the human renin promoter. We have cloned a protein that binds to this unique CNRE and demonstrated that this protein is liver X receptor-alpha (LXRalpha), a transcriptional factor of the nuclear receptor family. Transient expression of LXRalpha in human renin-producing Calu-6 cells increased cAMP inducibility of human renin promoter. Similarly, LXRalpha-stably transfected Calu-6 cells exhibited increased cAMP inducibility of renin promoter as well as the endogenous renin gene. Site-directed mutation of huREN/CNRE, which disrupted LXRalpha binding, decreased cAMP-induced transcriptional activity of human renin promoter. Furthermore, we demonstrated that the binding of LXRalpha derived from human juxtaglomerular cells, the main production site of renin in the kidney, to the huREN/CNRE in vivo. These results suggest that LXRalpha plays an important role in the cAMP-mediated regulation of human renin gene transcription by binding to CNRE.
Collapse
Affiliation(s)
- Kouichi Tamura
- Department of Medicine II, Yokohama City University School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Pan L, Wang Y, Jones CA, Glenn ST, Baumann H, Gross KW. Enhancer-dependent inhibition of mouse renin transcription by inflammatory cytokines. Am J Physiol Renal Physiol 2004; 288:F117-24. [PMID: 15367390 DOI: 10.1152/ajprenal.00333.2003] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Inflammatory cytokines have been shown to inhibit renin gene expression in the kidney in vivo and the kidney tumor-derived As4.1 cell line. In this report, we show that cytokines oncostatin M (OSM), IL-6, and IL-1beta inhibit transcriptional activity associated with 4.1 kb of the mouse renin 5'-flanking sequence in As4.1 cells. The 242-bp enhancer (-2866 to -2625 bp) is sufficient to mediate the observed inhibitory effects. Sequences within the enhancer required for inhibition by each of these cytokines have been determined by deletional and mutational analysis. Results indicate that a 39-bp region (CEC) containing a cAMP-responsive element, an E-box, and a steroid receptor-binding site, previously identified as the most critical elements for enhancer activity, is sufficient for the inhibition induced by IL-1beta. However, mutation of each of the three component sites does not abolish the inhibition by IL-1beta, suggesting that the target(s) of cytokine action may not be the transcription factors binding directly to these sites. This CEC region is also critical, but not sufficient, for the inhibition mediated by OSM and IL-6. These data suggest that the direct target of the associated cytokines may be coactivators interacting with transcription factors binding at the enhancer. Finally, we show that OSM treatment caused a 17-fold increase in promoter activity when only 2,625 bp of the Ren-1(c) flanking sequence were tested, in which the enhancer is not present. Three regions including -2625 to -1217 bp, the HOX.PBX binding site at -60 bp, and -59 to +6 bp have been found to contribute to this induction.
Collapse
Affiliation(s)
- Li Pan
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Elm and Carlton St., Buffalo, NY 14263-0001, USA
| | | | | | | | | | | |
Collapse
|
22
|
Nistala R, Zhang X, Sigmund CD. Differential expression of the closely linked KISS1, REN, and FLJ10761 genes in transgenic mice. Physiol Genomics 2004; 17:4-10. [PMID: 14709678 DOI: 10.1152/physiolgenomics.00205.2003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We previously reported the development and characterization of transgenic mice containing a large 160-kb P1 artificial chromosome (PAC) encompassing the renin (REN) locus from human chromosome 1. Here we demonstrate that PAC160 not only encodes REN, but also complete copies of the next upstream (KISS1) and downstream ( FLJ10761 ) gene along human chromosome 1. Incomplete copies of the second upstream (PEPP3) and downstream (SOX13) genes are also present. The gene order PEPP3-KISS1-REN-FLJ10761-SOX13 is conserved in mice containing either one or two copies of the REN locus. Despite the close localization of KISS1, REN, and FLJ10761 , they each exhibit distinct, yet overlapping tissue-specific expression profiles in humans. The tissue-specific expression patterns of REN and FLJ10761 were retained in transgenic mice containing PAC160. Expression of REN and FLJ10761 were also proportional to copy number. Expression of KISS1 in PAC160 mice showed both similarities and differences to humans. These data suggest that expression of gene blocks encoded on large genomic clones are retained when the clones are used to generate transgenic mice. Genomic elements which act to insulate genes from their neighbors are also apparently retained.
