1
|
Nagalakshmi VK, Smith JP, Matsuoka D, Gomez RA, Lopez MLSS. Piezo channels in JG cells do not regulate renin expression or renin release to the circulation. Clin Sci (Lond) 2024; 138:1527-1536. [PMID: 39564667 PMCID: PMC11843579 DOI: 10.1042/cs20242089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 11/21/2024]
Abstract
Renin-expressing juxtaglomerular (JG) cells possess an intrinsic pressure-sensing mechanism(s) that regulates renin synthesis and release in response to changes in perfusion pressure. Although we recently described the structure of the nuclear mechanotransducer that controls renin transcription, the acute pressure-sensing mechanism that controls the rapid release of renin has not been identified. In JG cells there is an inverse relationship between intracellular calcium and renin release, the 'calcium paradox'. Since the discovery of Piezo2 as the 'touch' receptors, there has been a significant interest in exploring whether they are also involved in other tissues beyond the skin. Given that Piezo receptors are permeable to calcium upon mechanical stimuli, it would be reasonable to hypothesize that Piezo2 controls renin synthesis and/or release in JG cells. To test this hypothesis, we used a variety of novel mouse models and JG cell-specific techniques to define whether Piezo2 controls renin expression and/or release in JG cells. Our in vivo data using constitutive and inducible Cre driver mouse lines and a variety of novel experimental approaches indicate that Piezo2 channels are not necessary for renin synthesis or release in JG cells during normal conditions or when homeostasis is threatened by hypotension, sodium depletion, or inverse changes in blood pressure. Furthermore, Piezo1 channels do not compensate for the lack of Piezo2 in JG cells. Efforts should be devoted to identifying the acute mechanosensory mechanisms controlling renin release.
Collapse
Affiliation(s)
- Vidya K. Nagalakshmi
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA
| | - Jason P. Smith
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA
| | - Daisuke Matsuoka
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA
| | - R. Ariel Gomez
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA
| | | |
Collapse
|
2
|
Smith JP, Paxton R, Medrano S, Sheffield NC, Sequeira-Lopez MLS, Ariel Gomez R. Inhibition of Renin Expression Is Regulated by an Epigenetic Switch From an Active to a Poised State. Hypertension 2024; 81:1869-1882. [PMID: 38989586 PMCID: PMC11337216 DOI: 10.1161/hypertensionaha.124.22886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 07/02/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Renin-expressing cells are myoendocrine cells crucial for the maintenance of homeostasis. Renin is regulated by cAMP, p300 (histone acetyltransferase p300)/CBP (CREB-binding protein), and Brd4 (bromodomain-containing protein 4) proteins and associated pathways. However, the specific regulatory changes that occur following inhibition of these pathways are not clear. METHODS We treated As4.1 cells (tumoral cells derived from mouse juxtaglomerular cells that constitutively express renin) with 3 inhibitors that target different factors required for renin transcription: H-89-dihydrochloride, PKA (protein kinase A) inhibitor; JQ1, Brd4 bromodomain inhibitor; and A-485, p300/CBP inhibitor. We performed assay for transposase-accessible chromatin with sequencing (ATAC-seq), single-cell RNA sequencing, cleavage under targets and tagmentation (CUT&Tag), and chromatin immunoprecipitation sequencing for H3K27ac (acetylation of lysine 27 of the histone H3 protein) and p300 binding on biological replicates of treated and control As4.1 cells. RESULTS In response to each inhibitor, Ren1 expression was significantly reduced and reversible upon washout. Chromatin accessibility at the Ren1 locus did not markedly change but was globally reduced at distal elements. Inhibition of PKA led to significant reductions in H3K27ac and p300 binding specifically within the Ren1 super-enhancer region. Further, we identified enriched TF (transcription factor) motifs shared across each inhibitory treatment. Finally, we identified a set of 9 genes with putative roles across each of the 3 renin regulatory pathways and observed that each displayed differentially accessible chromatin, gene expression, H3K27ac, and p300 binding at their respective loci. CONCLUSIONS Inhibition of renin expression in cells that constitutively synthesize and release renin is regulated by an epigenetic switch from an active to poised state associated with decreased cell-cell communication and an epithelial-mesenchymal transition. This work highlights and helps define the factors necessary for renin cells to alternate between myoendocrine and contractile phenotypes.
Collapse
Affiliation(s)
- Jason P. Smith
- Department of Pediatrics, Child Health Research Center, University of Virginia, Charlottesville, Virginia
| | - Robert Paxton
- Department of Pediatrics, Child Health Research Center, University of Virginia, Charlottesville, Virginia
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Silvia Medrano
- Department of Pediatrics, Child Health Research Center, University of Virginia, Charlottesville, Virginia
| | - Nathan C. Sheffield
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia
- Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia
| | | | - R. Ariel Gomez
- Department of Pediatrics, Child Health Research Center, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
3
|
Saito N, Toyoda M, Kondo M, Abe M, Sanechika N, Kimura M, Sawada K, Fukagawa M. Regulation of Renin Expression by Β1-Integrin in As4.1 Juxtaglomerular Line Cells. Biomedicines 2023; 11:biomedicines11020501. [PMID: 36831037 PMCID: PMC9953579 DOI: 10.3390/biomedicines11020501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
(1) Background: Renal dysfunction and hypertension are mutually aggravating factors; however, the details of their interaction remain unclear. In a study using renal tissue from diabetic rats, we found that β1-integrin, a cell-substrate adhesion molecule, is specifically phosphorylated in juxtaglomerular cells that secrete renin, a blood pressure regulator. (2) Methods: A mouse juxtaglomerular cell line (As4.1 cells) was used for the following experiments: drug-induced promotion of β1-integrin phosphorylation/dephosphorylation; knockdown of β1-integrin and the cell adhesion molecule connexin-40 (a candidate for the main body of baroreceptor); and pressurization to atmospheric pressure + 100 mmHg. culture in hypotonic liquid medium. The expression of renin under these conditions was measured by qRT-PCR. (3) Results: Phosphorylation of β1-integrin suppressed the expression of renin, while dephosphorylation conversely promoted it. β1-integrin and connexin-40 knockdown both promoted the expression of renin. Pneumatic pressurization and hypotonic medium culture both decreased the expression of renin, which was restored by the knockdown of β1-integrin. (4) Conclusions: β1-integrin plays an inhibitory role in the regulation of the expression of renin, which may be controlled by phosphorylation and dephosphorylation. It is hypothesized that β1-integrin and other adhesion factors regulate the expression of renin by altering the sensitivity of baroreceptors on the plasma membrane.
Collapse
Affiliation(s)
| | - Masao Toyoda
- Correspondence: ; Tel.: +81-463-93-1121 (ext. 2490)
| | | | | | | | | | | | | |
Collapse
|
4
|
Martini AG, Smith JP, Medrano S, Sheffield NC, Sequeira-Lopez MLS, Gomez RA. Determinants of renin cell differentiation: a single cell epi-transcriptomics approach. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.18.524595. [PMID: 36711565 PMCID: PMC9882312 DOI: 10.1101/2023.01.18.524595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Rationale Renin cells are essential for survival. They control the morphogenesis of the kidney arterioles, and the composition and volume of our extracellular fluid, arterial blood pressure, tissue perfusion, and oxygen delivery. It is known that renin cells and associated arteriolar cells descend from FoxD1 + progenitor cells, yet renin cells remain challenging to study due in no small part to their rarity within the kidney. As such, the molecular mechanisms underlying the differentiation and maintenance of these cells remain insufficiently understood. Objective We sought to comprehensively evaluate the chromatin states and transcription factors (TFs) that drive the differentiation of FoxD1 + progenitor cells into those that compose the kidney vasculature with a focus on renin cells. Methods and Results We isolated single nuclei of FoxD1 + progenitor cells and their descendants from FoxD1 cre/+ ; R26R-mTmG mice at embryonic day 12 (E12) (n cells =1234), embryonic day 18 (E18) (n cells =3696), postnatal day 5 (P5) (n cells =1986), and postnatal day 30 (P30) (n cells =1196). Using integrated scRNA-seq and scATAC-seq we established the developmental trajectory that leads to the mosaic of cells that compose the kidney arterioles, and specifically identified the factors that determine the elusive, myo-endocrine adult renin-secreting juxtaglomerular (JG) cell. We confirm the role of Nfix in JG cell development and renin expression, and identified the myocyte enhancer factor-2 (MEF2) family of TFs as putative drivers of JG cell differentiation. Conclusions We provide the first developmental trajectory of renin cell differentiation as they become JG cells in a single-cell atlas of kidney vascular open chromatin and highlighted novel factors important for their stage-specific differentiation. This improved understanding of the regulatory landscape of renin expressing JG cells is necessary to better learn the control and function of this rare cell population as overactivation or aberrant activity of the RAS is a key factor in cardiovascular and kidney pathologies.
Collapse
|
5
|
Chang JW, Kim S, Lee EY, Leem CH, Kim SH, Park CS. Cell-cell contacts via N-cadherin induce a regulatory renin secretory phenotype in As4.1 cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2022; 26:479-499. [PMID: 36302623 PMCID: PMC9614399 DOI: 10.4196/kjpp.2022.26.6.479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/05/2022] [Accepted: 09/19/2022] [Indexed: 11/07/2022]
Abstract
The lack of a clonal renin-secreting cell line has greatly hindered the investigation of the regulatory mechanisms of renin secretion at the cellular, biochemical, and molecular levels. In the present study, we investigated whether it was possible to induce phenotypic switching of the renin-expressing clonal cell line As4.1 from constitutive inactive renin secretion to regulated active renin secretion. When grown to postconfluence for at least two days in media containing fetal bovine serum or insulin-like growth factor-1, the formation of cell-cell contacts via N-cadherin triggered downstream cellular signaling cascades and activated smooth muscle-specific genes, culminating in phenotypic switching to a regulated active renin secretion phenotype, including responding to the key stimuli of active renin secretion. With the use of phenotype-switched As4.1 cells, we provide the first evidence that active renin secretion via exocytosis is regulated by phosphorylation/dephosphorylation of the 20 kDa myosin light chain. The molecular mechanism of phenotypic switching in As4.1 cells described here could serve as a working model for full phenotypic modulation of other secretory cell lines with incomplete phenotypes.
Collapse
Affiliation(s)
- Jai Won Chang
- Department of Physiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea.,Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Soohyun Kim
- Department of Physiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Eun Young Lee
- Department of Internal Medicine, The Catholic University of Korea, Seoul St. Mary's Hospital, Seoul 06591, Korea
| | - Chae Hun Leem
- Department of Physiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Suhn Hee Kim
- Department of Physiology, Jeonbuk National University Medical School, Jeonju 54907, Korea
| | - Chun Sik Park
- Department of Physiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| |
Collapse
|
6
|
Kumar S, Ali W, Mishra S, Pradhan A, Sethi R, Kushwaha R, Singh US, Perrone MA. Circulating Soluble Lectin-like Oxidized Low-Density Lipoprotein Receptor-1 (sLOX-1): A Diagnostic Indicator across the Spectrum of Acute Coronary Syndrome. J Clin Med 2021; 10:5567. [PMID: 34884269 PMCID: PMC8658522 DOI: 10.3390/jcm10235567] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/11/2021] [Accepted: 11/23/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Cardiac troponin is the best marker to diagnose acute coronary syndrome (ACS). However, early diagnosis using markers for plaque instability may be of significance. Soluble lectin-like oxidized low-density lipoprotein receptor-1 (sLOX-1) plays an important role in the pathogenesis of atherosclerosis plaque rupture and may be a potential biomarker of coronary artery disease (CAD), including ACS. The current study aims to evaluate sLOX-1 levels in the sera of patients with ACS as an independent marker of CAD with other established diagnostic markers and assess its level before and after percutaneous intervention (PCI) in predicting the risk of future recurrence of ACS. METHODS Peripheral blood was obtained from a total of 160 patients, including patients who underwent coronary angiography (n = 18, group I), patients of stable CAD who underwent percutaneous intervention (n = 50, group II), patients of the acute coronary syndrome (n = 64, group III), and healthy controls (n = 28, group IV). A serum sLOX-1 concentration was measured by the enzyme-linked immunosorbent assay (ELISA). RESULTS The results obtained showed a statistically significant raised level of sLOX-1 in pre/post PCI patients of stable CAD/ACS with male preponderance. The area under the curve for sLOX-1 was 0.925 for cases that are discriminated from controls with sensitivity and specificity of 87.88 and 100%, respectively. SLOX-1 showed 100% sensitivity and specificity in the discrimination of the stable CAD that underwent PCI vs. control with an AUC of 1.00. The recurrence of coronary artery disease was observed in 9 out of 132 (6.8%) cases. The post-interventional sLOX-1 level was significantly different and higher in recurrent cases (p = 0.027) of ACS/CAD. CONCLUSIONS sLOX-1 was a useful biomarker of stable CAD/ACS and has a potential in the risk prediction of a future recurrence of coronary artery disease.
Collapse
Affiliation(s)
- Sandeep Kumar
- Department of Pathology, King George’s Medical University, Lucknow 226003, India; (S.K.); (W.A.); (R.K.); (U.S.S.)
| | - Wahid Ali
- Department of Pathology, King George’s Medical University, Lucknow 226003, India; (S.K.); (W.A.); (R.K.); (U.S.S.)
| | - Sridhar Mishra
- Department of Pathology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow 226010, India;
| | - Akshyaya Pradhan
- Department of Cardiology, Lari Cardiology Centre, King George’s Medical University, Lucknow 226003, India; (A.P.); (R.S.)
| | - Rishi Sethi
- Department of Cardiology, Lari Cardiology Centre, King George’s Medical University, Lucknow 226003, India; (A.P.); (R.S.)
| | - Rashmi Kushwaha
- Department of Pathology, King George’s Medical University, Lucknow 226003, India; (S.K.); (W.A.); (R.K.); (U.S.S.)
| | - Uma Shankar Singh
- Department of Pathology, King George’s Medical University, Lucknow 226003, India; (S.K.); (W.A.); (R.K.); (U.S.S.)
| | | |
Collapse
|
7
|
Belyea BC, Santiago AE, Vasconez WA, Nagalakshmi VK, Xu F, Mehalic TC, Sequeira-Lopez MLS, Gomez RA. A primitive type of renin-expressing lymphocyte protects the organism against infections. Sci Rep 2021; 11:7251. [PMID: 33790364 PMCID: PMC8012387 DOI: 10.1038/s41598-021-86629-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 03/02/2021] [Indexed: 12/11/2022] Open
Abstract
The hormone renin plays a crucial role in the regulation of blood pressure and fluid-electrolyte homeostasis. Normally, renin is synthesized by juxtaglomerular (JG) cells, a specialized group of myoepithelial cells located near the entrance to the kidney glomeruli. In response to low blood pressure and/or a decrease in extracellular fluid volume (as it occurs during dehydration, hypotension, or septic shock) JG cells respond by releasing renin to the circulation to reestablish homeostasis. Interestingly, renin-expressing cells also exist outside of the kidney, where their function has remained a mystery. We discovered a unique type of renin-expressing B-1 lymphocyte that may have unrecognized roles in defending the organism against infections. These cells synthesize renin, entrap and phagocyte bacteria and control bacterial growth. The ability of renin-bearing lymphocytes to control infections-which is enhanced by the presence of renin-adds a novel, previously unsuspected dimension to the defense role of renin-expressing cells, linking the endocrine control of circulatory homeostasis with the immune control of infections to ensure survival.
Collapse
Affiliation(s)
- Brian C Belyea
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Araceli E Santiago
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Wilson A Vasconez
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Vidya K Nagalakshmi
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Fang Xu
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Theodore C Mehalic
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Maria Luisa S Sequeira-Lopez
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA.
| | - R Ariel Gomez
- Department of Pediatrics, Child Health Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA.
| |
Collapse
|
8
|
Role of Pericytes in the Development of the Renin/Angiotensin System: Induction of Functional Renin in Cultures of Pericytes. Methods Mol Biol 2021; 2235:169-180. [PMID: 33576977 DOI: 10.1007/978-1-0716-1056-5_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Renal pericytes have a critical importance for angiogenesis and vascular remodeling, medullary blood flow regulation, and development of fibrosis. An emerging role for kidney pericytes is their ability to induce renin expression and synthesis. Here, we present methods for purification of human renal pericytes, their primary culture, and differentiation into renin-producing cells. Possible applications of these protocols include investigations into (1) renin cell recruitment mechanisms, (2) modulation of renin expression/secretion by small molecules, and (3) renin expression/secretion in nonrenal pericytes. A potential therapeutic application of this work is the identification of new players regulating the renin-angiotensin system.
Collapse
|
9
|
Sauder KA, Stamatoiu AV, Leshchinskaya E, Ringham BM, Glueck DH, Dabelea D. Cord Blood Vitamin D Levels and Early Childhood Blood Pressure: The Healthy Start Study. J Am Heart Assoc 2020; 8:e011485. [PMID: 31020895 PMCID: PMC6512116 DOI: 10.1161/jaha.118.011485] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Vitamin D deficiency is associated with cardiovascular events among adults, but it is unclear whether early-life vitamin D deficiency influences cardiovascular risk factors in children. Methods and Results We measured total and bioavailable 25-dihydroxyvitamin D (25OHD) in cord blood and in blood from 4- to 6-year-old children, and we assessed cardiovascular risk factors (blood pressure, arterial stiffness, body size, and adiposity) at 4 to 6 years. We tested for racial/ethnic differences in total and bioavailable 25OHD (n=715) and modeled the adjusted association between cord blood 25OHD and childhood cardiovascular risk factors (n=171). We observed racial/ethnic differences in total and bioavailable 25OHD levels in both cord and child blood samples (all P<0.05). Each 25-nmol/L increase in cord blood total 25OHD was associated with a 2.5-mm Hg (SE 0.8) decrease in systolic blood pressure ( P=0.002) and a 1.7-mm Hg (SE 0.6) decrease in diastolic blood pressure ( P=0.01), independent of childhood 25OHD levels, race/ethnicity, and other covariates. There was no association between cord blood total 25OHD and any other cardiovascular risk factors. Cord blood levels of bioavailable and free 25OHD were not associated with any cardiovascular risk factor in childhood. Conclusions In this diverse prebirth cohort, we observed lower systolic and diastolic blood pressure among children with higher total 25OHD levels at birth. Our findings suggest that intrauterine exposure to vitamin D may contribute to early-life programming of offspring blood pressure. Intervention studies are needed to determine whether increasing fetal vitamin D exposure can reduce the risk of elevated blood pressure in childhood.
Collapse
Affiliation(s)
- Katherine A Sauder
- 1 Department of Pediatrics University of Colorado School of Medicine Aurora CO.,2 Department of Epidemiology Colorado School of Public Health Aurora CO.,3 Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center University of Colorado Aurora CO
| | - Alexandra V Stamatoiu
- 3 Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center University of Colorado Aurora CO
| | - Elina Leshchinskaya
- 3 Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center University of Colorado Aurora CO
| | - Brandy M Ringham
- 2 Department of Epidemiology Colorado School of Public Health Aurora CO.,3 Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center University of Colorado Aurora CO
| | - Deborah H Glueck
- 1 Department of Pediatrics University of Colorado School of Medicine Aurora CO.,3 Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center University of Colorado Aurora CO
| | - Dana Dabelea
- 1 Department of Pediatrics University of Colorado School of Medicine Aurora CO.,2 Department of Epidemiology Colorado School of Public Health Aurora CO.,3 Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center University of Colorado Aurora CO
| |
Collapse
|
10
|
Martinez MF, Martini AG, Sequeira-Lopez MLS, Gomez RA. Ctcf is required for renin expression and maintenance of the structural integrity of the kidney. Clin Sci (Lond) 2020; 134:1763-1774. [PMID: 32619009 PMCID: PMC7881370 DOI: 10.1042/cs20200184] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 06/23/2020] [Accepted: 07/03/2020] [Indexed: 12/17/2022]
Abstract
Renin cells are crucial for the regulation of blood pressure and fluid electrolyte homeostasis. We have recently shown that renin cells possess unique chromatin features at regulatory regions throughout the genome that may determine the identity and memory of the renin phenotype. The 3-D structure of chromatin may be equally important in the determination of cell identity and fate. CCCTC-binding factor (Ctcf) is a highly conserved chromatin organizer that may regulate the renin phenotype by controlling chromatin structure. We found that Ctcf binds at several conserved DNA sites surrounding and within the renin locus, suggesting that Ctcf may regulate the transcriptional activity of renin cells. In fact, deletion of Ctcf in cells of the renin lineage led to decreased endowment of renin-expressing cells accompanied by decreased circulating renin, hypotension, and severe morphological abnormalities of the kidney, including defects in arteriolar branching, and ultimately renal failure. We conclude that control of chromatin architecture by Ctcf is necessary for the appropriate expression of renin, control of renin cell number and structural integrity of the kidney.
Collapse
Affiliation(s)
- Maria Florencia Martinez
- Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, 22908. United States
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, 22908. United States
| | - Alexandre G. Martini
- Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, 22908. United States
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, 22908. United States
| | - Maria Luisa S. Sequeira-Lopez
- Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, 22908. United States
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, 22908. United States
- Department of Biology, University of Virginia School of Medicine, Charlottesville, Virginia, 22908. United States
| | - R. Ariel Gomez
- Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia, 22908. United States
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, 22908. United States
- Department of Biology, University of Virginia School of Medicine, Charlottesville, Virginia, 22908. United States
| |
Collapse
|
11
|
Martinez MF, Medrano S, Brown RI, Tufan T, Shang S, Bertoncello N, Guessoum O, Adli M, Belyea BC, Sequeira-Lopez MLS, Gomez RA. Super-enhancers maintain renin-expressing cell identity and memory to preserve multi-system homeostasis. J Clin Invest 2018; 128:4787-4803. [PMID: 30130256 PMCID: PMC6205391 DOI: 10.1172/jci121361] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 08/14/2018] [Indexed: 02/06/2023] Open
Abstract
Renin cells are crucial for survival - they control fluid-electrolyte and blood pressure homeostasis, vascular development, regeneration, and oxygen delivery to tissues. During embryonic development, renin cells are progenitors for multiple cell types that retain the memory of the renin phenotype. When there is a threat to survival, those descendants are transformed and reenact the renin phenotype to restore homeostasis. We tested the hypothesis that the molecular memory of the renin phenotype resides in unique regions and states of these cells' chromatin. Using renin cells at various stages of stimulation, we identified regions in the genome where the chromatin is open for transcription, mapped histone modifications characteristic of active enhancers such as H3K27ac, and tracked deposition of transcriptional activators such as Med1, whose deletion results in ablation of renin expression and low blood pressure. Using the rank ordering of super-enhancers, epigenetic rewriting, and enhancer deletion analysis, we found that renin cells harbor a unique set of super-enhancers that determine their identity. The most prominent renin super-enhancer may act as a chromatin sensor of signals that convey the physiologic status of the organism, and is responsible for the transformation of renin cell descendants to the renin phenotype, a fundamental process to ensure homeostasis.
Collapse
Affiliation(s)
| | | | | | - Turan Tufan
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Stephen Shang
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | | | - Omar Guessoum
- Child Health Research Center
- Department of Pediatrics
- Department of Biology, and
| | - Mazhar Adli
- Child Health Research Center
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | | | | | - R. Ariel Gomez
- Child Health Research Center
- Department of Pediatrics
- Department of Biology, and
| |
Collapse
|
12
|
Mrowka R, Kirchner S, Reuter S. How to switch on genes with CRISPR/Cas9? Acta Physiol (Oxf) 2018; 224:e13087. [PMID: 29729207 DOI: 10.1111/apha.13087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- R. Mrowka
- Experimentelle Nephrologie; Universitätsklinikum Jena, KIM III; Jena Germany
| | - S. Kirchner
- Experimentelle Nephrologie; Universitätsklinikum Jena, KIM III; Jena Germany
| | - S. Reuter
- Experimentelle Nephrologie; Universitätsklinikum Jena, KIM III; Jena Germany
| |
Collapse
|
13
|
Homeostatic Response of Mouse renin Gene Transcription in a Hypertensive Environment Is Mediated by a Novel 5' Enhancer. Mol Cell Biol 2018; 38:MCB.00566-17. [PMID: 29358217 DOI: 10.1128/mcb.00566-17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 01/17/2018] [Indexed: 01/22/2023] Open
Abstract
The renin-angiotensin system plays an essential role in blood pressure homeostasis. Because renin activity is reflected as a blood pressure phenotype, its gene expression in the kidney is tightly regulated by a feedback mechanism; i.e., renin gene transcription is suppressed in a hypertensive state. To address the molecular mechanisms controlling hypertension-responsive mouse renin (mRen) gene regulation, we deleted either 5' (17-kb) or 3' (78-kb) regions of the endogenous mRen gene and placed the animals in a hypertensive environment. While the mRen gene bearing the 3' deletion was appropriately downregulated, the one bearing the 5' deletion lost this hypertension responsiveness. Because the 17-kb sequence exhibited enhancer activity in vivo and in vitro, we narrowed down the enhancer to a 2.3-kb core using luciferase assays in As4.1 cells. When this 2.3-kb sequence was removed from the endogenous mRen gene in the mouse, its basal expression was dramatically reduced, and the hypertension responsiveness was significantly attenuated. Furthermore, we demonstrated that the angiotensin II signal played an important role in mRen gene suppression. We propose that in a hypertensive environment, the activity of this novel enhancer is attenuated, and, as a consequence, mRen gene transcription is suppressed to maintain blood pressure.
Collapse
|
14
|
Stodola TJ, Liu P, Liu Y, Vallejos AK, Geurts AM, Greene AS, Liang M. Genome-wide map of proximity linkage to renin proximal promoter in rat. Physiol Genomics 2018. [PMID: 29521603 DOI: 10.1152/physiolgenomics.00132.2017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
A challenge to understanding enhancer-gene relationships is that enhancers are not always sequentially close to the gene they regulate. Physical proximity mapping through sequencing can provide an unbiased view of the chromatin close to the proximal promoter of the renin gene ( Ren). Our objective was to determine genomic regions that physically interact with the renin proximal promoter, using two different genetic backgrounds, the Dahl salt sensitive and normotensive SS-13BN, which have been shown to have different regulation of plasma renin in vivo. The chromatin conformation capture method with sequencing focused at the Ren proximal promoter in rat-derived cardiac endothelial cells was used. Cells were fixed, chromatin close to the Ren promoter was captured, and fragments were sequenced. The clustering of mapped reads produced a genome-wide map of chromatin in contact with the Ren promoter. The largest number of contacts was found on chromosome 13, the chromosome with Ren, and contacts were found on all other chromosomes except chromosome X. These contacts were significantly enriched with genes positively correlated with Ren expression and with mapped quantitative trait loci associated with blood pressure, cardiovascular, and renal phenotypes. The results were reproducible in an independent biological replicate. The findings reported here represent the first map between a critical cardiovascular gene and physical interacting loci throughout the genome and will provide the basis for several new directions of research.
Collapse
Affiliation(s)
- Timothy J Stodola
- Center of Systems Molecular Medicine, Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Pengyuan Liu
- Center of Systems Molecular Medicine, Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin.,Cancer Center, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Yong Liu
- Center of Systems Molecular Medicine, Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Andrew K Vallejos
- Department of Biomedical Engineering, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Aron M Geurts
- Center of Systems Molecular Medicine, Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Andrew S Greene
- Center of Systems Molecular Medicine, Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin.,Department of Biomedical Engineering, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Mingyu Liang
- Center of Systems Molecular Medicine, Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin
| |
Collapse
|
15
|
Affiliation(s)
- Pablo Nakagawa
- From the Department of Pharmacology, UIHC Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City
| | - Curt D Sigmund
- From the Department of Pharmacology, UIHC Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City.
| |
Collapse
|
16
|
Baseline Vitamin D Deficiency Decreases the Effectiveness of Statins in HIV-Infected Adults on Antiretroviral Therapy. J Acquir Immune Defic Syndr 2017; 74:539-547. [PMID: 28045766 DOI: 10.1097/qai.0000000000001281] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
OBJECTIVE Vitamin D deficiency is common in HIV. Statins may increase vitamin D, and it is unknown whether vitamin D modifies the effect of statins on cardiovascular disease. DESIGN SATURN-HIV was a 96-week, randomized, placebo-controlled trial designed to evaluate the effect of rosuvastatin on immune activation and subclinical vascular disease in HIV-infected adults on antiretroviral therapy. This analysis focuses on the prespecified secondary endpoint 25-hydroxyvitamin D [25(OH)D] concentrations. METHODS Mixed effects linear modeling and analysis of variance were used to assess the rosuvastatin effect on plasma 25(OH)D concentrations over time and to determine whether baseline vitamin D modifies the rosuvastatin effect on changes in outcomes over the trial. RESULTS Hundred forty-seven adults were randomized (72 to rosuvastatin and 75 to placebo); 78% were men, 68% African American, with a mean age of 45 years. Baseline 25(OH)D concentrations were similar (overall mean 18 ng/mL) with 65% of participants below 20 ng/mL. Changes in 25(OH)D at 96 weeks were small and not significant within- or between-rosuvastatin and placebo groups. There were significant group by vitamin D status interactions for changes in low-density lipoprotein-cholesterol, proportion of patrolling monocytes expressing tissue factor (CD14dimCD16+TF+), lipoprotein-associated phospholipase A2, and common carotid artery intima media thickness at most time points. For each of these outcomes, the beneficial effects of rosuvastatin were either not apparent or attenuated in participants with 25(OH)D <20 ng/mL. CONCLUSIONS Although 25(OH)D did not change with rosuvastatin, baseline vitamin D deficiency decreased the effectiveness of rosuvastatin. Vitamin D supplementation may be warranted for deficient patients initiating statin therapy.
Collapse
|
17
|
Seghers F, Yerna X, Zanou N, Devuyst O, Vennekens R, Nilius B, Gailly P. TRPV4 participates in pressure-induced inhibition of renin secretion by juxtaglomerular cells. J Physiol 2016; 594:7327-7340. [PMID: 27779758 DOI: 10.1113/jp273595] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 10/20/2016] [Indexed: 12/31/2022] Open
Abstract
KEY POINTS Increase in blood pressure in the renal afferent arteriole is known to induce an increase in cytosolic calcium concentration ([Ca2+ ]i ) of juxtaglomerular (JG) cells and to result in a decreased secretion of renin. Mechanical stimulation of As4.1 JG cells induces an increase in [Ca2+ ]i that is inhibited by HC067047 and RN1734, two inhibitors of TRPV4, or by siRNA-mediated repression of TRPV4. Inhibition of TRPV4 impairs pressure-induced decrease in renin secretion. Compared to wild-type mice, Trpv4-/- mice present increased resting plasma levels of renin and aldosterone and present a significantly altered pressure-renin relationship. We suggest that TRPV4 channel participates in mechanosensation at the juxtaglomerular apparatus. ABSTRACT The renin-angiotensin system is a crucial blood pressure regulation system. It consists of a hormonal cascade where the rate-limiting enzyme is renin, which is secreted into the blood flow by renal juxtaglomerular (JG) cells in response to low pressure in the renal afferent arteriole. In contrast, an increase in blood pressure results in a decreased renin secretion. This is accompanied by a transitory increase in [Ca2+ ]i of JG cells. The inverse relationship between [Ca2+ ]i and renin secretion has been called the 'calcium paradox' of renin release. How increased pressure induces a [Ca2+ ]i transient in JG cells, is however, unknown. We observed that [Ca2+ ]i transients induced by mechanical stimuli in JG As4.1 cells were completely abolished by HC067047 and RN1734, two inhibitors of TRPV4. They were also reduced by half by siRNA-mediated repression of TRPV4 but not after repression or inhibition of TRPV2 or Piezo1 ion channels. Interestingly, the stimulation of renin secretion by the adenylate cyclase activator forskolin was totally inhibited by cyclic stretching of the cells. This effect was mimicked by stimulation with GSK1016790A and 4αPDD, two activators of TRPV4 and inhibited in the presence of HC067047. Moreover, in isolated perfused kidneys from Trpv4-/- mice, the pressure-renin relationship was significantly altered. In vivo, Trpv4-/- mice presented increased plasma levels of renin and aldosterone compared to wild-type mice. Altogether, our results suggest that TRPV4 is involved in the pressure-induced entry of Ca2+ in JG cells, which inhibits renin release and allows the negative feedback regulation on blood pressure.
Collapse
Affiliation(s)
- François Seghers
- Université catholique de Louvain, Institute of Neuroscience, Laboratory of Cell Physiology, av. Mounier 53, box B1.53.17, B-1200, Brussels, Belgium
| | - Xavier Yerna
- Université catholique de Louvain, Institute of Neuroscience, Laboratory of Cell Physiology, av. Mounier 53, box B1.53.17, B-1200, Brussels, Belgium
| | - Nadège Zanou
- Université catholique de Louvain, Institute of Neuroscience, Laboratory of Cell Physiology, av. Mounier 53, box B1.53.17, B-1200, Brussels, Belgium
| | - Olivier Devuyst
- University of Zurich, Institute of Physiology, CH-8057, Zurich, Switzerland
| | - Rudi Vennekens
- Katholieke Universiteit Leuven, Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, B-3000, Leuven, Belgium
| | - Bernd Nilius
- Katholieke Universiteit Leuven, Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, B-3000, Leuven, Belgium
| | - Philippe Gailly
- Université catholique de Louvain, Institute of Neuroscience, Laboratory of Cell Physiology, av. Mounier 53, box B1.53.17, B-1200, Brussels, Belgium
| |
Collapse
|
18
|
Ushiki A, Matsuzaki H, Ishida J, Fukamizu A, Tanimoto K. Long-Range Control of Renin Gene Expression in Tsukuba Hypertensive Mice. PLoS One 2016; 11:e0166974. [PMID: 27861631 PMCID: PMC5115840 DOI: 10.1371/journal.pone.0166974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 11/07/2016] [Indexed: 01/14/2023] Open
Abstract
Renin, a rate-limiting enzyme in the renin–angiotensin system, is regulated to maintain blood pressure homeostasis: renin gene expression in the kidney is suppressed in a hypertensive environment. We found that expression of a 15-kb human RENIN (hREN) transgene was aberrantly upregulated (>4.2-fold), while the endogenous mouse renin (mRen) gene was suppressed (>1.7-fold) in Tsukuba hypertensive mice (THM), a model for genetically induced hypertension. We then generated transgenic mice using a 13-kb mRen gene fragment that was homologous to the 15-kb hREN transgene and found that its expression was also upregulated (>3.1-fold) in THM, suggesting that putative silencing elements of the renin genes were distally located in the loci. We next examined the possible role of a previously identified mouse distal enhancer (mdE) located outside of the 13-kb mRen gene fragment. Deletion of the mdE in the context of a 156-kb mRen transgene did not affect its transcriptional repression in THM, implying that although the silencing element of the mRen gene is located within the 156-kb fragment tested, it is distinct from the mdE. Consistent with these results, deletion of the 63-kb region upstream of the mdE from the endogenous mRen gene locus abrogated its transcriptional repression in THM. We finally tested whether dysregulation of the short renin transgenes also occurred in the fetal or neonatal kidneys of THM and found that their expression was not aberrantly upregulated, demonstrating that aberrant regulation of short renin transgenes commences sometime between neonate and adult periods.
Collapse
Affiliation(s)
- Aki Ushiki
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hitomi Matsuzaki
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Life Science Center, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Junji Ishida
- Life Science Center, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Akiyoshi Fukamizu
- Life Science Center, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Keiji Tanimoto
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Life Science Center, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, Japan
- * E-mail:
| |
Collapse
|
19
|
Yu R. Animal models of spontaneous pancreatic neuroendocrine tumors. Mol Cell Endocrinol 2016; 421:60-7. [PMID: 26261055 DOI: 10.1016/j.mce.2015.08.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 07/10/2015] [Accepted: 08/04/2015] [Indexed: 01/20/2023]
Abstract
Pancreatic neuroendocrine tumors (PNETs) are usually low-grade neoplasms derived from the endocrine pancreas. PNETs can be functioning and cause well-described hormonal hypersecretion syndromes or non-functioning and cause only tumor mass effect. PNETs appear to be more common recently likely due to incidental detection by imaging. Although the diagnosis and management of PNETs have been evolving rapidly, much remains to be studied in the areas of molecular pathogenesis, molecular markers of tumor behavior, early detection, and targeted drug therapy. Unique challenges facing PNETs studies are long disease course, the deep location of pancreas and difficult access to pancreatic tissue, and the variety of tumors, which make animal models valuable tools for PNETs studies. Existing animal models of PNETs have provided insights into the pathogenesis and natural history of human PNETs. Future studies on animal models of PNETs should address early tumor detection, molecular markers of tumor behavior, and novel targeted therapies.
Collapse
Affiliation(s)
- Run Yu
- Division of Endocrinology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
20
|
Ihnatovych I, Sielski NL, Hofmann WA. Selective expression of myosin IC Isoform A in mouse and human cell lines and mouse prostate cancer tissues. PLoS One 2014; 9:e108609. [PMID: 25259793 PMCID: PMC4178219 DOI: 10.1371/journal.pone.0108609] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 09/01/2014] [Indexed: 12/15/2022] Open
Abstract
Myosin IC is a single headed member of the myosin superfamily. We recently identified a novel isoform and showed that the MYOIC gene in mammalian cells encodes three isoforms (isoforms A, B, and C). Furthermore, we demonstrated that myosin IC isoform A but not isoform B exhibits a tissue specific expression pattern. In this study, we extended our analysis of myosin IC isoform expression patterns by analyzing the protein and mRNA expression in various mammalian cell lines and in various prostate specimens and tumor tissues from the transgenic mouse prostate (TRAMP) model by immunoblotting, qRT-PCR, and by indirect immunohistochemical staining of paraffin embedded prostate specimen. Analysis of a panel of mammalian cell lines showed an increased mRNA and protein expression of specifically myosin IC isoform A in a panel of human and mouse prostate cancer cell lines but not in non-cancer prostate or other (non-prostate-) cancer cell lines. Furthermore, we demonstrate that myosin IC isoform A expression is significantly increased in TRAMP mouse prostate samples with prostatic intraepithelial neoplasia (PIN) lesions and in distant site metastases in lung and liver when compared to matched normal tissues. Our observations demonstrate specific changes in the expression of myosin IC isoform A that are concurrent with the occurrence of prostate cancer in the TRAMP mouse prostate cancer model that closely mimics clinical prostate cancer. These data suggest that elevated levels of myosin IC isoform A may be a potential marker for the detection of prostate cancer.
Collapse
Affiliation(s)
- Ivanna Ihnatovych
- Department of Physiology and Biophysics, University at Buffalo-State University of New York, Buffalo, New York, United States of America
| | - Neil L. Sielski
- Department of Physiology and Biophysics, University at Buffalo-State University of New York, Buffalo, New York, United States of America
| | - Wilma A. Hofmann
- Department of Physiology and Biophysics, University at Buffalo-State University of New York, Buffalo, New York, United States of America
- * E-mail:
| |
Collapse
|
21
|
Robinson AB, Tangpricha V, Yow E, Gurion R, Schanberg LE, McComsey GA. Vitamin D status is a determinant of atorvastatin effect on carotid intima medial thickening progression rate in children with lupus: an Atherosclerosis Prevention in Pediatric Lupus Erythematosus (APPLE) substudy. Lupus Sci Med 2014; 1:e000037. [PMID: 25396067 PMCID: PMC4225736 DOI: 10.1136/lupus-2014-000037] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 07/31/2014] [Accepted: 08/13/2014] [Indexed: 12/17/2022]
Abstract
Objective Epidemiological associations suggest that vitamin D status may play a role in inflammation and progression of atherosclerosis. Using frozen serum, carotid intima medial thickness (CIMT) measurements and other existing data from the Atherosclerosis Prevention in Pediatric Lupus Erythematosus (APPLE) trial, we assessed interactions between serum 25-hydroxyvitamin D (25(OH)D), atorvastatin randomisation and CIMT progression rate. Methods Participants in the 3-year APPLE trial were randomised to placebo or atorvastatin and CIMT progression rate was measured. Baseline frozen serum was used to measure 25(OH)D concentrations. Mixed effect longitudinal models for CIMT progression at 3 years were used to evaluate interaction between vitamin D deficiency (serum 25(OH)D <20 ng/mL) at baseline and atorvastatin or placebo treatment, adjusting for key systemic lupus erythematosus disease variables and cardiovascular risk factors. Results 201/221 APPLE participants had available samples and were included in this analysis; 61/201 (30%) had vitamin D deficiency at baseline. In adjusted longitudinal modelling, there was significant interaction between baseline vitamin D deficiency and atorvastatin randomisation in 3-year progression of mean-max CIMT. In four out of six carotid segments, there was a greater decrease in mean-max CIMT progression rate in subjects who were treated with atorvastatin compared with placebo if they had baseline serum 25(OH)D levels ≥20 ng/mL. Conclusions Subjects with serum 25(OH)D ≥20 ng/mL had less mean-max CIMT progression following 3 years of atorvastatin treatment. Results from secondary analyses must be interpreted cautiously, but findings suggest that underlying vitamin D deficiency may be involved in response to atorvastatin in atherosclerosis prevention. Trial registration number NCT00065806.
Collapse
Affiliation(s)
- Angela Byun Robinson
- Department of Pediatrics , Rainbow Babies and Children's Hospital/Case Medical Center , Cleveland, Ohio , USA
| | - Vin Tangpricha
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University School of Medicine and Atlanta VA Medical Center, Atlanta, Georgia, USA
| | - Eric Yow
- Duke Clinical Research Institute , Durham, North Carolina , USA
| | - Reut Gurion
- Department of Pediatrics , Rainbow Babies and Children's Hospital/Case Medical Center , Cleveland, Ohio , USA
| | - Laura E Schanberg
- Department of Pediatrics , Duke University Medical Center , Durham, North Carolina , USA
| | - Grace A McComsey
- Department of Medicine and Pediatrics , Case Medical Center , Cleveland, Ohio , USA
| | | |
Collapse
|
22
|
Karur S, Veerappa V, Nanjappa MC. Study of vitamin D deficiency prevalence in acute myocardial infarction. INTERNATIONAL JOURNAL OF CARDIOLOGY. HEART & VESSELS 2014; 3:57-59. [PMID: 29450171 PMCID: PMC5801269 DOI: 10.1016/j.ijchv.2014.03.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 03/08/2014] [Indexed: 11/28/2022]
Abstract
BACKGROUND Deficiency of 25-hydroxy vitamin D [25(OH)D] is a treatable condition that has been associated with coronary artery disease and many of its risk factors. A practical time to assess for 25(OH)D deficiency, and to initiate treatment, is at the time of an acute myocardial infarction(AMI). The prevalence of 25(OH)D deficiency and the characteristics associated with it in patients with acute myocardial infarction are unknown. METHODS In this study 25(OH)D was assessed in 314 subjects enrolled in a Sri Jayadeva Institute of Cardiovascular Science and Research(SJICS&R). Patients enrolled from December 1, 2011 to February 28, 2012 had serum samples sent to a centralized laboratory for analysis using the ELECYS assay. Normal 25(OH)D levels are ≥ 30 ng/ml, and patients with levels < 30 and > 20 ng/ml were classified as insufficient and those with levels ≤ 20 ng/ml as deficient. Vitamin D and other baseline characteristics were analyzed with T-test and chi-squared test. RESULTS Of the 314 enrolled patents, 212 (67.5%) were 25(OH)D deficient and 50(16%) were insufficient, for a total of 83.5% of patients with abnormally low 25(OH)D levels. No significant heterogeneity was observed among age or gender sub groups but 25(OH)D deficiency was more commonly seen in those with lower socioeconomic status, lower activity levels, diabetes, hypercholesterolemia(LDL), hypertriglyceridemia and in smokers. CONCLUSION Vitamin D deficiency is present in most of the patients with acute myocardial infarction and it is associated with many of its risk factors in our study.
Collapse
Affiliation(s)
- Satish Karur
- Department of Cardiology, Sri Jayadeva Institute of Cardiovascular Sciences and Research, Bannerghatta Road, Jayanagar 9th Block, Bangalore, Karnataka 560069, India
| | | | | |
Collapse
|
23
|
Conditional deletion of p53 and Rb in the renin-expressing compartment of the pancreas leads to a highly penetrant metastatic pancreatic neuroendocrine carcinoma. Oncogene 2013; 33:5706-15. [PMID: 24292676 DOI: 10.1038/onc.2013.514] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 09/18/2013] [Accepted: 10/23/2013] [Indexed: 12/17/2022]
Abstract
Efforts to model human pancreatic neuroendocrine tumors (PanNETs) in animals have been moderately successful, with minimal evidence for glucagonomas or metastatic spread. The renin gene, although classically associated with expression in the kidney, is also expressed in many other extrarenal tissues including the pancreas. To induce tumorigenesis within rennin-specific tissues, floxed alleles of p53 and Rb were selectively abrogated using Cre-recombinase driven by the renin promoter. The primary neoplasm generated is a highly metastatic islet cell carcinoma of the pancreas. Lineage tracing identifies descendants of renin-expressing cells as pancreatic alpha cells despite a lack of active renin expression in the mature pancreas. Both primary and metastatic tumors express high levels of glucagon; furthermore, an increased level of glucagon is found in the serum, identifying the pancreatic cancer as a functional glucagonoma. This new model is highly penetrant and exhibits robust frequency of metastases to the lymph nodes and the liver, mimicking human disease, and provides a useful platform for better understanding pancreatic endocrine differentiation and development, as well as islet cell carcinogenesis. The use of fluorescent reporters for lineage tracing of the cells contributing to disease initiation and progression provides an unique opportunity to dissect the timeline of disease, examining mechanisms of the metastatic process, as well as recovering primary and metastatic cells for identifying cooperating mutations that are necessary for progression of disease.
Collapse
|
24
|
Desch M, Hackmayer G, Todorov VT. Identification of ATF2 as a transcriptional regulator of renin gene. Biol Chem 2013; 393:93-100. [PMID: 22628303 DOI: 10.1515/bc-2011-157] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2011] [Accepted: 10/07/2011] [Indexed: 11/15/2022]
Abstract
The cAMP response element (enhCRE) in the distal enhancer regulatory region of renin gene is believed to play a major role in the control of renin transcription. enhCRE binds the CRE-binding protein (CREB), which is the main transcription factor target of cAMP signaling. Using the mouse renin-producing cell line As4.1 we found that activating transcription factor-2 (ATF2) also binds to enhCRE. N-terminal phosphorylation of ATF2, which controls its transactivation, is associated with downregulation of renin gene expression by the cytokine tumor necrosis factor-α (TNFα). The ubiquitin proteasome inhibitor MG132 also phosphorylates ATF2 and inhibits renin expression. Knockdown of ATF2 attenuated the suppression of renin gene expression by MG132, thus demonstrating that ATF2 mediates the inhibitory effect of MG132. In addition, MG132 increased the DNA-binding of ATF2 as well as the ratio of bound ATF2 to CREB. Using ATF2- and CREB-Gal4 fusion protein constructs coupled with luciferase reporter system we showed that ATF2 has a weaker transactivating capacity than CREB. These data suggest that ATF2 represses renin expression by drifting the transcriptional control of renin gene away from CREB. Accordingly, TNFα completely abrogated the cAMP-dependent stimulation of renin gene expression.
Collapse
Affiliation(s)
- Michael Desch
- Institute of Physiology, University of Regensburg, D93040 Regensburg, Germany
| | | | | |
Collapse
|
25
|
Han YH, Kim SZ, Kim SH, Park WH. 2,4-Dinitrophenol induces apoptosis in As4.1 juxtaglomerular cells through rapid depletion of GSH. Cell Biol Int 2013; 32:1536-45. [DOI: 10.1016/j.cellbi.2008.08.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2008] [Revised: 07/10/2008] [Accepted: 08/19/2008] [Indexed: 12/16/2022]
|
26
|
Regulation of mouse-renin gene by apurinic/apyrimidinic-endonuclease 1 (APE1/Ref-1) via recruitment of histone deacetylase 1 corepressor complex. J Hypertens 2012; 30:917-25. [PMID: 22441348 DOI: 10.1097/hjh.0b013e3283525124] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Apurinic/apyrimidinic-endonuclease 1 (APE1) heterozygous mice have chronically elevated blood pressure. Renin of the renin-angiotensin (ANG) system for blood pressure maintenance regulates production of ANG II, a vasoactive hormone. Renin expression and secretion from kidney juxtaglomerular cells are regulated by intracellular calcium. Our objective in this study is to investigate APE1's regulatory role in renin expression. METHODS Effect of APE1 on calcium-mediated modulation of renin expression was examined by real-time reverse transcriptase-PCR, Western analysis and renin promoter-dependent luciferase activity in APE1-knockdown, APE1-overexpressing or control mouse kidney As4.1 cells. Furthermore, coimmunoprecipitation and chromatin immunoprecipitation assays were utilized to examine the association of APE1 with histone deacetylase (HDAC)1 corepressor complex and their recruitment to renin enhancer. Finally, kidney renin mRNA level and plasma-renin activity were measured in wild-type and APE1-heterozygous mice. RESULTS Here we show that APE1 is involved in calcium-mediated repression of renin gene. Our results further indicate that APE1 is a component of HDAC1 corepressor complex bound to renin-enhancer region. Increase in intracellular calcium ion concentration enhances the association of APE1 with HDAC1 corepressor complex and their recruitment to the enhancer region. Furthermore, APE1's N-terminal region is critical for formation and recruitment of the enhancer-bound corepressor complex. Increased renin expression in kidneys and higher plasma-renin activity in APE1 heterozygous mice further supports APE1's corepressor role in vivo. CONCLUSION This study uncovers APE1's function as a novel negative regulator of renin expression, and thereby in blood pressure maintenance.
Collapse
|
27
|
Mayer S, Roeser M, Lachmann P, Ishii S, Suh JM, Harlander S, Desch M, Brunssen C, Morawietz H, Tsai SY, Tsai MJ, Hohenstein B, Hugo C, Todorov VT. Chicken ovalbumin upstream promoter transcription factor II regulates renin gene expression. J Biol Chem 2012; 287:24483-91. [PMID: 22645148 DOI: 10.1074/jbc.m111.329474] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
This study aimed to investigate the possible involvement of the orphan nuclear receptor chicken ovalbumin upstream promoter transcription factor II (COUP-TFII) in the regulation of renin gene expression. COUP-TFII colocalized with renin in the juxtaglomerular cells of the kidney, which are the main source of renin in vivo. Protein-DNA binding studies demonstrated that COUP-TFII binds to an imperfect direct repeat COUP-TFII recognition sequence (termed hereafter proxDR) in the proximal renin promoter. Because cAMP signaling plays a central role in the control of the renin gene expression, we suggested that COUP-TFII may modulate this cAMP effect. Accordingly, knockdown of COUP-TFII in the clonal renin-producing cell lines As4.1 and Calu-6 diminished the stimulation of the renin mRNA expression by cAMP agonists. In addition, the mutation of the proxDR element in renin promoter reporter gene constructs abrogated the inducibility by cAMP. The proxDR sequence was found to be necessary for the function of a proximal renin promoter cAMP-response element (CRE). Knockdown of COUP-TFII or cAMP-binding protein (CREB), which is the archetypal transcription factor binding to CRE, decreased the basal renin gene expression. However, the deficiency of COUP-TFII did not further diminish the renin expression when CREB was knocked down. In agreement with the cell culture studies, mutant mice deficient in COUP-TFII have lower renin expression than their control strain. Altogether our data show that COUP-TFII is involved in the control of renin gene expression.
Collapse
Affiliation(s)
- Sandra Mayer
- Laboratory for Experimental Nephrology and Division of Nephrology, Dresden University of Technology, D-01307 Dresden, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Beierwaltes WH. Hydrogen sulfide, renin, and regulating the second messenger cAMP. Focus on "Hydrogen sulfide regulates cAMP homeostasis and renin degranulation in As4.1 and rat renin-rich kidney cell". Am J Physiol Cell Physiol 2011; 302:C21-3. [PMID: 21998138 DOI: 10.1152/ajpcell.00375.2011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
29
|
Rienstra M, Cheng S, Larson MG, McCabe EL, Booth SL, Jacques PF, Lubitz SA, Yin X, Levy D, Magnani JW, Ellinor PT, Benjamin EJ, Wang TJ. Vitamin D status is not related to development of atrial fibrillation in the community. Am Heart J 2011; 162:538-41. [PMID: 21884873 DOI: 10.1016/j.ahj.2011.06.013] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Accepted: 06/20/2011] [Indexed: 01/19/2023]
Abstract
BACKGROUND Atrial fibrillation (AF) is common and is an important cause of cardiovascular morbidity and mortality. Vitamin D is an emerging risk factor in cardiovascular disease, and vitamin D status is modifiable. Thus, we sought to investigate whether vitamin D status predisposed to the development of AF in a community-based sample. METHODS We evaluated the relation between vitamin D status and development of AF in 2,930 participants of the Framingham Heart Study, Massachusetts, USA, without prevalent AF. The mean age was 65 ± 11 years, and 56% were women. Vitamin D status was assessed by measuring 25-hydroxyvitamin D (25[OH]D) concentrations. Multivariable Cox regression models were adjusted for AF risk factors and season. RESULTS During a mean follow-up of 9.9 years, 425 participants (15%) developed AF. In Cox proportional hazards models, 25(OH)D was not associated with development of AF, with a multivariable-adjusted hazard ratio of 0.99 per SD increment in 25(OH)D levels (95% CI 0.88-1.10, P = .81). Also, no relation was found in models including 25(OH)D as a dichotomous variable (above and below the cohort-specific 20th percentile; P = .59). CONCLUSION In our community-based sample, vitamin D status was not related to incident AF. Our data suggest that vitamin D deficiency does not promote the development of AF in the ambulatory setting.
Collapse
|
30
|
|
31
|
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: 0.9] [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
|
32
|
Rendina D, De Filippo G, Strazzullo P. Should vitamin D status be assessed in patients with congestive heart failure? Nutr Metab Cardiovasc Dis 2010; 20:627-632. [PMID: 20947319 DOI: 10.1016/j.numecd.2010.06.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Revised: 05/14/2010] [Accepted: 06/07/2010] [Indexed: 10/18/2022]
Abstract
For decades the vitamin D biological system has been considered almost exclusively as the master integrator of calcium-phosphate homeostasis and bone metabolism. More recently, the discovery that many human tissues and cells, which do not directly participate in mineral ion homeostasis, express the vitamin D receptor (VDR) and are able to convert the circulating pro-hormone 25-hydroxyvitamin D in its active form, 1,25-dihydroxyvitamin D, has provided new insights into the biological function of this peculiar endocrine system. Several reports have highlighted a variety of human diseases possibly related to vitamin D insufficiency or deficiency (respectively defined as 25-hydroxyvitamin D serum levels lower than 30 or lower than 20 ng/ml). In particular, experimental and observational studies, including those published in this journal issue, support the concept that vitamin D deficiency is involved in the pathogenesis of congestive heart failure, a disabling condition affecting over 15 million of patients worldwide. Considering that circulating levels of 25-hydroxyvitamin D represent the accepted clinical indicator of individual vitamin D status, the measurement of this pro-hormone can be regarded as an appropriate and cost-effective screening tool in patients with chronic heart failure.
Collapse
Affiliation(s)
- D Rendina
- Department of Clinical and Experimental Medicine, Federico II University of Naples Medical School, Naples, Italy
| | | | | |
Collapse
|
33
|
Han YH, Kim SZ, Kim SH, Park WH. Treatment with p38 inhibitor intensifies the death of MG132-treated As4.1 juxtaglomerular cells via the enhancement of GSH depletion. Drug Chem Toxicol 2010; 33:367-76. [PMID: 20545600 DOI: 10.3109/01480540903483458] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
MG132, as a proteasome inhibitor, has been shown to induce apoptotic cell death through the formation of reactive oxygen species (ROS). In this study, we investigated the effects of MG132 and/or MAPK inhibitors on As4.1 juxtaglomerular cells in relation to cell growth, cell death, ROS, and glutathione (GSH) levels. MG132 inhibited the growth of As4.1 cells and induced cell death, accompanied by the loss of mitochondrial membrane potential (MMP; DeltaPsi(m)) and activation of caspase-3 and -8. MG132 increased ROS levels, and GSH depleted cell numbers. The MEK inhibitor slightly reduced cell growth and caspase-3 activity in MG132-treated As4.1 cells and mildly increased MMP (DeltaPsi(m)) loss and O(2)(*-) level. However, it did not increase apoptosis and GSH depletion. The JNK inhibitor did not strongly influence cell growth, cell death, and GSH depletion by MG132, but increased caspase-3 activity, MMP (DeltaPsi(m)) loss, and O(2)(*-) level. Treatment with the p38 inhibitor magnified cell-growth inhibition and apoptosis by MG132. This agent also strongly increased caspase-8 activity, MMP (DeltaPsi(m)) loss, O(2)(*-) level, and GSH depletion. Conclusively, the p38 inhibitor strongly intensified cell death in MG132-treated As4.1 cells. The changes of GSH content by MG132 and/or MAPK inhibitors were closely related to the death of As4.1 cells.
Collapse
Affiliation(s)
- Yong Hwan Han
- Department of Physiology, Medical School, Centers for Healthcare Technology Development, Institute for Medical Sciences, Chonbuk National University, JeonJu, Republic of Korea
| | | | | | | |
Collapse
|
34
|
Han YH, Park WH. Proteasome inhibitor MG132 reduces growth of As4.1 juxtaglomerular cells via caspase-independent apoptosis. Arch Toxicol 2010; 84:689-98. [DOI: 10.1007/s00204-010-0550-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Accepted: 04/19/2010] [Indexed: 10/19/2022]
|
35
|
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: 199] [Impact Index Per Article: 13.3] [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
|
36
|
The changes of reactive oxygen species and glutathione by MG132, a proteasome inhibitor affect As4.1 juxtaglomerular cell growth and death. Chem Biol Interact 2010; 184:319-27. [DOI: 10.1016/j.cbi.2010.01.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2009] [Revised: 01/18/2010] [Accepted: 01/18/2010] [Indexed: 11/23/2022]
|
37
|
Han YH, Park WH. Pyrogallol-induced As4.1 juxtaglomerular cell death is attenuated by MAPK inhibitors via preventing GSH depletion. Arch Toxicol 2010; 84:631-40. [DOI: 10.1007/s00204-010-0526-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2009] [Accepted: 02/09/2010] [Indexed: 12/28/2022]
|
38
|
Abstract
Renin is the key regulated step in the enzymatic cascade that leads to angiotensin generation and the control of blood pressure and fluid/electrolyte homeostasis. In the adult unstressed animal, renin is synthesized and released by renal juxtaglomerular cells. However, when homeostasis is threatened, the number of cells that express and release renin increases and extends beyond the juxtaglomerular area; the result is an increase in circulating renin and the reestablishment of homeostasis. The increase in the number of renin cells, a process termed recruitment, is achieved by dedifferentiation and re-expression of renin in cells derived from the renin lineage. The mechanisms that regulate the related processes of reacquisition of the renin phenotype, renin synthesis, and renin release are beginning to be understood. Numerous studies point to cAMP as a central common factor for the regulation of renin phenotype. In addition, we are seeing the emergence of gap junctions and microRNAs as new and promising avenues for a more complete understanding of the complex regulation of the renin cell.
Collapse
Affiliation(s)
| | - R. Ariel Gomez
- University of Virginia School of Medicine, 409 Lane Road, MR4 Building, Room 2001, Charlottesville, VA 22908, USA.
| |
Collapse
|
39
|
Matsushita K, Morello F, Wu Y, Zhang L, Iwanaga S, Pratt RE, Dzau VJ. Mesenchymal stem cells differentiate into renin-producing juxtaglomerular (JG)-like cells under the control of liver X receptor-alpha. J Biol Chem 2010; 285:11974-82. [PMID: 20118482 DOI: 10.1074/jbc.m109.099671] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Renin is a key enzyme for cardiovascular and renal homeostasis and is produced by highly specialized endocrine cells in the kidney, known as juxtaglomerular (JG) cells. The nature and origin of these cells remain as mysteries. Previously, we have shown that the nuclear hormone receptor liver X receptor-alpha (LXRalpha) is a major transcriptional regulator of the expression of renin, c-myc, and other genes involved with growth/differentiation. In this study we test the hypothesis that LXRalpha plays an important role not only in renin expression but also in renin-containing cell differentiation, specifically from the mesenchymal stem cell (MSC), which may be the origin of the JG cell. Indeed, our data demonstrated that LXRalpha activation by its ligands or cAMP stimulated renin gene expression in both murine and human MSCs. Furthermore, sustained cAMP stimulation of murine MSCs overexpressing LXRalpha led to their differentiation into JG-like cells expressing renin and alpha-smooth muscle actin. These MSC-derived JG-like cells contained renin in secretory granules and released active renin in response to cAMP. In conclusion, the activation of LXRalpha stimulates renin expression and induces MSCs differentiation into renin-secreting, JG-like cells. Our results suggest that the MSC may be the origin of the juxtaglomerular cell and provide insight into novel understanding of pathophysiology of the renin-angiotensin system.
Collapse
Affiliation(s)
- Kenichi Matsushita
- Mandel Center for Hypertension and Atherosclerosis Research, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Tremblay AM, Dufour CR, Ghahremani M, Reudelhuber TL, Giguère V. Physiological genomics identifies estrogen-related receptor alpha as a regulator of renal sodium and potassium homeostasis and the renin-angiotensin pathway. Mol Endocrinol 2010; 24:22-32. [PMID: 19901197 PMCID: PMC5428150 DOI: 10.1210/me.2009-0254] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2009] [Accepted: 10/05/2009] [Indexed: 01/15/2023] Open
Abstract
Estrogen-related receptor alpha (ERRalpha) is an orphan nuclear receptor highly expressed in the kidney, an organ playing a central role in blood pressure regulation through electrolyte homeostasis and the renin-angiotensin system. Physiological analysis revealed that, relative to wild-type mice, ERRalpha null mice are hypotensive despite significant hypernatremia, hypokalemia, and slight hyperreninemia. Using a combination of genome-wide location analysis and expression profiling, we demonstrate that ERRalpha regulates the expression of channels involved in renal Na(+) and K(+) handling (Scnn1a, Atp1a1, Atp1b1) and altered in Bartter syndrome (Bsnd, Kcnq1). In addition, ERRalpha regulates the expression of receptors implicated in the systemic regulation of blood pressure (Ghr, Gcgr, Lepr, Npy1r) and of genes within the renin-angiotensin pathway (Ren1, Agt, Ace2). Our study thus identifies ERRalpha as a pleiotropic regulator of renal control of blood pressure, renal Na(+)/K(+) homeostasis, and renin-angiotensin pathway and suggests that modulation of ERRalpha activity could represent a potential avenue for the management of hypertension.
Collapse
Affiliation(s)
- Annie M Tremblay
- Rosalind and Goodman Cancer Centre, McGill University, Montréal, Québec, Canada
| | | | | | | | | |
Collapse
|
41
|
Itani H, Liu X, Sarsour EH, Goswami PC, Born E, Keen HL, Sigmund CD. Regulation of renin gene expression by oxidative stress. Hypertension 2009; 53:1070-6. [PMID: 19433777 PMCID: PMC2740736 DOI: 10.1161/hypertensionaha.109.130633] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2009] [Accepted: 04/06/2009] [Indexed: 01/20/2023]
Abstract
Increased arterial pressure, angiotensin II, and cytokines each result in feedback inhibition of renin gene expression. Because angiotensin II and cytokines can stimulate reactive oxygen species production, we tested the hypothesis that oxidative stress may be a mediator of this inhibition. Treatment of renin-expressing As4.1 cells with the potent cytokine tumor necrosis factor-alpha caused an increase in the steady-state levels of cellular reactive oxygen species, which was reversed by the antioxidant N-acetylcysteine. Exogenous H(2)O(2) caused a dose- and time-dependent decrease in the level of endogenous renin mRNA and decreased the transcriptional activity of a 4.1-kb renin promoter fused to luciferase, which was maximal when the renin enhancer was present. The effect of H(2)O(2) appeared to be specific to renin, because there was no change in the expression of beta-actin or cyclophilin mRNA or transcriptional activity of the SV40 promoter. The tumor necrosis factor-alpha-induced decrease in renin mRNA was partially reversed by either N-acetylcysteine or panepoxydone, a nuclear factor kappaB (NFkappaB) inhibitor. Interestingly, H(2)O(2) did not induce NFkappaB in As4.1 cells, and panepoxydone had no effect on the downregulation of renin mRNA by H(2)O(2). The transcriptional activity of a cAMP response element-luciferase construct was decreased by both tumor necrosis factor-alpha and H(2)O(2). These data suggest that cellular reactive oxygen species can negatively regulate renin gene expression via an NFkappaB-independent mechanism involving the renin enhancer and inhibiting cAMP response element-mediated transcription. Our data further suggest that tumor necrosis factor-alpha decreases renin expression through both NFkappaB-dependent and NFkappaB-independent mechanisms, the latter involving the production of reactive oxygen species.
Collapse
Affiliation(s)
- Hana Itani
- Molecular and Cellular Biology Graduate Program Roy J. and Lucille A. Carver College of Medicine University of Iowa Iowa City, IA, USA
| | - Xuebo Liu
- Department of Internal Medicine Roy J. and Lucille A. Carver College of Medicine University of Iowa Iowa City, IA, USA
| | - Ehab H. Sarsour
- Department of Radiation Oncology Roy J. and Lucille A. Carver College of Medicine University of Iowa Iowa City, IA, USA
| | - Prabhat C. Goswami
- Department of Radiation Oncology Roy J. and Lucille A. Carver College of Medicine University of Iowa Iowa City, IA, USA
| | - Ella Born
- Department of Internal Medicine Roy J. and Lucille A. Carver College of Medicine University of Iowa Iowa City, IA, USA
| | - Henry L. Keen
- Department of Internal Medicine Roy J. and Lucille A. Carver College of Medicine University of Iowa Iowa City, IA, USA
| | - Curt D. Sigmund
- Department of Internal Medicine Roy J. and Lucille A. Carver College of Medicine University of Iowa Iowa City, IA, USA
- Department of Molecular Physiology and Biophysics Roy J. and Lucille A. Carver College of Medicine University of Iowa Iowa City, IA, USA
- Center on Functional Genomics of Hypertension Roy J. and Lucille A. Carver College of Medicine University of Iowa Iowa City, IA, USA
| |
Collapse
|
42
|
Morris BJ. Fluorescence activated cell sorting of transiently transfected As4.1 cells shows renin enhancer directs on/off switching of renin promoter in vitro. Clin Exp Pharmacol Physiol 2008; 35:367-71. [PMID: 18307722 DOI: 10.1111/j.1440-1681.2008.04879.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
1. The proximal promoter of the renin gene is weak and its activity is influenced by a strong, far-upstream enhancer. This and the ability of renin expression in renal afferent arteriolar cells to be 'recruited' under chronic stimulation is consistent with the on/off switching (variegation) model of gene expression. If true, this would provide an example in which variegation controls a physiologically regulable gene. 2. The present study tested the hypothesis that renin promoter activity may accord with the variegation model, at least in individual juxtaglomerular (mouse As4.1) cells in vitro. 3. As4.1 cells were transiently transfected with constructs containing the mouse renin (Ren-1c) enhancer adjacent to the Ren-1c promoter and a linked reporter gene encoding enhanced green fluorescent protein (EGFP). The EGFP signal from individual cells was monitored by fluorescence activated cell sorting. 4. In the presence of the renin enhancer there was 10-fold higher EGFP expression in transfected cells compared with cells transfected with EGFP constructs containing the promoter alone. There was, moreover, an 8-fold increase in the number of EGFP expressing cells. However, EGFP expression in individual transfected cells was similar in the presence or absence of the enhancer. 5. Results from the in vitro system used suggest that the Ren-1c enhancer does not regulate the rate of promoter activity, but rather increases the probability of achieving an active transcriptional state. Limitations of these findings are discussed.
Collapse
Affiliation(s)
- Brian J Morris
- Basic and Clinical Genomics Laboratory, School of Medical Sciences and Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
43
|
Konoshita T, Fuchs S, Makino Y, Wakahara S, Miyamori I. A proximal direct repeat motif characterized as a negative regulatory element in the human renin gene. J Cell Biochem 2008; 102:1043-50. [PMID: 17455195 DOI: 10.1002/jcb.21341] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The regulation of renin gene expression is thought to be fundamental to regulation of the total renin-angiotensin system. The human renin gene contains a direct repeat (DR) motif AGGGGTCAC-AGGGCCA in the proximal region (-259/-245 bp), which contains similar sequence for nuclear receptor superfamily binding core motif, AGGTCA, and is the most similar to COUP-TFII consensus. The DR motif was evaluated as a functional cis-element with renal cortex and chorio-decidual cells by footprint assay, electromobility shift assay (EMSA) and reporter assay. The DR motif site was protected by footprint analysis with a clear hypersensitive and a minor hypersensitive region in good accordance with the DR of the consensus. One of the binding proteins was strongly suspected to be COUP-TFII-consensus-specific by EMSA. The DNA/protein complexes obtained with nuclear extract of renin producing cells could be completely blocked by homologous competitor and strongly blocked by the second-half mutant oligonucleotide of the DR motif but not by the first-half mutant oligonucleotide. Finally, the transcriptional activity of second-half mutant construct is slightly elevated and that first-half mutant construct is significantly stronger by twofold compared with wild type construct in reporter assay. These findings suggest that the DR motif site of the human renin gene functions as a negative regulatory element involved in a twofold repression of transcription and that member(s) of nucleic receptor superfamily bind the site and play important roles in the human renin gene expression with a possibility that one of the binding protein is COUP-TFII.
Collapse
Affiliation(s)
- Tadashi Konoshita
- Third Department of Internal Medicine, Fukui University School of Medicine, 23-3, Shimoaizuki, Matsuoka, 910-1193, Japan.
| | | | | | | | | |
Collapse
|
44
|
Wang TJ, Pencina MJ, Booth SL, Jacques PF, Ingelsson E, Lanier K, Benjamin EJ, D'Agostino RB, Wolf M, Vasan RS. Vitamin D deficiency and risk of cardiovascular disease. Circulation 2008; 117:503-11. [PMID: 18180395 PMCID: PMC2726624 DOI: 10.1161/circulationaha.107.706127] [Citation(s) in RCA: 1646] [Impact Index Per Article: 96.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Vitamin D receptors have a broad tissue distribution that includes vascular smooth muscle, endothelium, and cardiomyocytes. A growing body of evidence suggests that vitamin D deficiency may adversely affect the cardiovascular system, but data from longitudinal studies are lacking. METHODS AND RESULTS We studied 1739 Framingham Offspring Study participants (mean age 59 years; 55% women; all white) without prior cardiovascular disease. Vitamin D status was assessed by measuring 25-dihydroxyvitamin D (25-OH D) levels. Prespecified thresholds were used to characterize varying degrees of 25-OH D deficiency (< 15 ng/mL, < 10 ng/mL). Multivariable Cox regression models were adjusted for conventional risk factors. Overall, 28% of individuals had levels < 15 ng/mL, and 9% had levels < 10 ng/mL. During a mean follow-up of 5.4 years, 120 individuals developed a first cardiovascular event. Individuals with 25-OH D < 15 ng/mL had a multivariable-adjusted hazard ratio of 1.62 (95% confidence interval 1.11 to 2.36, P=0.01) for incident cardiovascular events compared with those with 25-OH D > or = 15 ng/mL. This effect was evident in participants with hypertension (hazard ratio 2.13, 95% confidence interval 1.30 to 3.48) but not in those without hypertension (hazard ratio 1.04, 95% confidence interval 0.55 to 1.96). There was a graded increase in cardiovascular risk across categories of 25-OH D, with multivariable-adjusted hazard ratios of 1.53 (95% confidence interval 1.00 to 2.36) for levels 10 to < 15 ng/mL and 1.80 (95% confidence interval 1.05 to 3.08) for levels < 10 ng/mL (P for linear trend=0.01). Further adjustment for C-reactive protein, physical activity, or vitamin use did not affect the findings. CONCLUSIONS Vitamin D deficiency is associated with incident cardiovascular disease. Further clinical and experimental studies may be warranted to determine whether correction of vitamin D deficiency could contribute to the prevention of cardiovascular disease.
Collapse
|
45
|
Han YH, Kim SZ, Kim SH, Park WH. Intracellular GSH level is a factor in As4.1 juxtaglomerular cell death by arsenic trioxide. J Cell Biochem 2008; 104:995-1009. [DOI: 10.1002/jcb.21685] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
46
|
Pentz ES, Lopez MLSS, Cordaillat M, Gomez RA. Identity of the renin cell is mediated by cAMP and chromatin remodeling: an in vitro model for studying cell recruitment and plasticity. Am J Physiol Heart Circ Physiol 2007; 294:H699-707. [PMID: 18055510 DOI: 10.1152/ajpheart.01152.2007] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The renin-angiotensin system (RAS) regulates blood pressure and fluid-electrolyte homeostasis. A key step in the RAS cascade is the regulation of renin synthesis and release by the kidney. We and others have shown that a major mechanism to control renin availability is the regulation of the number of cells capable of making renin. The kidney possesses a pool of cells, mainly in its vasculature but also in the glomeruli, capable of switching from smooth muscle to endocrine renin-producing cells when homeostasis is threatened. The molecular mechanisms governing the ability of these cells to turn the renin phenotype on and off have been very difficult to study in vivo. We, therefore, developed an in vitro model in which cells of the renin lineage are labeled with cyan fluorescent protein and cells actively making renin mRNA are labeled with yellow fluorescent protein. The model allowed us to determine that it is possible to culture cells of the renin lineage for numerous passages and that the memory to express the renin gene is maintained in culture and can be reenacted by cAMP and chromatin remodeling (histone H4 acetylation) at the cAMP-responsive element in the renin gene.
Collapse
Affiliation(s)
- Ellen Steward Pentz
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | | | | | | |
Collapse
|
47
|
Yuan W, Pan W, Kong J, Zheng W, Szeto FL, Wong KE, Cohen R, Klopot A, Zhang Z, Li YC. 1,25-dihydroxyvitamin D3 suppresses renin gene transcription by blocking the activity of the cyclic AMP response element in the renin gene promoter. J Biol Chem 2007; 282:29821-30. [PMID: 17690094 DOI: 10.1074/jbc.m705495200] [Citation(s) in RCA: 355] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
We have shown that 1,25-dihydroxyvitamin D(3) (1,25(OH)(2)D(3)) down-regulates renin expression. To explore the molecular mechanism, we analyzed the mouse Ren-1c gene promoter by luciferase reporter assays. Deletion analysis revealed two DNA fragments from -2,725 to -2,647 (distal fragment) and from -117 to +6 (proximal fragment) that are sufficient to mediate the repression. Mutation of the cAMP response element (CRE) in the distal fragment blunted forskolin stimulation as well as 1,25(OH)(2)D(3) inhibition of the transcriptional activity, suggesting the involvement of CRE in 1,25(OH)(2)D(3)-induced suppression. EMSA revealed that 1,25(OH)(2)D(3) markedly inhibited nuclear protein binding to the CRE in the promoter. ChIP and GST pull-down assays demonstrated that liganded VDR blocked the binding of CREB to the CRE by directly interacting with CREB with the ligand-binding domain, and the VDR-mediated repression can be rescued by CREB, CBP, or p300 overexpression. These data indicate that 1,25(OH)(2)D(3) suppresses renin gene expression at least in part by blocking the formation of CRE-CREB-CBP complex.
Collapse
Affiliation(s)
- Weihua Yuan
- Department of Medicine, Division of Biological Sciences, the University of Chicago, Chicago, Illinois 60637, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Fryer RM, Rakestraw PA, Nakane M, Dixon D, Banfor PN, Koch KA, Wu-Wong JR, Reinhart GA. Differential Inhibition of Renin mRNA Expression by Paricalcitol and Calcitriol in C57/BL6 Mice. ACTA ACUST UNITED AC 2007; 106:p76-81. [PMID: 17622742 DOI: 10.1159/000104875] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2006] [Accepted: 04/26/2007] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS Vitamin D receptor activators (VDRAs) may suppress renin expression and VDR-mediated renin inhibitors may offer a novel mechanism to control the RAS. METHODS We delineated the effects of paricalcitol and calcitriol on PTH, renin, and iCa(2+) in C57/BL6 mice administered vehicle, paricalcitol, or calcitriol (0.01, 0.03, 0.10, 0.33, 1.0 microg/kg s.c.) 3 days/week for 9 days. RESULTS Paricalcitol produced PTH suppression from 0.03 to 1.0 microg/kg (values between 9.7 +/- 3.3 and 20.7 +/- 4.7 pg/ml; vehicle = 88.0 +/- 16.9) and elicited dose-dependent reductions in renin/GAPDH expression at 0.33 and 1.0 microg/kg (0.037 +/- 0.002, 0.027 +/- 0.003; vehicle = 0.054 +/- 0.003) but produced no increases iCa(2+) at any dose tested. Calcitrol produced PTH suppression at all doses tested (between 6.4 +/- 1.2 and 29.5 +/- 17.2 pg/ml) and renin suppression at 0.10, 0.33, and 1.0 microg/kg (0.029 +/- 0.002, 0.031 +/- 0.003, and 0.038 +/- 0.02). However, at 0.33 and 1.0 mg/kg, calcitriol produced increases iCa(2+) (1.31 +/- 0.03 and 1.48 +/- 0.02 mmol/l; vehicle = 1.23 +/- 0.02 mmol/l). CONCLUSIONS Paricalcitol produces significant, dose-dependent suppression of renin expression in the absence of hypercalcemia at doses 10-fold above those necessary for PTH suppression. Calcitriol also produced suppression of renin at doses at least 10-fold above those required for PTH suppression, but increases in iCa(2+) were observed at doses only 3-fold above those necessary to elicit renin suppression.
Collapse
Affiliation(s)
- Ryan M Fryer
- Integrative Pharmacology, Global Pharmaceutical Research and Development, Abbott Laboratories, Abbott Park, IL 60064-6119, USA.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Matsushita K, Zhang Z, Pratt RE, Dzau VJ. Molecular mechanism of juxtaglomerular cell hyperplasia: a unifying hypothesis. ACTA ACUST UNITED AC 2007; 1:164-8. [DOI: 10.1016/j.jash.2007.02.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2007] [Revised: 02/12/2007] [Accepted: 02/19/2007] [Indexed: 01/02/2023]
|
50
|
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
|