Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 12(1): 18211, 2022 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-36307508

RESUMO

Genome editing tools such as CRISPR/Cas9 enable the rapid and precise manipulation of genomes. CRISPR-based genome editing has greatly simplified the study of gene function in cell lines, but its widespread use has also highlighted challenges of reproducibility. Phenotypic variability among different knockout clones of the same gene is a common problem confounding the establishment of robust genotype-phenotype correlations. Optimized genome editing protocols to enhance reproducibility include measures to reduce off-target effects. However, even if current state-of-the-art protocols are applied phenotypic variability is frequently observed. Here we identify heterogeneity of wild-type cells as an important and often neglected confounding factor in genome-editing experiments. We demonstrate that isolation of individual wild-type clones from an apparently homogenous stable cell line uncovers significant phenotypic differences between clones. Strikingly, we observe hundreds of differentially regulated transcripts (477 up- and 306 downregulated) when comparing two populations of wild-type cells. Furthermore, we show a variety of cellular and biochemical alterations in different wild-type clones in a range that is commonly interpreted as biologically relevant in genome-edited cells. Heterogeneity of wild-type cells thus contributes to variability in genome-edited cells when these are generated through isolation of clones. We show that the generation of monoclonal isogenic wild-type cells prior to genomic manipulation reduces phenotypic variability. We therefore propose to generate matched isogenic control cells prior to genome editing to increase reproducibility.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Sistemas CRISPR-Cas/genética , Reprodutibilidade dos Testes , Edição de Genes/métodos , Linhagem Celular , Células Cultivadas
3.
J Am Soc Nephrol ; 26(1): 48-54, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24904091

RESUMO

Mesangial cell injury has a major role in many CKDs. Because renin-positive precursor cells give rise to mesangial cells during nephrogenesis, this study tested the hypothesis that the same phenomenon contributes to glomerular regeneration after murine experimental mesangial injury. Mesangiolysis was induced by administration of an anti-mesangial cell serum in combination with LPS. In enhanced green fluorescent protein-reporter mice with constitutively labeled renin lineage cells, the size of the enhanced green fluorescent protein-positive area in the glomerular tufts increased after mesangial injury. Furthermore, we generated a novel Tet-on inducible triple-transgenic LacZ reporter line that allowed selective labeling of renin cells along renal afferent arterioles of adult mice. Although no intraglomerular LacZ expression was detected in healthy mice, about two-thirds of the glomerular tufts became LacZ positive during the regenerative phase after severe mesangial injury. Intraglomerular renin descendant LacZ-expressing cells colocalized with mesangial cell markers α8-integrin and PDGF receptor-ß but not with endothelial, podocyte, or parietal epithelial cell markers. In contrast with LacZ-positive cells in the afferent arterioles, LacZ-positive cells in the glomerular tuft did not express renin. These data demonstrate that extraglomerular renin lineage cells represent a major source of repopulating cells for reconstitution of the intraglomerular mesangium after injury.


Assuntos
Linhagem da Célula , Mesângio Glomerular/metabolismo , Rim/lesões , Renina/fisiologia , Animais , Animais Geneticamente Modificados , Doxiciclina/administração & dosagem , Enalapril/administração & dosagem , Feminino , Genes Reporter , Proteínas de Fluorescência Verde/metabolismo , Imageamento Tridimensional , Glomérulos Renais/metabolismo , Óperon Lac , Lipopolissacarídeos/química , Masculino , Camundongos , Camundongos Transgênicos , Renina/metabolismo , Células-Tronco/citologia
4.
Am J Physiol Renal Physiol ; 304(5): F578-84, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23303406

RESUMO

On the basis of evidence that within the adult kidney, the aldo-keto reductase AKR1B7 (aldo-keto reductase family 1, member 7, also known as mouse vas deferens protein, MVDP) is selectively expressed in renin-producing cells, we aimed to define a possible role of AKR1B7 for the regulation and function of renin cells in the kidney. We could confirm colocalization and corecruitment of renin and of AKR1B7 in wild-type kidneys. Renin cells in AKR1B7-deficient kidneys showed normal morphology, numbers, and intrarenal distribution. Plasma renin concentration (PRC) and renin mRNA levels of AKR1B7-deficient mice were normal at standard chow and were lowered by a high-salt diet directly comparable to wild-type mice. Treatment with a low-salt diet in combination with an angiotensin-converting enzyme inhibitor strongly increased PRC and renin mRNA in a similar fashion both in AKR1B7-deficient and wild-type mice. Under this condition, we also observed a strong retrograde recruitment of renin-expressing cell along the preglomerular vessels, however, without a difference between AKR1B7-deficient and wild-type mice. The isolated perfused mouse kidney model was used to study the acute regulation of renin secretion by ANG II and by perfusion pressure. Regarding these parameters, no differences were observed between AKR1B7-deficient and wild-type kidneys. In summary, our data suggest that AKR1B7 is not of major relevance for the regulation of renin production and secretion in spite of its striking coregulation with renin expression.


Assuntos
Aldeído Redutase/metabolismo , Rim/metabolismo , Renina/metabolismo , Aldeído Redutase/genética , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Enalapril/farmacologia , Rim/efeitos dos fármacos , Camundongos , Camundongos Knockout , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Renina/genética , Cloreto de Sódio na Dieta
5.
Hypertension ; 61(2): 400-7, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23297374

RESUMO

Chronic challenge of renin-angiotensin causes recruitment of renin-producing cells in the kidney along the media layer of afferent arterioles and hypertrophy of cells in the juxtaglomerular apparatus. This study aimed to define the role of nitric oxide (NO) with regard to the recruitment pattern of renin-producing cells and to the possible pathways along which NO could act. We considered the hypothesis that endothelium-derived NO acts via NO-sensitive guanylate cyclase. Mice were treated with low-salt diet in combination with the angiotensin I-converting enzyme inhibitor enalapril for 3 weeks, which led to a 13-fold increase in renin expression associated with marked recruitment of renin cells in afferent arterioles and hypertrophy of the juxtaglomerular apparatus in wild-type mice. In wild-type mice additionally treated with the nonselective NO synthase inhibitor L-NAME, the recruitment of renin-expressing cells along the afferent arterioles was absent and juxtaglomerular hypertrophy was diminished. An almost identical attenuation of renin cell recruitment as with L-NAME treatment in wild-type mice was found in mice lacking the endothelial isoform of NO synthase. Treatment of mice lacking NO-sensitive guanylate cyclase in renin-expressing cells and preglomerular smooth muscle cells with low-salt diet in combination with the angiotensin I-converting enzyme inhibitor enalapril for 3 weeks produced juxtaglomerular hypertrophy like in wild-type mice, but no recruitment in afferent arterioles. These findings suggest that endothelium-derived NO and concomitant formation of cGMP in preglomerular renin cell precursors supports recruitment of renin-expressing cells along preglomerular vessels, but not in the juxtaglomerular apparatus.


Assuntos
Endotélio Vascular/metabolismo , Guanilato Ciclase/metabolismo , Rim/metabolismo , Óxido Nítrico/metabolismo , Renina/metabolismo , Transdução de Sinais/fisiologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Sistema Justaglomerular/metabolismo , Rim/efeitos dos fármacos , Camundongos , Camundongos Knockout , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Transdução de Sinais/efeitos dos fármacos
6.
Am J Physiol Renal Physiol ; 302(10): F1278-85, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22357914

RESUMO

The salt intake of an organism controls the number of renin-producing cells in the kidney by yet undefined mechanisms. This study aimed to assess a possible mediator role of preglomerular blood pressure in the control of renin expression by oral salt intake. We used wild-type (WT) mice and mice lacking angiotensin II type 1a receptors (AT(1a)-/-) displaying an enhanced salt sensitivity to renin expression. In WT kidneys, we found renin-expressing cells at the ends of all afferent arterioles. A low-salt diet (0.02%) led to a moderate twofold increase in renin-expressing cells along afferent arterioles. In AT(1a)-/- mice, lowering of salt content led to a 12-fold increase in renin expression. Here, the renin-expressing cells were distributed along the preglomerular vascular tree in a typical distal-to-proximal distribution gradient which was most prominent at high salt intake and was obliterated at low salt intake by the appearance of renin-expressing cells in proximal parts of the preglomerular vasculature. While lowering of salt intake produced only a small drop in blood pressure in WT mice, the marked reduction of systolic blood pressure in AT(1a)-/- mice was accompanied by the disappearance of the distribution gradient from afferent arterioles to arcuate arteries. Unilateral renal artery stenosis in AT(1a)-/- mice on a normal salt intake produced a similar distribution pattern of renin-expressing cells as did low salt intake. Conversely, increasing blood pressure by administration of the NOS inhibitor N-nitro-l-arginine methyl ester or of the adrenergic agonist phenylephrine in AT(1a)-/- mice kept on low salt intake produced a similar distribution pattern of renin-producing cells as did normal salt intake alone. These findings suggest that changes in preglomerular blood pressure may be an important mediator of the influence of salt intake on the number and distribution of renin-producing cells in the kidney.


Assuntos
Pressão Sanguínea/fisiologia , Rim/fisiologia , Receptor Tipo 1 de Angiotensina/genética , Renina/genética , Cloreto de Sódio na Dieta/farmacologia , Agonistas de Receptores Adrenérgicos alfa 1/farmacologia , Animais , Arteríolas/fisiologia , Pressão Sanguínea/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Homeostase/fisiologia , Sistema Justaglomerular/fisiologia , Rim/irrigação sanguínea , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , NG-Nitroarginina Metil Éster/farmacologia , Fenilefrina/farmacologia , Receptor Tipo 1 de Angiotensina/metabolismo , Renina/metabolismo
7.
J Am Soc Nephrol ; 22(6): 1031-40, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21597036

RESUMO

Deletion of the gap-junction-forming protein connexin40 leads to renin-dependent hypertension in mice, but whether observed human variants in connexin40, such as A96S, promote hypertension is unknown. Here, we generated mice with the A96S variant in the mouse connexin40 gene. Although mice homozygous for the A96S mutations had normal expression patterns of connexin40 in the kidney, they were hypertensive, had sixfold higher plasma renin concentrations, and had 40% higher levels of renin mRNA than controls. Renin-expressing cells were aberrantly located outside the media layer of afferent arterioles, and increased renal perfusion pressure did not inhibit renin secretion from kidneys isolated from homozygous A96S mice. Treatment with a low-salt diet in combination with an ACE inhibitor increased renin mRNA levels, plasma renin concentrations, and the number of aberrantly localized renin-producing cells. Taken together, these findings suggest that the A96S mutation in connexin40 leads to renin-dependent hypertension in mice. Modulation of renin secretion by BP critically depends on functional connexin40; with the A96S mutation, the aberrant extravascular localization of renin-secreting cells in the kidney likely impairs the pressure-mediated inhibition of renin secretion.


Assuntos
Conexinas/genética , Hipertensão/genética , Hipertensão/fisiopatologia , Mutação/genética , Renina/fisiologia , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Animais , Dieta Hipossódica , Modelos Animais de Doenças , Feminino , Junções Comunicantes/fisiologia , Células HeLa , Humanos , Hipertensão/terapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transfecção , Resultado do Tratamento , Proteína alfa-5 de Junções Comunicantes
8.
Am J Physiol Renal Physiol ; 301(1): F71-7, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21389089

RESUMO

During nephrogenesis, renin expression shifts from large renal arteries toward smaller vessels in a defined spatiotemporal pattern, finally becoming restricted to the juxtaglomerular position. Chronic stimulation in adult kidneys leads to a recruitment of renin expression in the upstream vasculature. The mechanisms that control this characteristic switch-on and switch-off in the immature and adult kidney are not well-understood. Previous studies in mice with juxtaglomerular cell-specific deletion of the adenylyl cyclase-stimulatory G protein Gsα suggested that signaling along the cAMP pathway plays an essential role for renin expression during nephrogenesis and in the adult kidney. To identify the Gsα-dependent receptor that might be involved in activating this pathway, the present studies were performed to compare renin expression in wild types with that in mice with targeted deletions of ß(1) and ß(2)-adrenoceptors. The sympathetic nervous system is an important regulator of the renin system in the adult kidney so that activation of ß-adrenenoceptors may also participate in the activation of renin expression along the developing arterial tree and in upstream vasculature in adulthood. Compared with wild-types, renin expression was found to be significantly lower at all developmental stages in the kidneys of ß(1)/ß(2) Adr(-/-) mice. Three-dimensional analysis showed reduced renin expression in all segments of the vascular tree in mutants and a virtual absence of renin expression in the large arcuate arteries. Adult mutant kidneys showed the typical upstream renin expression after chronic stimulation. Tyrosine hydroxylase staining in fetal and postnatal kidneys revealed that sympathetic innervation of renin-producing cells occurs early in fetal development. Our data indicate that genetic disruption of ß-adrenergic receptors reduces basal renin expression along the developing preglomerular tree and in adult kidneys. Furthermore, ß-adrenergic receptor input is critical for the expression of renin in large renal vessels during early fetal development.


Assuntos
Vasos Sanguíneos/embriologia , Vasos Sanguíneos/metabolismo , Rim/embriologia , Rim/metabolismo , Receptores Adrenérgicos beta 1/fisiologia , Receptores Adrenérgicos beta 2/fisiologia , Renina/biossíntese , Animais , Arteríolas/metabolismo , Feminino , Desenvolvimento Fetal , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Gravidez , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 2/genética , Renina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sistema Nervoso Simpático/embriologia , Sistema Nervoso Simpático/fisiologia , Tirosina 3-Mono-Oxigenase/metabolismo
9.
Pflugers Arch ; 461(5): 567-77, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21424707

RESUMO

This study aimed to assess the role of cAMP target sequences enhancer cAMP response element (enhCRE) and cAMP and overlapping negative response element (CNRE) in the control of human renin gene (REN) in vivo. enhCRE and CNRE were silenced by mutations in a 12.2-kb human renin promoter fused to LacZ reporter gene. This construct was used to generate transgenic mice (RENMut-LacZ). The expression of the transgene was correctly targeted to the juxtaglomerular portions of renal afferent arterioles which express endogenous mouse renin. Therefore, enhCRE and CNRE do not seem to be relevant for the control of the cell-specific expression of the human renin gene. The ß-adrenoreceptor agonist isoproterenol (10 mg/kg/day, for 2 days) stimulated the endogenous renin, but not the LacZ mRNA expression. Treatment of RENMut-LacZ mice with the angiotensin converting enzyme inhibitor (enalapril 10 mg/kg/day, for 7 days) or their crossing to angiotensin receptor type 1a knockout mice led to increased renin and LacZ mRNA levels. Renin expression was upregulated by low-salt diet (0.03% NaCl, for 10 days) and downregulated by high-salt diet (4% NaCl, for 10 days). In contrast, low-salt diet did not influence, while high-salt diet inhibited the expression of LacZ. In summary, enhCRE and CNRE appear to be necessary for the transactivation of the human renin gene through ß-adrenoreceptors and by low-salt diet. Our data also suggest that different intracellular mechanisms mediate the effect of low- and high-salt intake on renin expression in vivo.


Assuntos
Dieta Hipossódica , Elementos Facilitadores Genéticos/fisiologia , Regulação da Expressão Gênica/genética , Renina/genética , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Animais , Linhagem Celular , AMP Cíclico/fisiologia , Humanos , Camundongos , Camundongos Transgênicos , Renina/biossíntese
10.
Am J Physiol Renal Physiol ; 296(5): F1006-12, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19261741

RESUMO

During metanephric kidney development, renin expression in the renal vasculature begins in larger vessels, shifting to smaller vessels and finally remaining restricted to the terminal portions of afferent arterioles at the entrance into the glomerular capillary network. The mechanisms determining the successive expression of renin along the vascular axis of the kidney are not well understood. Since the cAMP signaling cascade plays a central role in the regulation of both renin secretion and synthesis in the adult kidney, it seemed feasible that this pathway might also be critical for renin expression during kidney development. In the present study we determined the spatiotemporal development of renin expression and the development of the preglomerular arterial tree in mouse kidneys with renin cell-specific deletion of G(s)alpha, a core element for receptor activation of adenylyl cyclases. We found that in the absence of the G(s)alpha protein, renin expression was largely absent in the kidneys at any developmental stage, accompanied by alterations in the development of the preglomerular arterial tree. These data indicate that the maintenance of renin expression following a specific spatiotemporal pattern along the preglomerular vasculature critically depends on the availability of G(s)alpha. We infer from our data that the cAMP signaling pathway is not only critical for the regulation of renin synthesis and secretion in the mature kidney but that it also is critical for establishing the juxtaglomerular expression site of renin during development.


Assuntos
AMP Cíclico/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Glomérulos Renais , Circulação Renal/fisiologia , Renina/genética , Animais , Arteríolas/embriologia , Arteríolas/fisiologia , Cromograninas , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Sistema Justaglomerular/embriologia , Sistema Justaglomerular/fisiologia , Glomérulos Renais/irrigação sanguínea , Glomérulos Renais/embriologia , Glomérulos Renais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Renina/metabolismo , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...