Collapse
Affiliation(s)
- Ravi Nistala
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, USA
| | | | | |
Collapse
|
23
|
Pan L, Jones CA, Glenn ST, Gross KW. Identification of a novel region in the proximal promoter of the mouse renin gene critical for expression. Am J Physiol Renal Physiol 2004; 286:F1107-15. [PMID: 14761860 DOI: 10.1152/ajprenal.00319.2003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
An enhancer at -2.6 kb and a HOX.PBX-binding site at -60 bp have been demonstrated to be critical to expression of the mouse renin gene (Ren-1(c)) in As4.1 cells. In this report, we show that a region (-197 to -70) immediately 5' to the HOX.PBX-binding site is also critical for Ren-1(c) expression. Deletion of this region in a construct containing 4.1 kb of the Ren-1(c) 5'-flanking sequence resulted in a 99% reduction in Ren-1(c) promoter activity in As4.1 cells, suggesting the pivotal role for the region in the regulation of the mouse renin gene. Electrophoretic mobility shift and supershift assays have identified two nuclear factor I-binding sites and a Sp1/Sp3-binding site within the distal portion of the region (-197 to -103). Mutation of these three sites caused a 90% decrease in Ren-1(c) promoter activity. Mutational analysis and electrophoretic mobility shift assays have also identified three additional transcription factor-binding sites within the region from -103 to -69, each of which contributes to high-level expression of Ren-1(c) in As4.1 cells. Finally, we have shown that the Ren-1(c) enhancer is the target for endothelin-1 (ET-1)-induced inhibition of Ren-1(c) expression and the transcription factor-binding sites in the proximal promoter are required for the maximal ET-1 inhibitory effect.
Collapse
Affiliation(s)
- Li Pan
- Dept. of Molecular and Cellular Biology, Roswell Park Cancer Institute, Elm and Carlton Sts., Buffalo, NY 14263-0001, USA
| | | | | | | |
Collapse
|
24
|
Persson PB, Skalweit A, Mrowka R, Thiele BJ. Control of renin synthesis. Am J Physiol Regul Integr Comp Physiol 2003; 285:R491-7. [PMID: 12909573 DOI: 10.1152/ajpregu.00101.2003] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Studies published recently have considerably enhanced our understanding of the mechanisms controlling renin production. With regard to the control of renin transcription, two enhancer regions have been identified that markedly augment renin synthesis in cell lines. In the absence of this enhancer activity, the basic promoter of the renin gene increases transcription only two- to threefold. The location of one (Jones CA, Sigmund CD, McGowan RA, Kane-Haas CM, and Gross KW. Mol Endocrinol 4: 375-383, 1990) transcription enhancer in the mouse gene is at about -2.7 kb and in humans at roughly -11 kb. A second important region has been identified in a chorionic cell line to be located approximately 5 kb upstream of the transcription start site in humans. Another potentially important regulatory region may lie within approximately 3.9 kb upstream of the -11 kb enhancer, as suggested by several conserved sequences among species in this region. In addition to the control of renin transcription, it seems that renin translation and the stability of renin mRNA are also effectively regulated. This occurs via the 3'-untranslated region, to which several proteins can bind. The binding proteins were identified as hnRNP K and E1, dynamin, nucleolin, MINT homologous protein, and Y-Box 1.
Collapse
|
25
|
Mrowka R, Steinhage K, Patzak A, Persson PB. An evolutionary approach for identifying potential transcription factor binding sites: the renin gene as an example. Am J Physiol Regul Integr Comp Physiol 2003; 284:R1147-50. [PMID: 12626372 DOI: 10.1152/ajpregu.00448.2002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Evolutionary pressure has resulted in the conservation of certain nucleotide sequences. These conserved regions are potentially important for certain functions. Here we give an example of a comparison between noncoding sequences combined with other independent database information to shed light onto the regulation of the renin gene, a gene that has great importance for cardiovascular and renal homeostasis. To combine the information regarding conservation and weight matrices of transcription factor (TF) binding sites, an algorithm was developed (TFprofile). Notably, a local peak in the resulting binding profile coincides with a previously experimentally identified regulatory region for the renin gene. The existence of further peaks in the binding profile in the conserved 3.9-kb-long hRENc DNA block upstream of the renin gene suggests additional regions of potential importance for gene regulation. The algorithm TFprofile may be used to integrate information on cross-species evolutionary conservation and aspects of TF binding characteristics to provide putative regulatory DNA regions for experimental verification.
Collapse
Affiliation(s)
- Ralf Mrowka
- Johannes-Müller-Institut für Physiologie, Charité, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany.
| | | | | | | |
Collapse
|
26
|
Pan L, Glenn ST, Jones CA, Gronostajski RM, Gross KW. Regulation of renin enhancer activity by nuclear factor I and Sp1/Sp3. BIOCHIMICA ET BIOPHYSICA ACTA 2003; 1625:280-90. [PMID: 12591615 DOI: 10.1016/s0167-4781(03)00016-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Transcription of the mouse Ren-1(c) gene in kidney tumor-derived As4.1 cells, which express high levels of renin mRNA, is dependent on a proximal promoter element and a 242-bp enhancer region located 2.6 kb upstream of the transcription start site. We showed previously that the enhancer contains a cAMP responsive element (CRE) and an E-box. Mutation of either element resulted in almost complete loss of the Ren-1(c) expression. In this report we show that there are additional transcription factor-binding sites within the Ren-1(c) enhancer contributing to the enhancer activity. Electrophoretic mobility shift and supershift assays have identified four nuclear factor I (NFI)-binding sites, an Sp1/Sp3 site and an unidentified transcription factor-binding site (Ei) located upstream of the CRE and E-box. Mutation of the Sp1/Sp3 site or Ei reduced Ren-1(c) expression by 40% or 30%, respectively, while mutations of four NFI-binding sites resulted in an 89% decrease in expression. Thus, these protein-DNA interaction sites are essential for transcription of mouse renin genes. There are four homologous NFI genes (NFI-A, -B, -C and -X) in vertebrates and multiple alternatively spliced isoforms from each gene. Real-time reverse transcriptase-polymerase chain reaction (RT-PCR) assays have demonstrated that NFI-X is the predominant NFI mRNA expressed in As4.1 cells. Direct study of involvement of NFI-X in regulation of renin genes is underway.
Collapse
Affiliation(s)
- Li Pan
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263-0001, USA
| | | | | | | | | |
Collapse
|
27
|
Ryan MJ, Liu B, Herbowy MT, Gross KW, Hajduczok G. Intercellular communication between renin expressing As4.1 cells, endothelial cells and smooth muscle cells. Life Sci 2003; 72:1289-301. [PMID: 12570929 DOI: 10.1016/s0024-3205(02)02376-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Angiotensin II (AII) regulation of renin production by the juxtaglomerular (JG) cells of the kidney is commonly thought to occur through a direct feedback mechanism. However, recent evidence suggests that other cells in the vicinity may indirectly mediate AII's effect on renin production. Therefore we investigated whether an in vitro model of JG cells (As4.1) could have intercellular communication with endothelial or smooth muscle cells, which are in proximity to JG cells in vivo. 6-carboxyfluorescein was introduced to individual bovine aortic endothelial cells in co-culture with As4.1 cells. Coupling was observed 84% of the time at resting membrane potential and was attenuated by membrane depolarization or octanol (1 mM). Calcein green transfer between human aortic smooth muscle and As4.1 cells occurred 82% of the time and was inhibited by octanol. Expression of connexin 37, 40, 43, and 45 were detected in As4.1 cells using RT-PCR. Stimulation of As4.1 cells by AII failed to alter [Ca(2+)](i) or renin mRNA levels. These findings support the existence of gap junctions between renin producing cells and other cell types of the JG region. Moreover the lack of effect by AII suggest that feedback regulation of renin by AII may be due in part to intercellular communication with cells in proximity to JG cells.
Collapse
Affiliation(s)
- Michael J Ryan
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY 14214, USA.
| | | | | | | | | |
Collapse
|
28
|
Fuchs S, Philippe J, Germain S, Mathieu F, Jeunemaitre X, Corvol P, Pinet F. Functionality of two new polymorphisms in the human renin gene enhancer region. J Hypertens 2002; 20:2391-8. [PMID: 12473863 DOI: 10.1097/00004872-200212000-00018] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND The production of renin, which catalyses the rate-limiting step of the renin-angiotensin system, is strongly stimulated by a 225 bp enhancer element in the distal region of the promoter of the human gene (-5777 to -5552). OBJECTIVE To demonstrate the major role played by this enhancer in decoy experiments, to identify variants in this region, and to determine their effects on renin gene transcription. METHODS AND RESULTS We used this element as a decoy for transcription factors in human choriodecidual cells. The activity of the renin gene promoter was inhibited by 95% in the presence of this 225 bp enhancer element. This confirmation of the key role of this element suggested that changes in this region would be likely to affect renin gene expression. We therefore sequenced 70 genomic DNAs to identify variations in this region. We identified two new single nucleotide polymorphisms (SNPs) downstream from the 225 bp enhancer element at positions -5434 and -5312. We transfected choriodecidual cells with the four variants and found that a 592 bp region (-5870 to -5312) including the 225 bp element and the two SNPs had stronger enhancer activity than the 225 bp element alone, and that levels of transcription were 45% greater with the -5312T variant than with the -5312C variant, whereas none of the -5434 variants had an effect on renin transcription. Cis-regulatory elements close to the -5312 variant were identified in gel mobility shift assays on the basis of specific interactions between human choriodecidual cell nuclear extracts and an oligonucleotide including this polymorphism. CONCLUSION This study demonstrates that the human renin enhancer not only comprises the 225 bp element, but also extends to the region containing the -5312 SNP.
Collapse
Affiliation(s)
- Sébastien Fuchs
- INSERM Unit 36, Collège de France, 11 place Marcelin Berthelot, 75005 Paris, France.
| | | | | | | | | | | | | |
Collapse
|
29
|
Ryan MJ, Black TA, Millard SL, Gross KW, Hajduczok G. Endothelin-1 increases calcium and attenuates renin gene expression in As4.1 cells. Am J Physiol Heart Circ Physiol 2002; 283:H2458-65. [PMID: 12388321 DOI: 10.1152/ajpheart.00295.2002] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Endothelin-1 (ET-1) is a potent vasoconstrictor and blood pressure modulator. Renin secretion from juxtaglomerular (JG) cells is crucial for blood pressure and electrolyte homeostasis and has been shown to be modulated by ET-1; however, the cellular and molecular mechanism of this regulation is not clear. The purpose of this study was to gain a better understanding of the cellular and molecular pathways activated by ET-1 by using a renin-producing cell line As4.1. ET-1 caused an increase in As4.1 cell intracelluar Ca(2+) concentration ([Ca(2+)](i)) mediated by the ET(A) receptor as its antagonist, BQ-123, abolished the response. The nitric oxide donor nitroprusside, but not 8-bromo-cGMP, reduced the time necessary for successive ET-1 responses. Endothelin-3 had no effect on [Ca(2+)](i). ET-1 dose dependently increased total inositol phosphates with an EC(50) of 2.1 nM. ET-1 reduced renin mRNA by 68% independently of changes in message decay. With the use of a renin-luciferase reporter system in As4.1 cells, ET-1 reduced luciferase activity by 51%, suggesting that renin gene transcription is directly modified by ET-1.
Collapse
Affiliation(s)
- Michael J Ryan
- Department of Physiology and Biophysics, State University of New York at Buffalo, 14214, USA.
| | | | | | | | | |
Collapse
|
30
|
|
31
|
Sigmund CD. Regulation of renin expression and blood pressure by vitamin D(3). J Clin Invest 2002; 110:155-6. [PMID: 12122105 PMCID: PMC151071 DOI: 10.1172/jci16160] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Affiliation(s)
- Curt D Sigmund
- Department of Internal Medicine, 3181B Medical Education and Biomedical Research Facility, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, USA.
| |
Collapse
|
32
|
Abstract
With advances in genetic manipulation and molecular biological and physiological techniques, the mouse has become the animal model of choice for studying the genetic basis of human diseases. The two most commonly used methods for analyzing the function of a gene in vivo, overexpression (transgenic mouse) and deletion (knockout mouse), have been extremely useful in establishing the importance of genes in genetic disorders. The renin-angiotensin system (RAS) is one of the most widely studied systems controlling blood pressure. Although the primary site of Ang-II production is the plasma, all the components of the RAS cascade are expressed in many tissues, including the brain. This review briefly summarizes systemic and tissue-specific transgenic and knockout mouse models of the RAS for determining the role of this system in the regulation of blood pressure and in the pathogenesis of hypertension, with a focus on the RAS in the brain.
Collapse
Affiliation(s)
- Satoshi Morimoto
- Departments of Internal Medicine and Physiology & Biophisics, the University of Iowa College of Medicine, Iowa City, IA 52242, USA
| | | |
Collapse
|
33
|
Pan L, Black TA, Shi Q, Jones CA, Petrovic N, Loudon J, Kane C, Sigmund CD, Gross KW. Critical roles of a cyclic AMP responsive element and an E-box in regulation of mouse renin gene expression. J Biol Chem 2001; 276:45530-8. [PMID: 11564732 DOI: 10.1074/jbc.m103010200] [Citation(s) in RCA: 104] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Mouse As4.1 cells, obtained after transgene-targeted oncogenesis to induce neoplasia in renal renin expressing cells, express high levels of renin mRNA from their endogenous Ren-1(c) gene. We have previously identified a 242-base pair enhancer (coordinates -2866 to -2625 relative to the CAP site) upstream of the mouse Ren-1(c) gene. This enhancer, in combination with the proximal promoter (-117 to +6), activates transcription nearly 2 orders of magnitude in an orientation independent fashion. To further delimit sequences necessary for transcriptional activation, renin promoter-luciferase reporter gene constructs containing selected regions of the Ren-1(c) enhancer were analyzed after transfection into As4.1 cells. These results demonstrate that several regions are required for full enhancer activity. Sequences from -2699 to -2672, which are critical for the enhancer activity, contain a cyclic AMP responsive element (CRE) and an E-box. Electrophoretic mobility shift assays demonstrated that transcription factors CREB/CREM and USF1/USF2 in As4.1 cell nuclear extracts bind to oligonucleotides containing the Ren-1(c) CRE and E-box, respectively. These two elements are capable of synergistically activating transcription from the Ren-1(c) promoter. Moreover, mutation of either the CRE or E-box results in almost complete loss of enhancer activity, suggesting the critical roles these two elements play in regulating mouse Ren-1(c) gene expression. Although the Ren-1(c) gene contains a CRE, its expression is not induced by cAMP in As4.1 cells. This appears to reflect constitutive activation of protein kinase A in As4.1 cells since treatment with the protein kinase A inhibitor, H-89, caused a significant reduction in Ren-1(c) gene expression and this reduction is mediated through the CRE at -2699 to -2688.
Collapse
Affiliation(s)
- L Pan
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, New York 14263, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Shi SJ, Nguyen HT, Sharma GD, Navar LG, Pandey KN. Genetic disruption of atrial natriuretic peptide receptor-A alters renin and angiotensin II levels. Am J Physiol Renal Physiol 2001; 281:F665-73. [PMID: 11553513 DOI: 10.1152/ajprenal.2001.281.4.f665] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
We have studied cardiovascular and renal phenotypes in Npr1 (genetic determinant of natriuretic peptide receptor-A; NPRA) gene-disrupted mutant mouse model. The baseline systolic arterial pressure (SAP) in 0-copy mutant (-/-) mice (143 +/- 2 mmHg) was significantly higher than in 2-copy wild-type (+/+) animals (104 +/- 2 mmHg); however, the SAP in 1-copy heterozygotes (+/-) was at an intermediate value (120 +/- 4 mmHg). To determine whether Npr1 gene function affects the renin-angiotensin-aldosterone system (RAAS), we measured the components of RAAS in plasma, kidney, and adrenal gland of 0-copy, 1-copy, and 2-copy male mice. Newborn (2 days after the birth) 0-copy pups showed 2.5-fold higher intrarenal renin contents compared with 2-copy wild-type counterparts (0-copy 72 +/- 12 vs. 2-copy 30 +/- 7 microg ANG I. mg protein(-1). h(-1), respectively). The intrarenal ANG II level in 0-copy pups was also higher than in 2-copy controls (0-copy 33 +/- 5 vs. 2-copy 20 +/- 2 pg/mg protein, respectively). However, both young (3 wk) and adult (16 wk) 0-copy mutant mice showed a dramatic 50-80% reduction in plasma renin concentrations (PRCs) and in expression of renal renin message compared with 2-copy control animals. In contrast, the adrenal renin content and mRNA expression levels were 1.5- to 2-fold higher in 0-copy adult mice than in 2-copy animals. The results suggest that inhibition of renal and systemic RAAS is a compensatory response that prevents greater increases in elevated arterial pressures in adult NPRA null mutant mice. However, the greater renin and ANG II levels seen in 0-copy newborn pups provide evidence that the direct effect of NPRA activation on renin is an inhibitory response.
Collapse
Affiliation(s)
- S J Shi
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | | | | | | | | |
Collapse
|
35
|
Shi Q, Gross KW, Sigmund CD. Retinoic acid-mediated activation of the mouse renin enhancer. J Biol Chem 2001; 276:3597-603. [PMID: 11058598 DOI: 10.1074/jbc.m008361200] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Previous studies demonstrate that the mouse renin gene is regulated by a complex enhancer of transcription located 2.6 kilobases upstream of the transcription start site which is under both positive and negative influence. We demonstrate herein that a positive regulatory element (Eb) is repeated 10 bp upstream (Ec), and both are required for baseline activity of the enhancer. The Eb and Ec core sequences are identical to the consensus sequence for the nuclear hormone receptor superfamily of transcription factors, and transcriptional activity of constructs containing the enhancer is increased after treatment with retinoic acid. Maximal induction requires both Eb and Ec. Expression of endogenous renin and a renin-promoter controlled transgene in As4.1 cells, and kidney renin mRNA in C57BL/6J mice was induced after retinoid treatment. Gel mobility supershift analysis revealed the binding of RARalpha and RXRalpha to oligonucleotides containing both Eb and Ec. Reverse transcriptase-polymerase chain reaction analysis revealed that As4.1 cells express both receptor isoforms, along with RARgamma, but do not express RARbeta, RXRbeta, or RXRgamma. Co-transfection of an expression vector encoding wild-type RARalpha increased enhancer activity, whereas a dominant negative mutant of RARalpha significantly attenuated retinoic acid-induced activity of the enhancer. These results demonstrate the importance of the Eb and Ec motifs in controlling baseline activity of the renin enhancer, and suggest the potential importance of retinoids in regulating renin expression.
Collapse
Affiliation(s)
- Q Shi
- Department of Internal Medicine, The University of Iowa College of Medicine, Iowa City, Iowa 52242, USA
| | | | | |
Collapse
|
36
|
Ryan MJ, Black TA, Gross KW, Hajduczok G. Cyclic mechanical distension regulates renin gene transcription in As4.1 cells. Am J Physiol Endocrinol Metab 2000; 279:E830-7. [PMID: 11001765 DOI: 10.1152/ajpendo.2000.279.4.e830] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The renin-producing and -secreting juxtaglomerular (JG) cells are thought to function as the baroreceptor of the kidney. The mechanism by which changes in pressure, or mechanical force, regulate renin at the molecular level has not been elucidated. The renin gene-expressing and -secreting clonal cell line As4.1 was derived from transgene-targeted oncogenesis in mice and was used as a cellular model for JG cells. As4.1 cells subjected to cyclic mechanical distension for a period of 24 h at various frequencies (0. 05 or 0.5 Hz) and magnitudes (12 or 24% elongation) were analyzed via Northern analysis for renin mRNA levels. Results indicate that renin gene expression is decreased by 50-85% and returns to basal levels after a 24-h recovery period. Renin gene expression was attenuated independently of elevated cell growth or changes in renin message decay, suggesting that renin gene transcription is directly modulated by mechanical distension. Transient transfection of As4.1 cells with renin 5' flanking sequence-luciferase reporter gene constructs confirmed the role of mechanical stimulation in regulating renin gene transcription. A 43% inhibition of luciferase activity, by stretch, was observed in cells transfected with a 4,000 base pair 5' flanking sequence to the renin proximal promoter. These results demonstrate for the first time that changes in mechanical force can result in the regulation of renin gene transcription and thus provide further insight into the baroreceptor properties of renin-expressing cells.
Collapse
Affiliation(s)
- M J Ryan
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo 14214, USA
| | | | | | | |
Collapse
|
37
|
Sinn PL, Sigmund CD. Transgenic models as tools for studying the regulation of human renin expression. REGULATORY PEPTIDES 2000; 86:77-82. [PMID: 10672905 DOI: 10.1016/s0167-0115(99)00097-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Transgenic mice and rats have become popular tools to study the regulation of gene expression and the consequences of protein over-production. Over the past decade, numerous transgenic models have been developed to study the mechanisms of human renin gene expression and the participation of the renin-angiotensin system in the development of hypertension. Herein we will provide an overview of what has been learned from the use of transgenic models for studying the human renin gene.
Collapse
Affiliation(s)
- P L Sinn
- Department of Internal Medicine, The University of Iowa College of Medicine, Iowa City 52242, USA
| | | |
Collapse
|