Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 132(3): 422-33, 2008 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-18237772

RESUMEN

Cohesins mediate sister chromatid cohesion, which is essential for chromosome segregation and postreplicative DNA repair. In addition, cohesins appear to regulate gene expression and enhancer-promoter interactions. These noncanonical functions remained unexplained because knowledge of cohesin-binding sites and functional interactors in metazoans was lacking. We show that the distribution of cohesins on mammalian chromosome arms is not driven by transcriptional activity, in contrast to S. cerevisiae. Instead, mammalian cohesins occupy a subset of DNase I hypersensitive sites, many of which contain sequence motifs resembling the consensus for CTCF, a DNA-binding protein with enhancer blocking function and boundary-element activity. We find cohesins at most CTCF sites and show that CTCF is required for cohesin localization to these sites. Recruitment by CTCF suggests a rationale for noncanonical cohesin functions and, because CTCF binding is sensitive to DNA methylation, allows cohesin positioning to integrate DNA sequence and epigenetic state.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Cromosomas de los Mamíferos/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Animales , Secuencia de Bases , Factor de Unión a CCCTC , Diferenciación Celular , Línea Celular , Ensamble y Desensamble de Cromatina , Inmunoprecipitación de Cromatina , Citocinas/genética , Desoxirribonucleasa I/metabolismo , Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Linfocitos T/citología , Linfocitos T/metabolismo , Cohesinas
2.
Mol Biol Evol ; 37(7): 1986-2001, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32145025

RESUMEN

Genetic variation in the enzymes that catalyze posttranslational modification of proteins is a potentially important source of phenotypic variation during evolution. Ubiquitination is one such modification that affects turnover of virtually all of the proteins in the cell in addition to roles in signaling and epigenetic regulation. UBE2D3 is a promiscuous E2 enzyme, which acts as an ubiquitin donor for E3 ligases that catalyze ubiquitination of developmentally important proteins. We have used protein sequence comparison of UBE2D3 orthologs to identify a position in the C-terminal α-helical region of UBE2D3 that is occupied by a conserved serine in amniotes and by alanine in anamniote vertebrate and invertebrate lineages. Acquisition of the serine (S138) in the common ancestor to modern amniotes created a phosphorylation site for Aurora B. Phosphorylation of S138 disrupts the structure of UBE2D3 and reduces the level of the protein in mouse embryonic stem cells (ESCs). Substitution of S138 with the anamniote alanine (S138A) increases the level of UBE2D3 in ESCs as well as being a gain of function early embryonic lethal mutation in mice. When mutant S138A ESCs were differentiated into extraembryonic primitive endoderm, levels of the PDGFRα and FGFR1 receptor tyrosine kinases were reduced and primitive endoderm differentiation was compromised. Proximity ligation analysis showed increased interaction between UBE2D3 and the E3 ligase CBL and between CBL and the receptor tyrosine kinases. Our results identify a sequence change that altered the ubiquitination landscape at the base of the amniote lineage with potential effects on amniote biology and evolution.


Asunto(s)
Endodermo/enzimología , Evolución Molecular , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Enzimas Ubiquitina-Conjugadoras/genética , Vertebrados/genética , Sustitución de Aminoácidos , Animales , Aurora Quinasa B/metabolismo , Femenino , Humanos , Ratones , Fosforilación , Proteínas Tirosina Quinasas Receptoras/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Vertebrados/metabolismo
3.
J Immunol ; 202(8): 2502-2510, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30814307

RESUMEN

IFN-γ is a key cytokine of innate and adaptive immunity. It is important to understand temporal changes in IFN-γ production and how these changes relate to the role of IFN-γ in diverse models of infectious and autoimmune disease, making the ability to monitor and track IFN-γ production in vivo of a substantial benefit. IFN-γ ELISPOTs have been a central methodology to measure T cell immunity for many years. In this study, we add the capacity to analyze IFN-γ responses with high sensitivity and specificity, longitudinally, in vitro and in vivo. This allows the refinement of experimental protocols because immunity can be tracked in real-time through a longitudinal approach. We have generated a novel murine IFN-γ reporter transgenic model that allows IFN-γ production to be visualized and quantified in vitro and in vivo as bioluminescence using an imaging system. At baseline, in the absence of an inflammatory stimulus, IFN-γ signal from lymphoid tissue is detectable in vivo. Reporter transgenics are used in this study to track the IFN-γ response to Pseudomonas aeruginosa infection in the lung over time in vivo. The longitudinal development of the adaptive T cell immunity following immunization with Ag is identified from day 7 in vivo. Finally, we show that we are able to use this reporter transgenic to follow the onset of autoimmune T cell activation after regulatory T cell depletion in an established model of systemic autoimmunity. This IFN-γ reporter transgenic, termed "Gammaglow," offers a valuable new modality for tracking IFN-γ immunity, noninvasively and longitudinally over time.


Asunto(s)
Ensayo de Immunospot Ligado a Enzimas , Inmunidad Celular , Interferón gamma/inmunología , Mediciones Luminiscentes , Pulmón/inmunología , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Animales , Interferón gamma/genética , Pulmón/patología , Ratones , Ratones Transgénicos , Transgenes/inmunología
4.
Nature ; 524(7565): 356-60, 2015 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-26258299

RESUMEN

The typical response of the adult mammalian pulmonary circulation to a low oxygen environment is vasoconstriction and structural remodelling of pulmonary arterioles, leading to chronic elevation of pulmonary artery pressure (pulmonary hypertension) and right ventricular hypertrophy. Some mammals, however, exhibit genetic resistance to hypoxia-induced pulmonary hypertension. We used a congenic breeding program and comparative genomics to exploit this variation in the rat and identified the gene Slc39a12 as a major regulator of hypoxia-induced pulmonary vascular remodelling. Slc39a12 encodes the zinc transporter ZIP12. Here we report that ZIP12 expression is increased in many cell types, including endothelial, smooth muscle and interstitial cells, in the remodelled pulmonary arterioles of rats, cows and humans susceptible to hypoxia-induced pulmonary hypertension. We show that ZIP12 expression in pulmonary vascular smooth muscle cells is hypoxia dependent and that targeted inhibition of ZIP12 inhibits the rise in intracellular labile zinc in hypoxia-exposed pulmonary vascular smooth muscle cells and their proliferation in culture. We demonstrate that genetic disruption of ZIP12 expression attenuates the development of pulmonary hypertension in rats housed in a hypoxic atmosphere. This new and unexpected insight into the fundamental role of a zinc transporter in mammalian pulmonary vascular homeostasis suggests a new drug target for the pharmacological management of pulmonary hypertension.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Hipertensión Pulmonar/metabolismo , Hipoxia/metabolismo , Músculo Liso Vascular/metabolismo , Animales , Animales Congénicos , Arteriolas/metabolismo , Proteínas de Transporte de Catión/deficiencia , Proteínas de Transporte de Catión/genética , Bovinos , Hipoxia de la Célula , Proliferación Celular , Células Cultivadas , Cromosomas de los Mamíferos/genética , Enfermedad Crónica , Femenino , Técnicas de Silenciamiento del Gen , Homeostasis , Humanos , Hipertensión Pulmonar/genética , Hipoxia/genética , Espacio Intracelular/metabolismo , Masculino , Músculo Liso Vascular/citología , Ratas , Ratas Endogámicas F344 , Ratas Endogámicas WKY , Zinc/metabolismo
5.
J Immunol ; 194(10): 4705-4716, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25840911

RESUMEN

Epoxygenases belong to the cytochrome P450 family. They generate epoxyeicosatrienoic acids, which are known to have anti-inflammatory effects, but little is known about their role in macrophage function. By high-throughput sequencing of RNA in primary macrophages derived from rodents and humans, we establish the relative expression of epoxygenases in these cells. Zinc-finger nuclease-mediated targeted gene deletion of the major rat macrophage epoxygenase Cyp2j4 (ortholog of human CYP2J2) resulted in reduced epoxyeicosatrienoic acid synthesis. Cyp2j4(-/-) macrophages have relatively increased peroxisome proliferator-activated receptor-γ levels and show a profibrotic transcriptome, displaying overexpression of a specific subset of genes (260 transcripts) primarily involved in extracellular matrix, with fibronectin being the most abundantly expressed transcript. Fibronectin expression is under the control of epoxygenase activity in human and rat primary macrophages. In keeping with the in vitro findings, Cyp2j4(-/-) rats show upregulation of type I collagen following unilateral ureter obstruction of the kidney, and quantitative proteomics analysis (liquid chromatography-tandem mass spectrometry) showed increased renal type I collagen and fibronectin protein abundance resulting from experimentally induced crescentic glomerulonephritis in these rats. Taken together, these results identify the rat epoxygenase Cyp2j4 as a determinant of a profibrotic macrophage transcriptome that could have implications in various inflammatory conditions, depending on macrophage function.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Fibrosis/enzimología , Fibrosis/genética , Macrófagos/enzimología , Animales , Western Blotting , Cromatografía Liquida , Citocromo P-450 CYP2J2 , Familia 2 del Citocromo P450 , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Técnicas de Inactivación de Genes , Glomerulonefritis/enzimología , Glomerulonefritis/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Interferencia de ARN , Ratas , Ratas Endogámicas WKY , Reacción en Cadena en Tiempo Real de la Polimerasa , Espectrometría de Masas en Tándem , Transcriptoma
6.
Cell Physiol Biochem ; 39(5): 2077-2087, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27825168

RESUMEN

BACKGROUND: Serum & Glucocorticoid Regulated Kinase 1 (SGK1) plays a fundamental role in ion and solute transport processes in epithelia. In the endometrium, down-regulation of SGK1 during the window of receptivity facilitates embryo implantation whereas expression of a constitutively active mutant in the murine uterus blocks implantation. METHODS/RESULTS: Here, we report that treatment of endometrial epithelial cells with specific inhibitors of the phosphoinositide 3-kinase (PI3K)/AKT activity pathway results in reciprocal activation of SGK1. Flushing of the uterine lumen of mice with a cell permeable, substrate competitive phosphatidylinositol analogue that inhibits AKT activation (AKT inhibitor III) resulted in Sgk1 phosphorylation, down-regulation of the E3 ubiquitin-protein ligase Nedd4-2, and increased expression of epithelial Na+ channels (ENaC). Furthermore, exposure of the uterine lumen to AKT inhibitor III prior to embryo transfer induced a spectrum of early pregnancy defects, ranging from implantation failure to aberrant spacing of implantation sites. CONCLUSION: Taken together, our data indicate that the balanced activities of two related serine/threonine kinases, AKT and SGK1, critically govern the implantation process.


Asunto(s)
Implantación del Embrión/efectos de los fármacos , Proteínas Inmediatas-Precoces/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositoles/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Animales , Línea Celular , Endometrio/citología , Endometrio/efectos de los fármacos , Endometrio/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Canales Epiteliales de Sodio/genética , Canales Epiteliales de Sodio/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Proteínas Inmediatas-Precoces/agonistas , Proteínas Inmediatas-Precoces/metabolismo , Ratones , Ratones Endogámicos C57BL , Ubiquitina-Proteína Ligasas Nedd4 , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Embarazo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
7.
Cell Physiol Biochem ; 39(4): 1295-306, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27606670

RESUMEN

BACKGROUND: Serum & glucocorticoid inducible kinase (SGK1) regulates several ion channels, including amiloride sensitive epithelial Na+ channel (ENaC). SGK1 and ENaC in the luminal endometrium epithelium, are critically involved in embryo implantation, although little is known about their regulation. The present study explored whether SGK1 and ENaC are modulated by LEFTYA, a negative regulator of uterine receptivity. METHODS: Expression levels were determined by qRT-PCR and Western blotting, ENaC channel activity by whole cell patch clamp and transepithelial current by Ussing chamber experiments. RESULTS: Treatment of Ishikawa cells, an endometrial adenocarcinoma model cell line of endometrial epithelial cells, with LEFTYA rapidly up-regulated SGK1 and ENaC transcript and protein levels. Induction of ENaC in response to LEFTYA was blunted upon co-treatment with the SGK1 inhibitor EMD638683. ENaC levels also significantly upregulated upon expression of a constitutively active, but not a kinase dead, SGK1 mutant in Ishikawa cells. LEFTYA increased amiloride sensitive Na+-currents in Ishikawa cells and amiloride sensitive transepithelial current across the murine endometrium. Furthermore, LEFTYA induced the expression of ENaC in the endometrium of wild-type but not of Sgk1-deficient mice. CONCLUSIONS: LEFTYA regulates the expression and activity of ENaC in endometrial epithelial cells via SGK1. Aberrant regulation of SGK1 and ENaC by LEFTYA could contribute to the pathogenesis of unexplained infertility.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Canales Epiteliales de Sodio/genética , Proteínas Inmediatas-Precoces/genética , Factores de Determinación Derecha-Izquierda/farmacología , Proteínas Serina-Treonina Quinasas/genética , ARN Mensajero/genética , Amilorida/farmacología , Animales , Benzamidas/farmacología , Línea Celular Tumoral , Cámaras de Difusión de Cultivos , Endometrio/citología , Endometrio/efectos de los fármacos , Endometrio/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Canales Epiteliales de Sodio/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Hidrazinas/farmacología , Proteínas Inmediatas-Precoces/antagonistas & inhibidores , Proteínas Inmediatas-Precoces/deficiencia , Factores de Determinación Derecha-Izquierda/genética , Factores de Determinación Derecha-Izquierda/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Placa-Clamp , Cultivo Primario de Células , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/deficiencia , ARN Mensajero/metabolismo , Transducción de Señal
8.
J Am Soc Nephrol ; 25(1): 33-42, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24009238

RESUMEN

Vascular growth factors play an important role in maintaining the structure and integrity of the glomerular filtration barrier. In healthy adult glomeruli, the proendothelial survival factors vascular endothelial growth factor-A (VEGF-A) and angiopoietin-1 are constitutively expressed in glomerular podocyte epithelia. We demonstrate that this milieu of vascular growth factors is altered in streptozotocin-induced type 1 diabetic mice, with decreased angiopoietin-1 levels, VEGF-A upregulation, decreased soluble VEGF receptor-1 (VEGFR1), and increased VEGFR2 phosphorylation. This was accompanied by marked albuminuria, nephromegaly, hyperfiltration, glomerular ultrastructural alterations, and aberrant angiogenesis. We subsequently hypothesized that restoration of angiopoietin-1 expression within glomeruli might ameliorate manifestations of early diabetic glomerulopathy. Podocyte-specific inducible repletion of angiopoietin-1 in diabetic mice caused a 70% reduction of albuminuria and prevented diabetes-induced glomerular endothelial cell proliferation; hyperfiltration and renal morphology were unchanged. Furthermore, angiopoietin-1 repletion in diabetic mice increased Tie-2 phosphorylation, elevated soluble VEGFR1, and was paralleled by a decrease in VEGFR2 phosphorylation and increased endothelial nitric oxide synthase Ser(1177) phosphorylation. Diabetes-induced nephrin phosphorylation was also reduced in mice with angiopoietin-1 repletion. In conclusion, targeted angiopoietin-1 therapy shows promise as a renoprotective tool in the early stages of diabetic kidney disease.


Asunto(s)
Angiopoyetina 1/metabolismo , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/terapia , Terapia Molecular Dirigida , Angiopoyetina 1/deficiencia , Angiopoyetina 1/genética , Angiopoyetina 2/genética , Angiopoyetina 2/metabolismo , Animales , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/terapia , Nefropatías Diabéticas/patología , Humanos , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Mutantes , Podocitos/metabolismo , Podocitos/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
9.
Proc Natl Acad Sci U S A ; 108(48): 19270-5, 2011 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-22084104

RESUMEN

Timing of organ development during embryogenesis is coordinated such that at birth, organ and fetal size and maturity are appropriately proportioned. The extent to which local developmental timers are integrated with each other and with the signaling interactions that regulate morphogenesis to achieve this end is not understood. Using the absolute requirement for a signaling pathway activity (bone morphogenetic protein, BMP) during a critical stage of tooth development, we show that suboptimal levels of BMP signaling do not lead to abnormal morphogenesis, as suggested by mutants affecting BMP signaling, but to a 24-h stalling of the intrinsic developmental clock of the tooth. During this time, BMP levels accumulate to reach critical levels whereupon tooth development restarts, accelerates to catch up with development of the rest of the embryo and completes normal morphogenesis. This suggests that individual organs can autonomously control their developmental timing to adjust their stage of development to that of other organs. We also find that although BMP signaling is critical for the bud-to-cap transition in all teeth, levels of BMP signaling are regulated differently in multicusped teeth. We identify an interaction between two homeodomain transcription factors, Barx1 and Msx1, which is responsible for setting critical levels of BMP activity in multicusped teeth and provides evidence that correlates the levels of Barx1 transcriptional activity with cuspal complexity. This study highlights the importance of absolute levels of signaling activity for development and illustrates remarkable self-regulation in organogenesis that ensures coordination of developmental processes such that timing is subordinate to developmental structure.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Proteínas de Homeodominio/metabolismo , Factor de Transcripción MSX1/metabolismo , Odontogénesis/fisiología , Transducción de Señal/fisiología , Diente/embriología , Factores de Transcripción/metabolismo , Factores de Edad , Animales , Cartilla de ADN/genética , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Hibridación in Situ , Ratones , Ratones Noqueados , Microtomografía por Rayos X
10.
Commun Biol ; 7(1): 442, 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38600349

RESUMEN

Aryl hydrocarbon receptor (AHR) signalling integrates biological processes that sense and respond to environmental, dietary, and metabolic challenges to ensure tissue homeostasis. AHR is a transcription factor that is inactive in the cytosol but upon encounter with ligand translocates to the nucleus and drives the expression of AHR targets, including genes of the cytochrome P4501 family of enzymes such as Cyp1a1. To dynamically visualise AHR activity in vivo, we generated reporter mice in which firefly luciferase (Fluc) was non-disruptively targeted into the endogenous Cyp1a1 locus. Exposure of these animals to FICZ, 3-MC or to dietary I3C induced strong bioluminescence signal and Cyp1a1 expression in many organs including liver, lung and intestine. Longitudinal studies revealed that AHR activity was surprisingly long-lived in the lung, with sustained Cyp1a1 expression evident in discrete populations of cells including columnar epithelia around bronchioles. Our data link diet to lung physiology and also reveal the power of bespoke Cyp1a1-Fluc reporters to longitudinally monitor AHR activity in vivo.


Asunto(s)
Citocromo P-450 CYP1A1 , Receptores de Hidrocarburo de Aril , Ratones , Animales , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A1/metabolismo , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Luciferasas/genética , Hígado/metabolismo , Pulmón/metabolismo
11.
Proc Natl Acad Sci U S A ; 106(2): 516-21, 2009 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-19116266

RESUMEN

B cell development requires the coordinated rearrangement of Ig heavy (IgH) and light chain loci (IgL). Most mature B cells express a single B cell receptor of unique specificity, and a central question in immunology concerns the mechanisms that prevent the productive rearrangement of >1 IgH and IgL allele per cell. Probabilistic models of allelic exclusion maintain that simultaneous rearrangement of both alleles is rare, because the likelihood of undergoing rearrangement is low for a given Ig allele. Strong support for this idea came from studies in which a GFP marker was inserted into the Igk locus. In this system, the probability of high-level germ-line transcription and subsequent locus rearrangement appeared to be low in pre-B cells. Readdressing the validity of GFP expression as a reporter for the level of germ-line transcription, we found a striking discordance between GFP transcript and protein levels at the pre-B cell stage, which is explained at least in part by the developmentally regulated usage of 2 alternative Igk-J germ-line promoters. These results question the validity of the kappa-GFP system as evidence for probabilistic models of allelic exclusion.


Asunto(s)
Alelos , Linfocitos B/inmunología , Reordenamiento Génico , Modelos Estadísticos , Receptores de Antígenos de Linfocitos B/genética , Animales , Linfocitos B/citología , Células de la Médula Ósea , Células Cultivadas , Proteínas Fluorescentes Verdes , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Ligeras de Inmunoglobulina/genética , Inmunoglobulinas/genética , Ratones , Transcripción Genética
12.
Nat Commun ; 13(1): 2464, 2022 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-35513363

RESUMEN

Transmission of epigenetic information between generations occurs in nematodes, flies and plants, mediated by specialised small RNA pathways, modified histones and DNA methylation. Similar processes in mammals can also affect phenotype through intergenerational or trans-generational mechanisms. Here we generate a luciferase knock-in reporter mouse for the imprinted Dlk1 locus to visualise and track epigenetic fidelity across generations. Exposure to high-fat diet in pregnancy provokes sustained re-expression of the normally silent maternal Dlk1 in offspring (loss of imprinting) and increased DNA methylation at the somatic differentially methylated region (sDMR). In the next generation heterogeneous Dlk1 mis-expression is seen exclusively among animals born to F1-exposed females. Oocytes from these females show altered gene and microRNA expression without changes in DNA methylation, and correct imprinting is restored in subsequent generations. Our results illustrate how diet impacts the foetal epigenome, disturbing canonical and non-canonical imprinting mechanisms to modulate the properties of successive generations of offspring.


Asunto(s)
Epigénesis Genética , Impresión Genómica , Animales , Variación Biológica Poblacional , Metilación de ADN , Dieta Alta en Grasa , Femenino , Mamíferos , Ratones , Embarazo
13.
Brain Commun ; 3(1): fcaa130, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33758823

RESUMEN

Epilepsy is a serious neurological disorder affecting about 1% of the population worldwide. Epilepsy may arise as a result of acquired brain injury, or as a consequence of genetic predisposition. To date, genome-wide association studies and exome sequencing approaches have provided limited insights into the mechanisms of acquired brain injury. We have previously reported a pro-epileptic gene network, which is conserved across species, encoding inflammatory processes and positively regulated by sestrin3 (SESN3). In this study, we investigated the phenotype of SESN3 knock-out rats in terms of susceptibility to seizures and observed a significant delay in status epilepticus onset in SESN3 knock-out compared to control rats. This finding confirms previous in vitro and in vivo evidence indicating that SESN3 may favour occurrence and/or severity of seizures. We also analysed the phenotype of SESN3 knock-out rats for common comorbidities of epilepsy, i.e., anxiety, depression and cognitive impairment. SESN3 knock-out rats proved less anxious compared to control rats in a selection of behavioural tests. Taken together, the present results suggest that SESN3 may regulate mechanisms involved in the pathogenesis of epilepsy and its comorbidities.

14.
Dev Biol ; 331(1): 38-49, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19394325

RESUMEN

Nevoid basal cell carcinoma syndrome (NBCCS) is an autosomal dominant or spontaneous disorder characterized by multiple cutaneous basal cell carcinomas, odontogenic keratocysts, skeletal anomalies and facial dysmorphology, including cleft lip and palate. Causative mutations for NBCCS occur in the PTCH1 gene on chromosome 9q22.3-q31, which encodes the principle receptor for the Hedgehog signalling pathway. We have investigated the molecular basis of craniofacial defects seen in NBCCS using a transgenic mouse model expressing Shh in basal epithelium under a Keratin-14 promoter. These mice have an absence of flat bones within the skull vault, hypertelorism, open-bite malocclusion, cleft palate and arrested tooth development. Significantly, increased Hedgehog signal transduction in these mice can influence cell fate within the craniofacial region. In medial edge epithelium of the palate, Shh activity prevents apoptosis and subsequent palatal shelf fusion. In contrast, high levels of Shh in odontogenic epithelium arrests tooth development at the bud stage, secondary to a lack of cell proliferation in this region. These findings illustrate the importance of appropriately regulated Hedgehog signalling during early craniofacial development and demonstrate that oro-facial clefting and hypodontia seen in NBCCS can occur as a direct consequence of increased Shh signal activity within embryonic epithelial tissues.


Asunto(s)
Síndrome del Nevo Basocelular/genética , Proteínas Hedgehog/genética , Diente/crecimiento & desarrollo , Anomalías Múltiples/genética , Animales , Síndrome del Nevo Basocelular/patología , Muerte Celular , División Celular , Mapeo Cromosómico , Cromosomas Humanos Par 9 , Fisura del Paladar/genética , Cartilla de ADN , Modelos Animales de Enfermedad , Humanos , Hibridación in Situ , Queratina-14/genética , Meduloblastoma/patología , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Diente/embriología , Diente/patología
15.
Dev Cell ; 4(4): 481-95, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12689588

RESUMEN

Previous studies have implicated the Eed-Enx1 Polycomb group complex in the maintenance of imprinted X inactivation in the trophectoderm lineage in mouse. Here we show that recruitment of Eed-Enx1 to the inactive X chromosome (Xi) also occurs in random X inactivation in the embryo proper. Localization of Eed-Enx1 complexes to Xi occurs very early, at the onset of Xist expression, but then disappears as differentiation and development progress. This transient localization correlates with the presence of high levels of the complex in totipotent cells and during early differentiation stages. Functional analysis demonstrates that Eed-Enx1 is required to establish methylation of histone H3 at lysine 9 and/or lysine 27 on Xi and that this, in turn, is required to stabilize the Xi chromatin structure.


Asunto(s)
Compensación de Dosificación (Genética) , Embrión de Mamíferos/embriología , N-Metiltransferasa de Histona-Lisina , Metiltransferasas/metabolismo , Proteínas Represoras/metabolismo , Células Madre Totipotentes/metabolismo , Cromosoma X/genética , Secuencia de Aminoácidos/genética , Animales , Diferenciación Celular/genética , Células Cultivadas , Cromatina/genética , Cromatina/metabolismo , Metilación de ADN , Femenino , Feto , Regulación del Desarrollo de la Expresión Génica/genética , Histona Metiltransferasas , Histonas/genética , Histonas/metabolismo , Lisina/genética , Lisina/metabolismo , Masculino , Metiltransferasas/genética , Ratones , Ratones Endogámicos C57BL , Complejo Represivo Polycomb 2 , Proteínas del Grupo Polycomb , Proteína Metiltransferasas , ARN Largo no Codificante , ARN no Traducido/genética , ARN no Traducido/metabolismo , Proteínas Represoras/genética , Células Madre Totipotentes/citología
16.
Hypertension ; 74(3): 687-696, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31327268

RESUMEN

Metabolic syndrome is a cause of coronary artery disease and type 2 diabetes mellitus. Camk2n1 resides in genomic loci for blood pressure, left ventricle mass, and type 2 diabetes mellitus, and in the spontaneously hypertensive rat model of metabolic syndrome, Camk2n1 expression is cis-regulated in left ventricle and fat and positively correlates with adiposity. Therefore, we knocked out Camk2n1 in spontaneously hypertensive rat to investigate its role in metabolic syndrome. Compared with spontaneously hypertensive rat, Camk2n1-/- rats had reduced cardiorenal CaMKII (Ca2+/calmodulin-dependent kinase II) activity, lower blood pressure, enhanced nitric oxide bioavailability, and reduced left ventricle mass associated with altered hypertrophic networks. Camk2n1 deficiency reduced insulin resistance, visceral fat, and adipogenic capacity through the altered cell cycle and complement pathways, independent of CaMKII. In human visceral fat, CAMK2N1 expression correlated with adiposity and genomic variants that increase CAMK2N1 expression associated with increased risk of coronary artery disease and type 2 diabetes mellitus. Camk2n1 regulates multiple networks that control metabolic syndrome traits and merits further investigation as a therapeutic target in humans.


Asunto(s)
Proteínas Portadoras/genética , Hipertensión/genética , Hipertrofia Ventricular Izquierda/genética , Síndrome Metabólico/fisiopatología , Adiposidad/genética , Animales , Proteínas de Unión al Calcio , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/fisiopatología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Hipertensión/fisiopatología , Hipertrofia Ventricular Izquierda/fisiopatología , Síndrome Metabólico/genética , Distribución Aleatoria , Ratas , Ratas Endogámicas SHR , Medición de Riesgo , Sensibilidad y Especificidad
17.
J Mol Med (Berl) ; 96(2): 173-182, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29230527

RESUMEN

Early embryo development and endometrial differentiation are initially independent processes, and synchronization, imposed by a limited window of implantation, is critical for reproductive success. A putative negative regulator of endometrial receptivity is LEFTY2, a member of the transforming growth factor (TGF)-ß family. LEFTY2 is highly expressed in decidualizing human endometrial stromal cells (HESCs) during the late luteal phase of the menstrual cycle, coinciding with the closure of the window of implantation. Here, we show that flushing of the uterine lumen in mice with recombinant LEFTY2 inhibits the expression of key receptivity genes, including Cox2, Bmp2, and Wnt4, and blocks embryo implantation. In Ishikawa cells, a human endometrial epithelial cell line, LEFTY2 downregulated the expression of calcium release-activated calcium channel protein 1, encoded by ORAI1, and inhibited store-operated Ca2+ entry (SOCE). Furthermore, LEFTY2 and the Orai1 blockers 2-APB, MRS-1845, as well as YM-58483, inhibited, whereas the Ca2+ ionophore, ionomycin, strongly upregulated COX2, BMP2 and WNT4 expression in decidualizing HESCs. These findings suggest that LEFTY2 closes the implantation window, at least in part, by downregulating Orai1, which in turn limits SOCE and antagonizes expression of Ca2+-sensitive receptivity genes. KEY MESSAGES: •Endometrial receptivity is negatively regulated by LEFTY2. •LEFTY2 inhibits the expression of key murine receptivity genes, including Cox2, Bmp2 and Wnt4, and blocks embryo implantation. •LEFTY2 downregulates the expression of Orai1 and inhibits SOCE. •LEFTY2 and the Orai1 blockers 2-APB, MRS-1845, and YM-58483 inhibit COX2, BMP2, and WNT4 expression in endometrial cells. •Targeting LEFTY2 and Orai1 may represent a novel approach for treating unexplained infertility.


Asunto(s)
Calcio/fisiología , Endometrio/fisiología , Factores de Determinación Derecha-Izquierda/fisiología , Proteína ORAI1/fisiología , Animales , Células Cultivadas , Regulación hacia Abajo , Endometrio/citología , Femenino , Humanos , Ratones Endogámicos C57BL
18.
Hypertension ; 2017 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-28739975

RESUMEN

CFB (complement factor B) is elevated in adipose tissue and serum from patients with type 2 diabetes mellitus and cardiovascular disease, but the causal relationship to disease pathogenesis is unclear. Cfb is also elevated in adipose tissue and serum of the spontaneously hypertensive rat, a well-characterized model of metabolic syndrome. To establish the role of CFB in metabolic syndrome, we knocked out the Cfb gene in the spontaneously hypertensive rat. Cfb-/- rats showed improved glucose tolerance and insulin sensitivity, redistribution of visceral to subcutaneous fat, increased adipocyte mitochondrial respiration, and marked changes in gene expression. Cfb-/- rats also had lower blood pressure, increased ejection fraction and fractional shortening, and reduced left ventricular mass. These changes in metabolism and gene expression, in adipose tissue and left ventricle, suggest new adipose tissue-intrinsic and blood pressure-independent mechanisms for insulin resistance and cardiac hypertrophy in the spontaneously hypertensive rat. In silico analysis of the human CFB locus revealed 2 cis-regulated expression quantitative trait loci for CFB expression significantly associated with visceral fat, circulating triglycerides and hypertension in genome-wide association studies. Together, these data demonstrate a key role for CFB in the development of spontaneously hypertensive rat metabolic syndrome phenotypes and of related traits in humans and indicate the potential for CFB as a novel target for treatment of cardiometabolic disease.

19.
Cell Rep ; 18(5): 1090-1099, 2017 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-28147266

RESUMEN

Imprinted genes are regulated according to parental origin and can influence embryonic growth and metabolism and confer disease susceptibility. Here, we designed sensitive allele-specific reporters to non-invasively monitor imprinted Cdkn1c expression in mice and showed that expression was modulated by environmental factors encountered in utero. Acute exposure to chromatin-modifying drugs resulted in de-repression of paternally inherited (silent) Cdkn1c alleles in embryos that was temporary and resolved after birth. In contrast, deprivation of maternal dietary protein in utero provoked permanent de-repression of imprinted Cdkn1c expression that was sustained into adulthood and occurred through a folate-dependent mechanism of DNA methylation loss. Given the function of imprinted genes in regulating behavior and metabolic processes in adults, these results establish imprinting deregulation as a credible mechanism linking early-life adversity to later-life outcomes. Furthermore, Cdkn1c-luciferase mice offer non-invasive tools to identify factors that disrupt epigenetic processes and strategies to limit their long-term impact.


Asunto(s)
Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Impresión Genómica/fisiología , Alelos , Animales , Cromatina/fisiología , Metilación de ADN/fisiología , Epigénesis Genética/fisiología , Ratones
20.
FASEB J ; 19(12): 1680-2, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16099943

RESUMEN

Agouti-related protein (AgRP) and neuropeptide Y (NPY) are colocalized in arcuate nucleus (arcuate) neurons implicated in the regulation of energy balance. Both AgRP and NPY stimulate food intake when administered into the third ventricle and are up-regulated in states of negative energy balance. However, mice with targeted deletion of either NPY or AgRP or both do not have major alterations in energy homeostasis. Using bacterial artificial chromosome (BAC) transgenesis we have targeted expression of a neurotoxic CAG expanded form of ataxin-3 to AgRP-expressing neurons in the arcuate. This resulted in a 47% loss of AgRP neurons by 16 weeks of age, a significantly reduced body weight, (wild-type mice (WT) 34.7+/-0.7 g vs. transgenic mice (Tg) 28.6+/-0.6 g, P<0.001), and reduced food intake (WT 5.0+/-0.2 vs. Tg 3.6+/-0.1 g per day, P<0.001). Transgenic mice had significantly reduced total body fat, plasma insulin, and increased brown adipose tissue UCP1 expression. Transgenic mice failed to respond to peripherally administered ghrelin but retained sensitivity to PYY 3-36. These data suggest that postembryonic partial loss of AgRP/NPY neurons leads to a lean, hypophagic phenotype.


Asunto(s)
Neuronas/metabolismo , Neuropéptido Y/genética , Proteínas/genética , Tejido Adiposo/metabolismo , Proteína Relacionada con Agouti , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Glucemia/metabolismo , Northern Blotting , Composición Corporal , Peso Corporal , Proteínas Portadoras/metabolismo , Cromosomas Artificiales Bacterianos/metabolismo , ADN Complementario/metabolismo , Exones , Conducta Alimentaria , Eliminación de Gen , Ghrelina , Hipotálamo/metabolismo , Hipotálamo/patología , Hibridación in Situ , Insulina/sangre , Péptidos y Proteínas de Señalización Intercelular , Canales Iónicos , Leptina/sangre , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Proteínas Mitocondriales , Modelos Genéticos , Neuropéptido Y/metabolismo , Hormonas Peptídicas/metabolismo , Fenotipo , Reacción en Cadena de la Polimerasa , Estructura Terciaria de Proteína , Proteínas/metabolismo , ARN Mensajero/metabolismo , Radioinmunoensayo , Delgadez/genética , Factores de Tiempo , Transgenes , Proteína Desacopladora 1 , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA