Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-38991008

RESUMO

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.
Hypertension ; 2024 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-38989586

RESUMO

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 ATAC-seq, single-cell RNA sequencing, CUT&Tag, and chromatin immunoprecipitation sequencing for H3K27ac 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.

3.
bioRxiv ; 2024 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-38585851

RESUMO

Renin is a crucial enzyme involved in the regulation of blood pressure and electrolyte balance. It has been shown that renin expressing cells arise from the Foxd1+ stromal progenitors, however the factors involved in guiding Foxd1+ cells towards the renin-secreting cell fate remain poorly understood. Tcf21, also known as Pod1 or Capsulin, is a bHLH transcription factor that is expressed in the metanephric mesenchyme and plays a crucial role in kidney development. We have previously shown that deletion of Tcf21 in Foxd1+ cells ( Foxd1 Cre/+ ;Tcf21 f/f ) results in paucity of vascular mural cells and in disorganized renal arterial tree with fewer, shorter, and thinner arterioles. Here, we sought to examine the relationship between Tcf21 and renin cells during kidney development and test whether Tcf21 is implicated in the regulation of juxtaglomerular cell differentiation. Immunostaining for renin demonstrated that kidneys of Foxd1 Cre/+ ;Tcf21 f/f have fewer renin-positive spots at E16.5 and E18.5 compared with controls. In-situ hybridization for renin mRNA showed reduced expression in Foxd1 Cre/+ ;Tcf21 f/f kidneys at E14.5, E16.5, and E18.5. Together, these data suggest that stromal expression of Tcf21 is required for the emergence of renin cells. To dissect the role of Tcf21 in juxtaglomerular (JG) cells, we deleted Tcf21 upon renin promoter activation ( Ren1 dCre/+ ;Tcf21 f/f ). Interestingly, the Ren1 dCre/+ ;Tcf21 f/f kidney showed normal arterial tree at E16.5 identical to controls. Furthermore, inactivation of Tcf21 upon renin expression did not alter kidney morphology in two- and four-month-old mice. Finally, expression renin mRNA was similar between Ren1 dCre/+ ;Tcf21 f/f and controls at 2 months. Taken together, these findings suggest that Tcf21 expression in Foxd1+ cells is essential for specifying the fate of these cells into juxtaglomerular cells. However, once renin cell identity is assumed, Tcf21 is dispensable. Uncovering the regulation of Foxd1+ cells and their derivatives, including the JG cell lineage, is crucial for understanding the mechanisms underlying renal vasculature formation.

5.
Am J Physiol Renal Physiol ; 325(2): F188-F198, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37345845

RESUMO

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.


Assuntos
Nefropatias , Renina , Camundongos , Animais , Renina/genética , Renina/metabolismo , Fator de Transcrição GATA3/genética , Fator de Transcrição GATA3/metabolismo , Rim/metabolismo , Glomérulos Renais/metabolismo , Nefropatias/patologia , Zinco/metabolismo
6.
Acta Physiol (Oxf) ; 238(4): e14014, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37309075

RESUMO

AIM: Ureteral obstruction leads to significant changes in kidney renin expression. It is unclear whether those changes are responsible for the progression of kidney damage, repair, or regeneration. In the current study, we aimed to elucidate the contribution of renin-producing cells (RPCs) and the cells of the renin lineage (CoRL) towards kidney damage and regeneration using a model of partial and reversible unilateral ureteral obstruction (pUUO) in neonatal mice. METHODS: Renin cells are progenitors for other renal cell types collectively called CoRL. We labeled the CoRL with green fluorescent protein (GFP) using genetic approaches. We performed lineage tracing to analyze the changes in the distribution of CoRL during and after the release of obstruction. We also ablated the RPCs and CoRL by cell-specific expression of Diphtheria Toxin Sub-unit A (DTA). Finally, we evaluated the kidney damage and regeneration during and after the release of obstruction in the absence of CoRL. RESULTS: In the obstructed kidneys, there was a 163% increase in the renin-positive area and a remarkable increase in the distribution of GFP+ CoRL. Relief of obstruction abrogated these changes. In addition, DTA-expressing animals did not respond to pUUO with increased RPCs and CoRL. Moreover, reduction in CoRL significantly compromised the kidney's ability to recover from the damage after the release of obstruction. CONCLUSIONS: CoRL play a role in the regeneration of the kidneys post-relief of obstruction.


Assuntos
Rim , Obstrução Ureteral , Camundongos , Animais , Rim/metabolismo , Renina/metabolismo , Animais Recém-Nascidos , Obstrução Ureteral/metabolismo , Camundongos Transgênicos , Regeneração
7.
bioRxiv ; 2023 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-36711565

RESUMO

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.

8.
JCI Insight ; 6(24)2021 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-34762601

RESUMO

Inhibitors of the renin-angiotensin system (RAS) are widely used to treat hypertension. Using mice harboring fluorescent cell lineage tracers, single-cell RNA-Seq, and long-term inhibition of RAS in both mice and humans, we found that deletion of renin or inhibition of the RAS leads to concentric thickening of the intrarenal arteries and arterioles. This severe disease was caused by the multiclonal expansion and transformation of renin cells from a classical endocrine phenotype to a matrix-secretory phenotype: the cells surrounded the vessel walls and induced the accumulation of adjacent smooth muscle cells and extracellular matrix, resulting in blood flow obstruction, focal ischemia, and fibrosis. Ablation of the renin cells via conditional deletion of ß1 integrin prevented arteriolar hypertrophy, indicating that renin cells are responsible for vascular disease. Given these findings, prospective morphological studies in humans are necessary to determine the extent of renal vascular damage caused by the widespread use of inhibitors of the RAS.


Assuntos
Hipertensão/fisiopatologia , Rim/irrigação sanguínea , Sistema Renina-Angiotensina/fisiologia , Animais , Humanos , Camundongos
9.
J Clin Invest ; 128(11): 4787-4803, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30130256

RESUMO

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.


Assuntos
Epigênese Genética , Código das Histonas , Histonas/metabolismo , Homeostase , Subunidade 1 do Complexo Mediador/metabolismo , Renina/biossíntese , Células-Tronco/metabolismo , Animais , Histonas/genética , Subunidade 1 do Complexo Mediador/genética , Camundongos , Camundongos Transgênicos , Renina/genética , Células-Tronco/citologia
10.
Hypertension ; 70(1): 119-128, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28533331

RESUMO

Experimental or spontaneous genomic mutations of the renin-angiotensin system or its pharmacological inhibition in early life leads to renal abnormalities, including poorly developed renal medulla, papillary atrophy, hydronephrosis, inability to concentrate the urine, polyuria, polydipsia, renal failure, and anemia. At the core of such complex phenotype is the presence of unique vascular abnormalities: the renal arterioles do not branch or elongate properly and they have disorganized, concentric hypertrophy. This lesion has been puzzling because it is often found in hypertensive individuals whereas mutant or pharmacologically inhibited animals are hypotensive. Remarkably, when renin cells are ablated with diphtheria toxin, the vascular hypertrophy does not occur, suggesting that renin cells per se may contribute to the vascular disease. To test this hypothesis, on a Ren1c-/- background, we generated mutant mice with reporter expression (Ren1c-/-;Ren1c-Cre;R26R.mTmG and Ren1c-/-;Ren1c-Cre;R26R.LacZ) to trace the fate of reninnull cells. To assess whether reninnull cells maintain their renin promoter active, we used Ren1c-/-;Ren1c-YFP mice that transcribe YFP (yellow fluorescent protein) directed by the renin promoter. We also followed the expression of Akr1b7 and miR-330-5p, markers of cells programmed for the renin phenotype. Contrary to what we expected, reninnull cells did not die or disappear. Instead, they survived, increased in number along the renal arterial tree, and maintained an active molecular memory of the myoepitheliod renin phenotype. Furthermore, null cells of the renin lineage occupied the walls of the arteries and arterioles in a chaotic, directionless pattern directly contributing to the concentric arterial hypertrophy.


Assuntos
Hipertensão , Medula Renal/metabolismo , Artéria Renal , Sistema Renina-Angiotensina/fisiologia , Renina , Animais , Hipertensão/genética , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Hipertrofia , Medula Renal/patologia , Medula Renal/fisiopatologia , Camundongos , Artéria Renal/metabolismo , Artéria Renal/patologia , Artéria Renal/fisiopatologia , Renina/genética , Renina/metabolismo
11.
J Am Soc Nephrol ; 26(1): 67-80, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24904090

RESUMO

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.


Assuntos
Regulação da Expressão Gênica , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Sistema Justaglomerular/metabolismo , Animais , Sítios de Ligação , Comunicação Celular , Linhagem da Célula , Proliferação de Células , Cromossomos Artificiais Bacterianos , Deleção de Genes , Genes Reporter , Células-Tronco Hematopoéticas/citologia , Rim/irrigação sanguínea , Rim/metabolismo , Camundongos , Camundongos Knockout , Microcirculação , Mutação , Miócitos de Músculo Liso/citologia , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Regiões Promotoras Genéticas , Renina/genética
12.
Am J Pathol ; 184(12): 3226-38, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25307343

RESUMO

Obstructive nephropathy, the leading cause of kidney failure in children, can be anatomic or functional. The underlying causes of functional hydronephrosis are not well understood. miRNAs, which are small noncoding RNAs, regulate gene expression at the post-transcriptional level. We found that miR-145-5p, a member of the miR-143/145 cluster that is highly expressed in smooth muscle cells of the renal vasculature, was present in the pelvicalyceal system and the ureter. To evaluate whether the miR-143/145 cluster is involved in urinary tract function we performed morphologic, functional, and gene expression studies in mice carrying a whole-body deletion of miR-143/145. miR-143/145-deficient mice developed hydronephrosis, characterized by severe papillary atrophy and dilatation of the pelvicalyceal system without obvious physical obstruction. Moreover, mutant mice showed abnormal ureteral peristalsis. The number of ureter contractions was significantly higher in miR-143/145-deficient mice. Peristalsis was replaced by incomplete, short, and more frequent contractions that failed to completely propagate in a proximal-distal direction. Microarray analysis showed 108 differentially expressed genes in ureters of miR-143/145-deficient mice. Ninety genes were up-regulated and 18 genes were down-regulated, including genes with potential regulatory roles in smooth muscle contraction and extracellular matrix-receptor interaction. We show that miR-143/145 are important for the normal peristalsis of the ureter and report an association between the expression of these miRNAs and hydronephrosis.


Assuntos
Deleção de Genes , Hidronefrose/genética , MicroRNAs/genética , Animais , Nitrogênio da Ureia Sanguínea , Creatinina/sangue , Modelos Animais de Doenças , Feminino , Heterozigoto , Hidronefrose/metabolismo , Hibridização In Situ , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Músculo Liso/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Peristaltismo , Ureter/metabolismo
13.
Nat Commun ; 5: 3273, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24549417

RESUMO

The cell of origin and triggering events for leukaemia are mostly unknown. Here we show that the bone marrow contains a progenitor that expresses renin throughout development and possesses a B-lymphocyte pedigree. This cell requires RBP-J to differentiate. Deletion of RBP-J in these renin-expressing progenitors enriches the precursor B-cell gene programme and constrains lymphocyte differentiation, facilitated by H3K4me3 activating marks in genes that control the pre-B stage. Mutant cells undergo neoplastic transformation, and mice develop a highly penetrant B-cell leukaemia with multi-organ infiltration and early death. These renin-expressing cells appear uniquely vulnerable as other conditional models of RBP-J deletion do not result in leukaemia. The discovery of these unique renin progenitors in the bone marrow and the model of leukaemia described herein may enhance our understanding of normal and neoplastic haematopoiesis.


Assuntos
Células da Medula Óssea/metabolismo , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Leucemia de Células B/etiologia , Leucemia Experimental/etiologia , Renina/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Medula Óssea/patologia , Células da Medula Óssea/patologia , Epigênese Genética , Feminino , Hematopoese , Humanos , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/genética , Leucemia de Células B/metabolismo , Leucemia de Células B/patologia , Leucemia Experimental/metabolismo , Leucemia Experimental/patologia , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Baço/patologia , Adulto Jovem
14.
Pediatr Nephrol ; 29(4): 721-6, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24337407

RESUMO

Renin-expressing cells appear early in the embryo and are distributed broadly throughout the body as organogenesis ensues. Their appearance in the metanephric kidney is a relatively late event in comparison with other organs such as the fetal adrenal gland. The functions of renin cells in extra renal tissues remain to be investigated. In the kidney, they participate locally in the assembly and branching of the renal arterial tree and later in the endocrine control of blood pressure and fluid-electrolyte homeostasis. Interestingly, this endocrine function is accomplished by the remarkable plasticity of renin cell descendants along the kidney arterioles and glomeruli which are capable of reacquiring the renin phenotype in response to physiological demands, increasing circulating renin and maintaining homeostasis. Given that renin cells are sensors of the status of the extracellular fluid and perfusion pressure, several signaling mechanisms (ß-adrenergic receptors, Notch pathway, gap junctions and the renal baroreceptor) must be coordinated to ensure the maintenance of renin phenotype--and ultimately the availability of renin--during basal conditions and in response to homeostatic threats. Notably, key transcriptional (Creb/CBP/p300, RBP-J) and posttranscriptional (miR-330, miR125b-5p) effectors of those signaling pathways are prominent in the regulation of renin cell identity. The next challenge, it seems, would be to understand how those factors coordinate their efforts to control the endocrine and contractile phenotypes of the myoepithelioid granulated renin-expressing cell.


Assuntos
Rim/embriologia , Rim/metabolismo , Organogênese/fisiologia , Renina/metabolismo , Células-Tronco/metabolismo , Animais , Humanos
15.
Am J Physiol Renal Physiol ; 302(1): F29-37, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21993888

RESUMO

We have shown that microRNAs (miRNAs) are necessary for renin cell specification and kidney vascular development. Here, we used a screening strategy involving microarray and in silico analyses, along with in situ hybridization and in vitro functional assays to identify miRNAs important for renin cell identity. Microarray studies using vascular smooth muscle cells (SMCs) of the renin lineage and kidney cortex under normal conditions and after reacquisition of the renin phenotype revealed that of 599 miRNAs, 192 were expressed in SMCs and 234 in kidney cortex. In silico analysis showed that the highly conserved miR-330 and miR-125b-5p have potential binding sites in smoothelin (Smtn), calbindin 1, smooth muscle myosin heavy chain, α-smooth muscle actin, and renin genes important for the myoepithelioid phenotype of the renin cell. RT-PCR studies confirmed miR-330 and miR-125b-5p expression in kidney and SMCs. In situ hybridization revealed that under normal conditions, miR-125b-5p was expressed in arteriolar SMCs and in juxtaglomerular (JG) cells. Under conditions that induce reacquisition of the renin phenotype, miR-125b-5p was downregulated in arteriolar SMCs but remained expressed in JG cells. miR-330, normally absent, was expressed exclusively in JG cells of treated mice. In vitro functional studies showed that overexpression of miR-330 inhibited Smtn expression in SMCs. On the other hand, miR-125b-5p increased Smtn expression, whereas its inhibition reduced Smtn expression. Our results demonstrate that miR-330 and miR-125b-5p are markers of JG cells and have opposite effects on renin lineage cells: one inhibiting and the other favoring their smooth muscle phenotype.


Assuntos
Sistema Justaglomerular/metabolismo , MicroRNAs/genética , Renina/genética , Animais , Linhagem da Célula , Proteínas do Citoesqueleto/biossíntese , Bases de Dados Genéticas , Feminino , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Musculares/biossíntese , Músculo Liso Vascular/fisiologia , Fenótipo , Renina/metabolismo
16.
Biochim Biophys Acta ; 1790(12): 1587-91, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19800395

RESUMO

We propose a model in which cell loss in the aging brain is seen as a root cause of behavioral changes that compromise quality of life, including the onset of generalized anxiety disorder, in elderly individuals. According to this model, as stem cells in neurogenic regions of the adult brain lose regenerative capacity, worn-out, dead, or damaged neurons fail to be replaced, leaving gaps in function. As most replacement involves inhibitory interneurons, either directly or indirectly, the net result is the acquisition over time of a hyper-excitable state. The stress axis is subserved by all three neurogenic regions in the adult brain, making it particularly susceptible to these age-dependent changes. We outline a molecular mechanism by which hyper-excitation of the stress axis in turn activates the tumor suppressor p53. This reinforces the loss of stem cell proliferative capacity and interferes with the feedback mechanism by which the glucocorticoid receptor turns off neuroendocrine pathways and resets the axis.


Assuntos
Envelhecimento/fisiologia , Ansiedade/etiologia , Encéfalo/fisiopatologia , Proteína Supressora de Tumor p53/fisiologia , Adulto , Animais , Humanos , Sistema Hipotálamo-Hipofisário/fisiopatologia , Modelos Biológicos , Neurogênese/genética , Neurogênese/fisiologia , Sistema Hipófise-Suprarrenal/fisiopatologia , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/fisiologia , Estresse Psicológico/genética , Estresse Psicológico/fisiopatologia , Proteína Supressora de Tumor p53/genética
17.
Exp Gerontol ; 44(1-2): 93-100, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-18598747

RESUMO

In higher organisms dependent on the regenerative ability of tissue stem cells to maintain tissue integrity throughout adulthood, the failure of stem cells to replace worn out, dead, or damaged cells is seen as one mechanism that limits life span. In these organisms, tumor suppressors such as p53 are central participants in the control of longevity because they regulate stem cell proliferation. Several recent reports have identified p53 as a longevity gene in organisms such as Caenorhabditis elegans and Drosophila melanogaster, which lack proliferative stem cells in all but the germline and have relatively short life spans. This has forced us to reevaluate the role of p53 in the control of life span. We discuss how p53 might regulate longevity in both long- and short-lived species by controlling the activity of insulin-like molecules that operate in proliferating and non-proliferating compartments of adult somatic tissues. We also discuss the hierarchical structure of life span regulation where loss of p53 has life span extending effects. Finally, we suggest a molecular mechanism by which p53 might facilitate the response to severe nutrient deprivation that allows metabolically active cells to survive periods of starvation. Paradoxically, loss of p53 function in these cells would compromise life span.


Assuntos
Insulina/metabolismo , Transdução de Sinais/fisiologia , Somatomedinas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Caenorhabditis elegans , Proliferação de Células , Sobrevivência Celular , Drosophila melanogaster , Humanos , Expectativa de Vida , Camundongos , Estresse Oxidativo , Inanição/metabolismo , Células-Tronco/fisiologia
18.
Neurobiol Aging ; 30(3): 483-97, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17850928

RESUMO

The question of whether or not stem cell loss drives aging in the brain has not been fully resolved. Here, we used mice over-expressing the short isoform of p53 (DeltaNp53 or p44) as a model of aging to gain insight into the cellular mechanisms underlying age-related functional deficits in the brain. By BrdU labeling, we observed an accelerated decline in the number of subventricular zone proliferating cells with age in p44Tg mice compared to mice with normal p53 expression. A 2-3-fold reduction in the number of slowly dividing stem cells was evident in the subventricular zone of 9-12-month-old p44Tg mice, but not in younger p44Tg mice or in normal mice. Consequently, the supply of new olfactory bulb neurons was also reduced. The number and size of neurospheres generated from subventricular zone cells from p44Tg mice was significantly reduced, and cells derived from these neurospheres had limited self-renewal and amplification capacities. At the cellular level, p44 lengthened the cell cycle and affected cell cycle reentry properties, evident by an increased proportion of cells in G0. At the functional level, p44 expression resulted in impaired olfactory discrimination in 15-16-month-old mice. This phenotype is driven by constitutive activation of p53 and constitutive expression of p21(Cip1/waf1) in neural stem cells. Our results demonstrate that p53 plays a crucial role in the maintenance of the regenerative capacity of the brain by regulating the proliferation of stem and progenitor cells.


Assuntos
Encéfalo/fisiologia , Diferenciação Celular/fisiologia , Neurônios/fisiologia , Regeneração/fisiologia , Células-Tronco/fisiologia , Proteína Supressora de Tumor p53/genética , Animais , Encéfalo/citologia , Diferenciação Celular/genética , Proliferação de Células , Células Cultivadas , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/citologia , Regeneração/genética , Células-Tronco/citologia , Proteína Supressora de Tumor p53/fisiologia
19.
Biochem Biophys Res Commun ; 331(3): 828-33, 2005 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-15865938

RESUMO

In the adult mammalian brain, neuronal turnover continues to replenish cells in existing neuronal circuits, such as those involved either in odor discrimination or in learning and memory, throughout life. With age, however, the capacity for neurogenesis diminishes and these functions become impaired. Neuronal turnover is a two-step process, which first generates excess neuronal progenitors and then eliminates all but the few that differentiate into fully functional neurons. This process requires a fine balance between cell proliferation and cell death. Altered activity of the tumor suppressor p53 can upset this balance by affecting the rate of cell proliferation, but not the rate of cell death, in neurogenic regions of the adult brain. Genetically engineered mice in which p53 activity is increased demonstrate that premature loss of neurogenic capacity is linked to accelerated organismal aging.


Assuntos
Ventrículos Cerebrais/citologia , Giro Denteado/citologia , Neurônios/citologia , Proteína Supressora de Tumor p53/fisiologia , Envelhecimento/fisiologia , Animais , Morte Celular/fisiologia , Proliferação de Células , Genes p53 , Camundongos , Degeneração Neural/fisiopatologia , Bulbo Olfatório/citologia , Células-Tronco/fisiologia
20.
Biol Reprod ; 68(1): 67-76, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12493697

RESUMO

In neurons, arc (activity regulated, cytoskeleton associated) is an immediate early gene (IEG) that is rapidly and transiently induced by excitatory stimulation. It is believed to mediate rapid strengthening of signaling structures at activated synaptic sites. Unlike most IEGs, arc does not encode nuclear transcription factor, but an effector molecule that associates with the actin cytoskeleton. Cytoskeletal rearrangements of microtubule- and actin-based networks that occur at activated synapses also take place in similar fashion during the formation of the acrosome, the site of regulated exocytosis at fertilization. In this paper, arc is reported to be highly expressed both at the RNA and protein levels in postmeiotic germ cells in the testis of adult mice. Immunofluorescence studies reveal that arc is first present in the perinuclear region of round, elongating, and elongate spermatids, where it colocalizes with the developing acrosome. In isolated mature sperm, arc is present in the acrosomal region of the sperm head, the centriole region of the neck, and the principal piece of the tail. Arc is lost to varying degrees during sperm capacitation and in acrosome-reacted sperm. Phalloidin staining of mature sperm cells reveals an overlapping pattern of filamentous-actin and arc expression. These results support a role for arc and the actin cytoskeleton in the acrosome formation, the sperm acrosome reaction, and in sperm cell motility.


Assuntos
Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Espermatozoides/metabolismo , Acrossomo/metabolismo , Reação Acrossômica , Actinas/metabolismo , Animais , Encéfalo/metabolismo , Citoesqueleto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genes Precoces , Glicoproteínas/metabolismo , Imuno-Histoquímica , Masculino , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Capacitação Espermática , Cauda do Espermatozoide/metabolismo , Espermátides/metabolismo , Espermatogênese , Testículo/crescimento & desenvolvimento , Testículo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...