Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
Add more filters

Publication year range
1.
Article in English | MEDLINE | ID: mdl-38991008

ABSTRACT

Fate mapping and genetic manipulation of renin cells have relied on either non-inducible Cre lines that can introduce developmental effects of gene deletion or BAC transgene-based inducible models that may be prone to spurious and/or ectopic gene expression. To circumvent these problems, we generated an inducible mouse model in which CreERT2 is under the control of the endogenous Akr1b7 gene, an independent marker of renin cells that is expressed in a few extrarenal tissues. We confirmed the proper expression of Cre using Akr1b7CreERT2/+;R26RmTmG/+ mice in which Akr1b7+/renin+ cells become GFP+ upon tamoxifen administration. In embryos and neonates, GFP was found in Juxtaglomerular cells, along the arterioles, and in the mesangium, and in adults, GFP was present mainly in Juxtaglomerular cells. In mice treated with captopril and a low salt diet to induce recruitment of renin cells, GFP extended along the afferent arterioles and in the mesangium. We generated Akr1b7CreERT2/+;Ren1cFl/-;R26RmTmG/+ mice to conditionally delete renin in adult mice and found a marked reduction in kidney renin mRNA and protein, and mean arterial pressure in mutant animals. When subjected to a homeostatic threat, mutant mice were unable to recruit renin+ cells. Most importantly, these mice developed concentric vascular hypertrophy ruling out potential developmental effects on the vasculature due to the lack of renin. We conclude that Akr1b7CreERT2 mice constitute an excellent model for the fate mapping of renin cells and for the spatial and temporal control of gene expression in renin cells.

2.
Am J Physiol Renal Physiol ; 325(2): F188-F198, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37345845

ABSTRACT

Renin cells are precursors for other cell types in the kidney and show high plasticity in postnatal life in response to challenges to homeostasis. Our previous single-cell RNA-sequencing studies revealed that the dual zinc-finger transcription factor Gata3, which is important for cell lineage commitment and differentiation, is expressed in mouse renin cells under normal conditions and homeostatic threats. We identified a potential Gata3-binding site upstream of the renin gene leading us to hypothesize that Gata3 is essential for renin cell identity. We studied adult mice with conditional deletion of Gata3 in renin cells: Gata3fl/fl;Ren1dCre/+ (Gata3-cKO) and control Gata3fl/fl;Ren1d+/+ counterparts. Gata3 immunostaining revealed that Gata3-cKO mice had significantly reduced Gata3 expression in juxtaglomerular, mesangial, and smooth muscle cells, indicating a high degree of deletion of Gata3 in renin lineage cells. Gata3-cKO mice exhibited a significant increase in blood urea nitrogen, suggesting hypovolemia and/or compromised renal function. By immunostaining, renin-expressing cells appeared very thin compared with their normal plump shape in control mice. Renin cells were ectopically localized to Bowman's capsule in some glomeruli, and there was aberrant expression of actin-α2 signals in the mesangium, interstitium, and Bowman's capsule in Gata3-cKO mice. Distal tubules showed dilated morphology with visible intraluminal casts. Under physiological threat, Gata3-cKO mice exhibited a lower increase in mRNA levels than controls. Hematoxylin-eosin, periodic acid-Schiff, and Masson's trichrome staining showed increased glomerular fusion, absent cubical epithelial cells in Bowman's capsule, intraglomerular aneurysms, and tubular dilation. In conclusion, our results indicate that Gata3 is crucial to the identity of cells of the renin lineage.NEW & NOTEWORTHY Gata3, a dual zinc-finger transcription factor, is responsible for the identity and localization of renin cells in the kidney. Mice with a conditional deletion of Gata3 in renin lineage cells have abnormal kidneys with juxtaglomerular cells that lose their characteristic location and are misplaced outside and around arterioles and glomeruli. The fundamental role of Gata3 in renin cell development offers a new model to understand how transcription factors control cell location, function, and pathology.


Subject(s)
Kidney Diseases , Renin , Mice , Animals , Renin/genetics , Renin/metabolism , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Kidney/metabolism , Kidney Glomerulus/metabolism , Kidney Diseases/pathology , Zinc/metabolism
3.
Circ Res ; 128(7): 887-907, 2021 04 02.
Article in English | MEDLINE | ID: mdl-33793334

ABSTRACT

Renin cells are essential for survival perfected throughout evolution to ensure normal development and defend the organism against a variety of homeostatic threats. During embryonic and early postnatal life, they are progenitors that participate in the morphogenesis of the renal arterial tree. In adult life, they are capable of regenerating injured glomeruli, control blood pressure, fluid-electrolyte balance, tissue perfusion, and in turn, the delivery of oxygen and nutrients to cells. Throughout life, renin cell descendants retain the plasticity or memory to regain the renin phenotype when homeostasis is threatened. To perform all of these functions and maintain well-being, renin cells must regulate their identity and fate. Here, we review the major mechanisms that control the differentiation and fate of renin cells, the chromatin events that control the memory of the renin phenotype, and the major pathways that determine their plasticity. We also examine how chronic stimulation of renin cells alters their fate leading to the development of a severe and concentric hypertrophy of the intrarenal arteries and arterioles. Lastly, we provide examples of additional changes in renin cell fate that contribute to equally severe kidney disorders.


Subject(s)
Hypertension/etiology , Kidney/cytology , Renin/physiology , Animals , Arterioles/embryology , Blood Pressure/physiology , Cell Communication , Cell Differentiation , Cell Plasticity , Chromatin/physiology , Chromatin Assembly and Disassembly/physiology , Connexins/physiology , Homeostasis , Humans , Integrins/physiology , Juxtaglomerular Apparatus/cytology , Kidney/blood supply , Kidney/embryology , Kidney Glomerulus/physiology , Mice , MicroRNAs/physiology , Phenotype , Regeneration/physiology , Renal Artery , Renin/metabolism , Renin-Angiotensin System/physiology , Stem Cells/physiology , Water-Electrolyte Balance
5.
Semin Cell Dev Biol ; 91: 132-146, 2019 07.
Article in English | MEDLINE | ID: mdl-29879472

ABSTRACT

The kidney vasculature has a unique and complex architecture that is central for the kidney to exert its multiple and essential physiological functions with the ultimate goal of maintaining homeostasis. An appropriate development and coordinated assembly of the different vascular cell types and their association with the corresponding nephrons is crucial for the generation of a functioning kidney. In this review we provide an overview of the renal vascular anatomy, histology, and current knowledge of the embryological origin and molecular pathways involved in its development. Understanding the cellular and molecular mechanisms involved in renal vascular development is the first step to advance the field of regenerative medicine.


Subject(s)
Kidney/blood supply , Neovascularization, Physiologic/physiology , Nephrons/blood supply , Renal Artery/anatomy & histology , Renal Veins/anatomy & histology , Animals , Gene Expression Regulation, Developmental , Humans , Kidney/embryology , Kidney/metabolism , Neovascularization, Physiologic/genetics , Nephrons/embryology , Nephrons/metabolism , Regenerative Medicine/methods , Regenerative Medicine/trends , Renal Artery/embryology , Renal Artery/metabolism , Renal Veins/embryology , Renal Veins/metabolism
6.
Am J Physiol Renal Physiol ; 321(3): F378-F388, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34338032

ABSTRACT

Developmentally heterogeneous renin-expressing cells serve as progenitors for mural, glomerular, and tubular cells during nephrogenesis and are collectively termed renin lineage cells (RLCs). In this study, we quantified different renal vascular and tubular cell types based on specific markers and assessed proliferation and de novo differentiation in the RLC population. We used kidney sections of mRenCre-mT/mG mice throughout nephrogenesis. Marker positivity was evaluated in whole digitalized sections. At embryonic day 16, RLCs appeared in the developing kidney, and the expression of all stained markers in RLCs was observed. The proliferation rate of RLCs did not differ from the proliferation rate of non-RLCs. RLCs expanded mainly by de novo differentiation (neogenesis). Fractions of RLCs originating from the stromal progenitors of the metanephric mesenchyme (renin-producing cells, vascular smooth muscle cells, and mesangial cells) decreased during nephrogenesis. In contrast, aquaporin-2-positive RLCs in the collecting duct system, which embryonically emerges almost exclusively from the ureteric bud, expanded postpartum. The cubilin-positive RLC fraction in the proximal tubule, deriving from the cap mesenchyme, remained constant. In summary, RLCs were continuously detectable in the vascular and tubular compartments of the kidney during nephrogenesis. Therein, various patterns of RLC differentiation that depend on the embryonic origin of the cells were identified.NEW & NOTEWORTHY The unifying feature of the renal renin lineage cells (RLCs) is their origin from renin-expressing progenitors. RLCs evolve to an embryologically heterogeneous large population in structures with different ancestry. RLCs are also targets for the widely used renin-angiotensin-system blockers, which modulate their phenotype. Unveiling the different differentiation patterns of RLCs in the developing kidney contributes to understanding changes in their cell fate in response to homeostatic challenges and the use of antihypertensive drugs.


Subject(s)
Cell Differentiation/physiology , Kidney Glomerulus/metabolism , Kidney/metabolism , Mesangial Cells/metabolism , Renin/metabolism , Animals , Cell Lineage/physiology , Mesoderm/metabolism , Mice , Stem Cells/metabolism
7.
Article in English | MEDLINE | ID: mdl-33385525

ABSTRACT

The renin-angiotensin system (RAS) evolved early among vertebrates and remains functioning throughout the vertebrate phylogeny and has adapted to various environments. The RAS is crucial for the regulation of blood pressure, fluid-electrolyte balance and tissue homeostasis. The RAS is also expressed during early ontogeny in renal and extra-renal tissues, and exerts unique vascular growth and differentiation functions. In this brief review, we describe advances from molecular-genetic and whole animal approaches and discuss similarities and unique aspects of the RAS in the context of embryonic development and vertebrates' phylogeny.


Subject(s)
Endothelium, Vascular/metabolism , Renin-Angiotensin System/physiology , Renin/metabolism , Animals , Arteries/metabolism , Arterioles/metabolism , Blood Pressure , Cell Differentiation , Chickens , Homeostasis , Humans , Kidney/metabolism , Phylogeny , RNA, Messenger/metabolism , Water-Electrolyte Balance
8.
Kidney Int ; 98(3): 630-644, 2020 09.
Article in English | MEDLINE | ID: mdl-32446934

ABSTRACT

Kidney function and blood pressure homeostasis are regulated by purinergic signaling mechanisms. These autocrine/paracrine signaling pathways are initiated by the release of cellular ATP, which influences kidney hemodynamics and steady-state renin secretion from juxtaglomerular cells. However, the mechanism responsible for ATP release that supports tonic inputs to juxtaglomerular cells and regulates renin secretion remains unclear. Pannexin 1 (Panx1) channels localize to both afferent arterioles and juxtaglomerular cells and provide a transmembrane conduit for ATP release and ion permeability in the kidney and the vasculature. We hypothesized that Panx1 channels in renin-expressing cells regulate renin secretion in vivo. Using a renin cell-specific Panx1 knockout model, we found that male Panx1 deficient mice exhibiting a heightened activation of the renin-angiotensin-aldosterone system have markedly increased plasma renin and aldosterone concentrations, and elevated mean arterial pressure with altered peripheral hemodynamics. Following ovariectomy, female mice mirrored the male phenotype. Furthermore, constitutive Panx1 channel activity was observed in As4.1 renin-secreting cells, whereby Panx1 knockdown reduced extracellular ATP accumulation, lowered basal intracellular calcium concentrations and recapitulated a hyper-secretory renin phenotype. Moreover, in response to stress stimuli that lower blood pressure, Panx1-deficient mice exhibited aberrant "renin recruitment" as evidenced by reactivation of renin expression in pre-glomerular arteriolar smooth muscle cells. Thus, renin-cell Panx1 channels suppress renin secretion and influence adaptive renin responses when blood pressure homeostasis is threatened.


Subject(s)
Connexins , Renin , Adenosine Triphosphate , Animals , Blood Pressure , Connexins/genetics , Female , Homeostasis , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics
9.
Clin Sci (Lond) ; 134(13): 1763-1774, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32619009

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.


Subject(s)
CCCTC-Binding Factor/metabolism , Kidney/metabolism , Renin/metabolism , Animals , CCCTC-Binding Factor/genetics , Chromatin , Female , Kidney/anatomy & histology , Male , Mice , Mice, Knockout , Renin/genetics
11.
Gen Comp Endocrinol ; 296: 113533, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32561435

ABSTRACT

Renin or a renin-like enzyme evolved in ancestral vertebrates and is conserved along the vertebrate phylogeny. The ontogenic development of renin, however, is not well understood in nonmammalian vertebrates. We aimed to determine the expression patterns and relative abundance of renin mRNA in pre- and postnatal chickens (Gallus gallus, White Leghorn breed). Embryonic day 13 (E13) embryos show renal tubules, undifferentiated mesenchymal structures, and a small number of developing glomeruli. Maturing glomeruli are seen in post-hatch day 4 (D4) and day 30 (D30) kidneys, indicating that nephrogenic activity still exists in kidneys of 4-week-old chickens. In E13 embryos, renin mRNA measured by quantitative polymerase chain reaction in the adrenal glands is equivalent to the expression in the kidneys, whereas in post-hatch D4 and D30 maturing chicks, renal renin expressions increased 2-fold and 11-fold, respectively. In contrast, relative renin expression in the adrenals became lower than in the kidneys. Furthermore, renin expression is clearly visible by in situ hybridization in the juxtaglomerular (JG) area in D4 and D30 chicks, but not in E13 embryos. The results suggest that in chickens, renin evolved in both renal and extrarenal organs at an early stage of ontogeny and, with maturation, became localized to the JG area. Clear JG structures are not morphologically detectable in E13 embryos, but are visible in 30-day-old chicks, supporting this concept.


Subject(s)
Chickens/genetics , Gene Expression Regulation , Renin/genetics , Animals , Chick Embryo , Juxtaglomerular Apparatus/cytology , Juxtaglomerular Apparatus/metabolism , Organogenesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Renin/metabolism , Renin-Angiotensin System
12.
Am J Physiol Regul Integr Comp Physiol ; 316(5): R640-R650, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30943054

ABSTRACT

Formation of the metanephric kidney requires coordinated interaction among the stroma, ureteric bud, and cap mesenchyme. The transcription factor Foxd1, a specific marker of renal stromal cells, is critical for normal kidney development. The prorenin receptor (PRR), a receptor for renin and prorenin, is also an accessory subunit of the vacuolar proton pump V-ATPase. Global loss of PRR is embryonically lethal in mice, indicating an essential role of the PRR in embryonic development. Here, we report that conditional deletion of the PRR in Foxd1+ stromal progenitors in mice (cKO) results in neonatal mortality. The kidneys of surviving mice show reduced expression of stromal markers Foxd1 and Meis1 and a marked decrease in arterial and arteriolar development with the subsequent decreased number of glomeruli, expansion of Six2+ nephron progenitors, and delay in nephron differentiation. Intrarenal arteries and arterioles in cKO mice were fewer and thinner and showed a marked decrease in the expression of renin, suggesting a central role for the PRR in the development of renin-expressing cells, which in turn are essential for the proper formation of the renal arterial tree. We conclude that stromal PRR is crucial for the appropriate differentiation of the renal arterial tree, which in turn may restrict excessive expansion of nephron progenitors to promote a coordinated and proper morphogenesis of the nephrovascular structures of the mammalian kidney.


Subject(s)
Kidney/growth & development , Nephrons/metabolism , Organogenesis/physiology , Receptors, Cell Surface/metabolism , Animals , Cell Differentiation/physiology , Gene Expression Regulation/physiology , Kidney/metabolism , Mice, Transgenic , Renin/metabolism , Stem Cells/cytology , Transcription Factors/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Prorenin Receptor
15.
Clin Sci (Lond) ; 132(23): 2519-2545, 2018 12 12.
Article in English | MEDLINE | ID: mdl-30442812

ABSTRACT

Congenital obstructive nephropathy is a major cause of chronic kidney disease (CKD) in children. The contribution of changes in the identity of renal cells to the pathology of obstructive nephropathy is poorly understood. Using a partial unilateral ureteral obstruction (pUUO) model in genetically modified neonatal mice, we traced the fate of cells derived from the renal stroma, cap mesenchyme, ureteric bud (UB) epithelium, and podocytes using Foxd1Cre, Six2Cre, HoxB7Cre, and Podocyte.Cre mice respectively, crossed with double fluorescent reporter (membrane-targetted tandem dimer Tomato (mT)/membrane-targetted GFP (mG)) mice. Persistent obstruction leads to a significant loss of tubular epithelium, rarefaction of the renal vasculature, and decreased renal blood flow (RBF). In addition, Forkhead Box D1 (Foxd1)-derived pericytes significantly expanded in the interstitial space, acquiring a myofibroblast phenotype. Degeneration of Sine Oculis Homeobox Homolog 2 (Six2) and HoxB7-derived cells resulted in significant loss of glomeruli, nephron tubules, and collecting ducts. Surgical release of obstruction resulted in striking regeneration of tubules, arterioles, interstitium accompanied by an increase in blood flow to the level of sham animals. Contralateral kidneys with remarkable compensatory response to kidney injury showed an increase in density of arteriolar branches. Deciphering the mechanisms involved in kidney repair and regeneration post relief of obstruction has potential therapeutic implications for infants and children and the growing number of adults suffering from CKD.


Subject(s)
Cell Differentiation , Cell Lineage , Cell Proliferation , Hydronephrosis/prevention & control , Kidney/surgery , Regeneration , Ureteral Obstruction/surgery , Animals , Animals, Newborn , Cell Tracking/methods , Disease Models, Animal , Fibrosis , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Hydronephrosis/genetics , Hydronephrosis/metabolism , Hydronephrosis/pathology , Kidney/metabolism , Kidney/pathology , Kidney/physiopathology , Mice, Transgenic , Neovascularization, Physiologic , Oxidative Stress , Phenotype , Renal Circulation , Signal Transduction , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Ureteral Obstruction/genetics , Ureteral Obstruction/metabolism , Ureteral Obstruction/pathology
16.
Physiology (Bethesda) ; 31(1): 25-33, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26661526

ABSTRACT

Renin progenitors appear early and are found in multiple tissues throughout the embryo. Besides their well known role in blood pressure and fluid homeostasis, renin progenitors participate in tissue morphogenesis, repair, and regeneration, and may integrate immune and endocrine responses. In the bone marrow, renin cells offer clues to understand normal and neoplastic hematopoiesis.


Subject(s)
Homeostasis/physiology , Renin-Angiotensin System/physiology , Renin/metabolism , Animals , Blood Pressure/physiology , Bone Marrow/metabolism , Bone Marrow/physiology , Hematopoiesis/physiology , Humans , Neoplasms/metabolism , Neoplasms/physiopathology
17.
Kidney Int ; 92(6): 1419-1432, 2017 12.
Article in English | MEDLINE | ID: mdl-28688581

ABSTRACT

Renin lineage cells (RLCs) serve as a progenitor cell reservoir during nephrogenesis and after renal injury. The maintenance mechanisms of the RLC pool are still poorly understood. Since RLCs were also identified as a progenitor cell population in bone marrow we first considered that these may be their source in the kidney. However, transplantation experiments in adult mice demonstrated that bone marrow-derived cells do not give rise to RLCs in the kidney indicating their non-hematopoietic origin. Therefore we tested whether RLCs develop in the kidney through neogenesis (de novo differentiation) from cells that have never expressed renin before. We used a murine model to track neogenesis of RLCs by flow cytometry, histochemistry, and intravital kidney imaging. During nephrogenesis RLCs first appear at e14, form a distinct population at e16, and expand to reach a steady state level of 8-10% of all kidney cells in adulthood. De novo differentiated RLCs persist as a clearly detectable population through embryogenesis until at least eight months after birth. Pharmacologic stimulation of renin production with enalapril or glomerular injury induced the rate of RLC neogenesis in the adult mouse kidney by 14% or more than three-fold, respectively. Thus, the renal RLC niche is constantly filled by local de novo differentiation. This process could be stimulated consequently representing a new potential target to beneficially influence repair and regeneration after kidney injury.


Subject(s)
Acute Kidney Injury/pathology , Cell Differentiation/physiology , Glomerular Mesangium/physiology , Regeneration/drug effects , Renin/metabolism , Stem Cells/physiology , Acute Kidney Injury/chemically induced , Animals , Biopsy , Bone Marrow Cells/metabolism , Bone Marrow Cells/physiology , Bone Marrow Transplantation/methods , Cell Lineage/drug effects , Cell Lineage/physiology , Enalapril/pharmacology , Glomerular Mesangium/cytology , Glomerular Mesangium/drug effects , Glomerular Mesangium/pathology , Humans , Lipopolysaccharides/toxicity , Mesangial Cells/drug effects , Mesangial Cells/pathology , Mesangial Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Renin/genetics , Stem Cells/drug effects
18.
J Am Soc Nephrol ; 27(7): 1984-95, 2016 07.
Article in English | MEDLINE | ID: mdl-26534925

ABSTRACT

The close relationship between endothelial and hematopoietic precursors during early development of the vascular system suggested the possibility of a common yet elusive precursor for both cell types. Whether similar or related progenitors for endothelial and hematopoietic cells are present during organogenesis is unclear. Using inducible transgenic mice that specifically label endothelial and hematopoietic precursors, we performed fate-tracing studies combined with colony-forming assays and crosstransplantation studies. We identified a progenitor, marked by the expression of helix-loop-helix transcription factor stem cell leukemia (SCL/Tal1). During organogenesis of the kidney, SCL/Tal1(+) progenitors gave rise to endothelium and blood precursors with multipotential colony-forming capacity. Furthermore, appropriate morphogenesis of the kidney vasculature, including glomerular capillary development, arterial mural cell coating, and lymphatic vessel development, required sphingosine 1-phosphate (S1P) signaling via the G protein-coupled S1P receptor 1 in these progenitors. Overall, these results show that SCL/Tal1(+) progenitors with hemogenic capacity originate and differentiate within the early embryonic kidney by hemovasculogenesis (the concomitant formation of blood and vessels) and underscore the importance of the S1P pathway in vascular development.


Subject(s)
Blood Vessels/embryology , Kidney/blood supply , Organogenesis , Receptors, Lysosphingolipid/physiology , Animals , Endothelium, Vascular/embryology , Mice , Sphingosine-1-Phosphate Receptors
19.
Kidney Int ; 90(2): 244-246, 2016 08.
Article in English | MEDLINE | ID: mdl-27418087

ABSTRACT

The kidney vasculature is extremely complex, yet, despite recent progress, our understanding of how the renal vascular system develops is limited. By using advanced tissue engineering techniques and in vivo and in vitro depletion of specific populations of endothelial cell precursors, Halt et al. have identified a CD146-expressing precursor as an important player in the development of the renal vasculature.


Subject(s)
Endothelial Cells , Kidney/blood supply , Endothelium , Endothelium, Vascular , Humans
20.
J Am Soc Nephrol ; 26(1): 67-80, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24904090

ABSTRACT

Recombination signal binding protein for Ig-κJ region (RBP-J), the major downstream effector of Notch signaling, is necessary to maintain the number of renin-positive juxtaglomerular cells and the plasticity of arteriolar smooth muscle cells to re-express renin when homeostasis is threatened. We hypothesized that RBP-J controls a repertoire of genes that defines the phenotype of the renin cell. Mice bearing a bacterial artificial chromosome reporter with a mutated RBP-J binding site in the renin promoter had markedly reduced reporter expression at the basal state and in response to a homeostatic challenge. Mice with conditional deletion of RBP-J in renin cells had decreased expression of endocrine (renin and Akr1b7) and smooth muscle (Acta2, Myh11, Cnn1, and Smtn) genes and regulators of smooth muscle expression (miR-145, SRF, Nfatc4, and Crip1). To determine whether RBP-J deletion decreased the endowment of renin cells, we traced the fate of these cells in RBP-J conditional deletion mice. Notably, the lineage staining patterns in mutant and control kidneys were identical, although mutant kidneys had fewer or no renin-expressing cells in the juxtaglomerular apparatus. Microarray analysis of mutant arterioles revealed upregulation of genes usually expressed in hematopoietic cells. Thus, these results suggest that RBP-J maintains the identity of the renin cell by not only activating genes characteristic of the myo-endocrine phenotype but also, preventing ectopic gene expression and adoption of an aberrant phenotype, which could have severe consequences for the control of homeostasis.


Subject(s)
Gene Expression Regulation , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Juxtaglomerular Apparatus/metabolism , Animals , Binding Sites , Cell Communication , Cell Lineage , Cell Proliferation , Chromosomes, Artificial, Bacterial , Gene Deletion , Genes, Reporter , Hematopoietic Stem Cells/cytology , Kidney/blood supply , Kidney/metabolism , Mice , Mice, Knockout , Microcirculation , Mutation , Myocytes, Smooth Muscle/cytology , Oligonucleotide Array Sequence Analysis , Phenotype , Promoter Regions, Genetic , Renin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL