Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Nephrol Dial Transplant ; 36(2): 237-246, 2021 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-33097957

RESUMEN

BACKGROUND: An underlying monogenic cause of early-onset chronic kidney disease (CKD) can be detected in ∼20% of individuals. For many etiologies of CKD manifesting before 25 years of age, >200 monogenic causative genes have been identified to date, leading to the elucidation of mechanisms of renal pathogenesis. METHODS: In 51 families with echogenic kidneys and CKD, we performed whole-exome sequencing to identify novel monogenic causes of CKD. RESULTS: We discovered a homozygous truncating mutation in the transcription factor gene transcription factor CP2-like 1 (TFCP2L1) in an Arabic patient of consanguineous descent. The patient developed CKD by the age of 2 months and had episodes of severe hypochloremic, hyponatremic and hypokalemic alkalosis, seizures, developmental delay and hypotonia together with cataracts. We found that TFCP2L1 was localized throughout kidney development particularly in the distal nephron. Interestingly, TFCP2L1 induced the growth and development of renal tubules from rat mesenchymal cells. Conversely, the deletion of TFCP2L1 in mice was previously shown to lead to reduced expression of renal cell markers including ion transporters and cell identity proteins expressed in different segments of the distal nephron. TFCP2L1 localized to the nucleus in HEK293T cells only upon coexpression with its paralog upstream-binding protein 1 (UBP1). A TFCP2L1 mutant complementary DNA (cDNA) clone that represented the patient's mutation failed to form homo- and heterodimers with UBP1, an essential step for its transcriptional activity. CONCLUSION: Here, we identified a loss-of-function TFCP2L1 mutation as a potential novel cause of CKD in childhood accompanied by a salt-losing tubulopathy.


Asunto(s)
Transición Epitelial-Mesenquimal , Enfermedades Renales/etiología , Mutación , Proteínas Represoras/genética , Animales , Niño , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Células HEK293 , Humanos , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Ratones , Ratones Noqueados , Ratas , Proteínas Represoras/metabolismo , Análisis de la Célula Individual , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Secuenciación del Exoma
2.
N Engl J Med ; 376(8): 742-754, 2017 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-28121514

RESUMEN

BACKGROUND: The DiGeorge syndrome, the most common of the microdeletion syndromes, affects multiple organs, including the heart, the nervous system, and the kidney. It is caused by deletions on chromosome 22q11.2; the genetic driver of the kidney defects is unknown. METHODS: We conducted a genomewide search for structural variants in two cohorts: 2080 patients with congenital kidney and urinary tract anomalies and 22,094 controls. We performed exome and targeted resequencing in samples obtained from 586 additional patients with congenital kidney anomalies. We also carried out functional studies using zebrafish and mice. RESULTS: We identified heterozygous deletions of 22q11.2 in 1.1% of the patients with congenital kidney anomalies and in 0.01% of population controls (odds ratio, 81.5; P=4.5×10-14). We localized the main drivers of renal disease in the DiGeorge syndrome to a 370-kb region containing nine genes. In zebrafish embryos, an induced loss of function in snap29, aifm3, and crkl resulted in renal defects; the loss of crkl alone was sufficient to induce defects. Five of 586 patients with congenital urinary anomalies had newly identified, heterozygous protein-altering variants, including a premature termination codon, in CRKL. The inactivation of Crkl in the mouse model induced developmental defects similar to those observed in patients with congenital urinary anomalies. CONCLUSIONS: We identified a recurrent 370-kb deletion at the 22q11.2 locus as a driver of kidney defects in the DiGeorge syndrome and in sporadic congenital kidney and urinary tract anomalies. Of the nine genes at this locus, SNAP29, AIFM3, and CRKL appear to be critical to the phenotype, with haploinsufficiency of CRKL emerging as the main genetic driver. (Funded by the National Institutes of Health and others.).


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Deleción Cromosómica , Síndrome de DiGeorge/genética , Haploinsuficiencia , Riñón/anomalías , Proteínas Nucleares/genética , Sistema Urinario/anomalías , Adolescente , Animales , Niño , Cromosomas Humanos Par 22 , Exoma , Femenino , Heterocigoto , Humanos , Lactante , Recién Nacido , Masculino , Ratones , Modelos Animales , Análisis de Secuencia de ADN , Adulto Joven , Pez Cebra
3.
Development ; 142(6): 1125-36, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25758223

RESUMEN

Healthy placental development is essential for reproductive success; failure of the feto-maternal interface results in pre-eclampsia and intrauterine growth retardation. We found that grainyhead-like 2 (GRHL2), a CP2-type transcription factor, is highly expressed in chorionic trophoblast cells, including basal chorionic trophoblast (BCT) cells located at the chorioallantoic interface in murine placentas. Placentas from Grhl2-deficient mouse embryos displayed defects in BCT cell polarity and basement membrane integrity at the chorioallantoic interface, as well as a severe disruption of labyrinth branching morphogenesis. Selective Grhl2 inactivation only in epiblast-derived cells rescued all placental defects but phenocopied intraembryonic defects observed in global Grhl2 deficiency, implying the importance of Grhl2 activity in trophectoderm-derived cells. ChIP-seq identified 5282 GRHL2 binding sites in placental tissue. By integrating these data with placental gene expression profiles, we identified direct and indirect Grhl2 targets and found a marked enrichment of GRHL2 binding adjacent to genes downregulated in Grhl2(-/-) placentas, which encoded known regulators of placental development and epithelial morphogenesis. These genes included that encoding the serine protease inhibitor Kunitz type 1 (Spint1), which regulates BCT cell integrity and labyrinth formation. In human placenta, we found that human orthologs of murine GRHL2 and its targets displayed co-regulation and were expressed in trophoblast cells in a similar domain as in mouse placenta. Our data indicate that a conserved Grhl2-coordinated gene network controls trophoblast branching morphogenesis, thereby facilitating development of the site of feto-maternal exchange. This might have implications for syndromes related to placental dysfunction.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Redes Reguladoras de Genes/fisiología , Morfogénesis/fisiología , Placentación , Factores de Transcripción/metabolismo , Trofoblastos/fisiología , Sitios de Unión/genética , Inmunoprecipitación de Cromatina , Femenino , Técnica del Anticuerpo Fluorescente , Redes Reguladoras de Genes/genética , Humanos , Inmunohistoquímica , Análisis por Micromatrices , Microscopía Electrónica , Embarazo , Proteínas Inhibidoras de Proteinasas Secretoras/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
4.
J Am Soc Nephrol ; 28(6): 1729-1740, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28028135

RESUMEN

Two metrics, a rise in serum creatinine concentration and a decrease in urine output, are considered tantamount to the injury of the kidney tubule and the epithelial cells thereof (AKI). Yet neither criterion emphasizes the etiology or the pathogenetic heterogeneity of acute decreases in kidney excretory function. In fact, whether decreased excretory function due to contraction of the extracellular fluid volume (vAKI) or due to intrinsic kidney injury (iAKI) actually share pathogenesis and should be aggregated in the same diagnostic group remains an open question. To examine this possibility, we created mouse models of iAKI and vAKI that induced a similar increase in serum creatinine concentration. Using laser microdissection to isolate specific domains of the kidney, followed by RNA sequencing, we found that thousands of genes responded specifically to iAKI or to vAKI, but very few responded to both stimuli. In fact, the activated gene sets comprised different, functionally unrelated signal transduction pathways and were expressed in different regions of the kidney. Moreover, we identified distinctive gene expression patterns in human urine as potential biomarkers of either iAKI or vAKI, but not both. Hence, iAKI and vAKI are biologically unrelated, suggesting that molecular analysis should clarify our current definitions of acute changes in kidney excretory function.


Asunto(s)
Lesión Renal Aguda/clasificación , Lesión Renal Aguda/genética , Transcriptoma , Animales , Femenino , Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL
5.
J Am Soc Nephrol ; 26(11): 2704-15, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25788534

RESUMEN

Grainyhead transcription factors control epithelial barriers, tissue morphogenesis, and differentiation, but their role in the kidney is poorly understood. Here, we report that nephric duct, ureteric bud, and collecting duct epithelia express high levels of grainyhead-like homolog 2 (Grhl2) and that nephric duct lumen expansion is defective in Grhl2-deficient mice. In collecting duct epithelial cells, Grhl2 inactivation impaired epithelial barrier formation and inhibited lumen expansion. Molecular analyses showed that GRHL2 acts as a transcriptional activator and strongly associates with histone H3 lysine 4 trimethylation. Integrating genome-wide GRHL2 binding as well as H3 lysine 4 trimethylation chromatin immunoprecipitation sequencing and gene expression data allowed us to derive a high-confidence GRHL2 target set. GRHL2 transactivated a group of genes including Ovol2, encoding the ovo-like 2 zinc finger transcription factor, as well as E-cadherin, claudin 4 (Cldn4), and the small GTPase Rab25. Ovol2 induction alone was sufficient to bypass the requirement of Grhl2 for E-cadherin, Cldn4, and Rab25 expression. Re-expression of either Ovol2 or a combination of Cldn4 and Rab25 was sufficient to rescue lumen expansion and barrier formation in Grhl2-deficient collecting duct cells. Hence, we identified a Grhl2/Ovol2 network controlling Cldn4 and Rab25 expression that facilitates lumen expansion and barrier formation in subtypes of renal epithelia.


Asunto(s)
Epitelio/metabolismo , Regulación de la Expresión Génica , Riñón/embriología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Sitios de Unión , Núcleo Celular/metabolismo , Inmunoprecipitación de Cromatina , Claudina-4/metabolismo , ADN/química , Técnicas de Transferencia de Gen , Histonas/química , Humanos , Inmunohistoquímica , Riñón/metabolismo , Túbulos Renales Colectores/metabolismo , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Unión Proteica , Proteínas/metabolismo , Transducción de Señal , Transcripción Genética
6.
Development ; 137(22): 3835-45, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20978075

RESUMEN

Differentiation of epithelial cells and morphogenesis of epithelial tubes or layers is closely linked with the establishment and remodeling of the apical junctional complex, which includes adherens junctions and tight junctions. Little is known about the transcriptional control of apical junctional complex components. Here, we show that the transcription factor grainyhead-like 2 (Grhl2), an epithelium-specific mammalian homolog of Drosophila Grainyhead, is essential for adequate expression of the adherens junction gene E-cadherin and the tight junction gene claudin 4 (Cldn4) in several types of epithelia, including gut endoderm, surface ectoderm and otic epithelium. We have generated Grhl2 mutant mice to demonstrate defective molecular composition of the apical junctional complex in these compartments that coincides with the occurrence of anterior and posterior neural tube defects. Mechanistically, we show that Grhl2 specifically associates with cis-regulatory elements localized at the Cldn4 core promoter and within intron 2 of the E-cadherin gene. Cldn4 promoter activity in epithelial cells is crucially dependent on the availability of Grhl2 and on the integrity of the Grhl2-associated cis-regulatory element. At the E-cadherin locus, the intronic Grhl2-associated cis-regulatory region contacts the promoter via chromatin looping, while loss of Grhl2 leads to a specific decrease of activating histone marks at the E-cadherin promoter. Together, our data provide evidence that Grhl2 acts as a target gene-associated transcriptional activator of apical junctional complex components and, thereby, crucially participates in epithelial differentiation.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Uniones Intercelulares/química , Factores de Transcripción/metabolismo , Animales , Cadherinas/metabolismo , Diferenciación Celular , Línea Celular , Claudina-4 , Perros , Células Epiteliales/citología , Células Epiteliales/metabolismo , Humanos , Uniones Intercelulares/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción/genética
8.
Nat Genet ; 51(1): 117-127, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30578417

RESUMEN

Congenital anomalies of the kidney and urinary tract (CAKUT) are a major cause of pediatric kidney failure. We performed a genome-wide analysis of copy number variants (CNVs) in 2,824 cases and 21,498 controls. Affected individuals carried a significant burden of rare exonic (that is, affecting coding regions) CNVs and were enriched for known genomic disorders (GD). Kidney anomaly (KA) cases were most enriched for exonic CNVs, encompassing GD-CNVs and novel deletions; obstructive uropathy (OU) had a lower CNV burden and an intermediate prevalence of GD-CNVs; and vesicoureteral reflux (VUR) had the fewest GD-CNVs but was enriched for novel exonic CNVs, particularly duplications. Six loci (1q21, 4p16.1-p16.3, 16p11.2, 16p13.11, 17q12 and 22q11.2) accounted for 65% of patients with GD-CNVs. Deletions at 17q12, 4p16.1-p16.3 and 22q11.2 were specific for KA; the 16p11.2 locus showed extensive pleiotropy. Using a multidisciplinary approach, we identified TBX6 as a driver for the CAKUT subphenotypes in the 16p11.2 microdeletion syndrome.


Asunto(s)
Variaciones en el Número de Copia de ADN/genética , Riñón/anomalías , Sistema Urinario/anomalías , Anomalías Urogenitales/genética , Reflujo Vesicoureteral/genética , Deleción Cromosómica , Femenino , Predisposición Genética a la Enfermedad/genética , Estudio de Asociación del Genoma Completo/métodos , Humanos , Masculino
9.
Science ; 360(6390): 758-763, 2018 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-29622724

RESUMEN

Our understanding of kidney disease pathogenesis is limited by an incomplete molecular characterization of the cell types responsible for the organ's multiple homeostatic functions. To help fill this knowledge gap, we characterized 57,979 cells from healthy mouse kidneys by using unbiased single-cell RNA sequencing. On the basis of gene expression patterns, we infer that inherited kidney diseases that arise from distinct genetic mutations but share the same phenotypic manifestation originate from the same differentiated cell type. We also found that the collecting duct in kidneys of adult mice generates a spectrum of cell types through a newly identified transitional cell. Computational cell trajectory analysis and in vivo lineage tracing revealed that intercalated cells and principal cells undergo transitions mediated by the Notch signaling pathway. In mouse and human kidney disease, these transitions were shifted toward a principal cell fate and were associated with metabolic acidosis.


Asunto(s)
Rastreo Celular/métodos , Perfilación de la Expresión Génica/métodos , Enfermedades Renales/genética , Túbulos Renales Colectores/citología , Túbulos Renales Colectores/metabolismo , Análisis de la Célula Individual/métodos , Animales , Plasticidad de la Célula , Marcadores Genéticos , Humanos , Ratones , Receptores Notch/metabolismo , Análisis de Secuencia de ARN/métodos , Transducción de Señal
10.
Elife ; 62017 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-28577314

RESUMEN

Although most nephron segments contain one type of epithelial cell, the collecting ducts consists of at least two: intercalated (IC) and principal (PC) cells, which regulate acid-base and salt-water homeostasis, respectively. In adult kidneys, these cells are organized in rosettes suggesting functional interactions. Genetic studies in mouse revealed that transcription factor Tfcp2l1 coordinates IC and PC development. Tfcp2l1 induces the expression of IC specific genes, including specific H+-ATPase subunits and Jag1. Jag1 in turn, initiates Notch signaling in PCs but inhibits Notch signaling in ICs. Tfcp2l1 inactivation deletes ICs, whereas Jag1 inactivation results in the forfeiture of discrete IC and PC identities. Thus, Tfcp2l1 is a critical regulator of IC-PC patterning, acting cell-autonomously in ICs, and non-cell-autonomously in PCs. As a result, Tfcp2l1 regulates the diversification of cell types which is the central characteristic of 'salt and pepper' epithelia and distinguishes the collecting duct from all other nephron segments.


Asunto(s)
Tipificación del Cuerpo , Regulación del Desarrollo de la Expresión Génica , Túbulos Renales Colectores/embriología , Proteínas Represoras/metabolismo , Transcripción Genética , Animales , Ratones
11.
J Clin Invest ; 124(7): 2963-76, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24937428

RESUMEN

α-Intercalated cells (A-ICs) within the collecting duct of the kidney are critical for acid-base homeostasis. Here, we have shown that A-ICs also serve as both sentinels and effectors in the defense against urinary infections. In a murine urinary tract infection model, A-ICs bound uropathogenic E. coli and responded by acidifying the urine and secreting the bacteriostatic protein lipocalin 2 (LCN2; also known as NGAL). A-IC-dependent LCN2 secretion required TLR4, as mice expressing an LPS-insensitive form of TLR4 expressed reduced levels of LCN2. The presence of LCN2 in urine was both necessary and sufficient to control the urinary tract infection through iron sequestration, even in the harsh condition of urine acidification. In mice lacking A-ICs, both urinary LCN2 and urinary acidification were reduced, and consequently bacterial clearance was limited. Together these results indicate that A-ICs, which are known to regulate acid-base metabolism, are also critical for urinary defense against pathogenic bacteria. They respond to both cystitis and pyelonephritis by delivering bacteriostatic chemical agents to the lower urinary system.


Asunto(s)
Proteínas de Fase Aguda/orina , Infecciones por Escherichia coli/prevención & control , Túbulos Renales Colectores/metabolismo , Lipocalinas/orina , Proteínas Oncogénicas/orina , Proteínas Proto-Oncogénicas/orina , Infecciones Urinarias/prevención & control , Escherichia coli Uropatógena , Equilibrio Ácido-Base , Proteínas de Fase Aguda/deficiencia , Proteínas de Fase Aguda/genética , Animales , Modelos Animales de Enfermedad , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/orina , Femenino , Humanos , Concentración de Iones de Hidrógeno , Hierro/metabolismo , Túbulos Renales Colectores/patología , Lipocalina 2 , Lipocalinas/genética , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Oncogénicas/deficiencia , Proteínas Oncogénicas/genética , Receptor Toll-Like 4/metabolismo , Infecciones Urinarias/microbiología , Infecciones Urinarias/orina
12.
Hepatology ; 44(4): 967-75, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17006929

RESUMEN

In mammalian liver, high glutamine synthetase (GS) expression is restricted to hepatocytes surrounding the terminal venules. The most important enhancer of the GS gene is located approximately 2520 base pairs (bp) upstream from the transcriptional start point. The nature of the transcription factors that bind to the enhancers has remained enigmatic. In this study, we purified nuclear proteins binding to the element. Supershift assays and footprint experiments with purified protein identified activated STAT5 as a transcription factor binding to a site within the enhancer. In addition, a second binding site close to the STAT5 site was observed that also binds a protein present in nuclear extracts. Sequence analysis indicated that the second site may bind a member of the LEF/TCF transcription factor family. Reporter gene assays demonstrate that the STAT5 binding site mediates enhancement of expression whereas the LEF/TCF site functions as a silencer of growth hormone-mediated enhancement in normal hepatocytes. LEF/TCF-sites are known to function as silencers in the absence and as enhancers in the presence of activated beta-catenin. In conclusion, the GS 5' enhancer contains elements important for GS expression in cells carrying an activated form of beta-catenin as previously shown in experimentally induced hepatocellular carcinomas.


Asunto(s)
Regulación de la Expresión Génica , Glutamato-Amoníaco Ligasa/biosíntesis , Hepatocitos/metabolismo , Factor de Transcripción STAT5/metabolismo , Factores de Transcripción TCF/metabolismo , Animales , Elementos de Facilitación Genéticos , Genes Reporteros , Glutamato-Amoníaco Ligasa/genética , Masculino , Proteínas Nucleares/metabolismo , Ratas , Ratas Sprague-Dawley , Análisis de Secuencia , Transfección
13.
Hepatology ; 41(6): 1225-32, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15880568

RESUMEN

The most striking phenomenon of glutamine synthetase (GS) expression in the liver is its unique restriction to cells surrounding the terminal hepatic venules. Expression is positively regulated by elements located in the 5'-upstream region and in the first intron of the gene. It was long believed that transcription factors present in GS-positive cells and absent in GS-negative cells are responsible for the phenomenon of zonal expression. However, strong enhancers are equally active in both types of cells. Therefore, the existence of a silencer mechanism in GS-negative hepatocytes was postulated. In the present study, a GS silencer element was investigated that was previously identified within the first intron and was shown to be able to prevent glucocorticoid-induced expression in cells negative for a transacting factor designated GS silencer element-binding protein. Reporter gene assays with the silencer element in combination with the most potent 5'-enhancer of the GS gene demonstrate that the silencer element is able to prevent enhancement mediated by the 5'-enhancer in combination with a heterologous as well as with the homologous promoter. More importantly, the effect of the silencer is shown to be restricted to GS-negative hepatocytes. In conclusion, the phenomenon of zonal expression of GS in the liver is caused by a protein present in GS-negative cells and absent in GS-positive cells that interacts with the silencer element in the first intron and not by a differential expression of enhancer-binding proteins.


Asunto(s)
Glutamato-Amoníaco Ligasa/metabolismo , Intrones , Hígado/enzimología , Elementos Silenciadores Transcripcionales , Animales , Elementos de Facilitación Genéticos , Expresión Génica , Genes Reporteros , Hepatocitos/metabolismo , Masculino , Regiones Promotoras Genéticas , Ratas , Ratas Sprague-Dawley , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA