Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Psychiatry ; 28(7): 2946-2963, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37131076

RESUMEN

While persistence of fear memories is essential for survival, a failure to inhibit fear in response to harmless stimuli is a feature of anxiety disorders. Extinction training only temporarily suppresses fear memory recovery in adults, but it is highly effective in juvenile rodents. Maturation of GABAergic circuits, in particular of parvalbumin-positive (PV+) cells, restricts plasticity in the adult brain, thus reducing PV+ cell maturation could promote the suppression of fear memories following extinction training in adults. Epigenetic modifications such as histone acetylation control gene accessibility for transcription and help couple synaptic activity to changes in gene expression. Histone deacetylase 2 (Hdac2), in particular, restrains both structural and functional synaptic plasticity. However, whether and how Hdac2 controls the maturation of postnatal PV+ cells is not well understood. Here, we show that PV+- cell specific Hdac2 deletion limits spontaneous fear memory recovery in adult mice, while enhancing PV+ cell bouton remodeling and reducing perineuronal net aggregation around PV+ cells in prefrontal cortex and basolateral amygdala. Prefrontal cortex PV+ cells lacking Hdac2, show reduced expression of Acan, a critical perineuronal net component, which is rescued by Hdac2 re-expression. Pharmacological inhibition of Hdac2 before extinction training is sufficient to reduce both spontaneous fear memory recovery and Acan expression in wild-type adult mice, while these effects are occluded in PV+-cell specific Hdac2 conditional knockout mice. Finally, a brief knock-down of Acan expression mediated by intravenous siRNA delivery before extinction training but after fear memory acquisition is sufficient to reduce spontaneous fear recovery in wild-type mice. Altogether, these data suggest that controlled manipulation of PV+ cells by targeting Hdac2 activity, or the expression of its downstream effector Acan, promotes the long-term efficacy of extinction training in adults.


Asunto(s)
Condicionamiento Psicológico , Parvalbúminas , Ratones , Animales , Parvalbúminas/metabolismo , Regulación hacia Abajo , Condicionamiento Psicológico/fisiología , Memoria/fisiología , Miedo/fisiología , Ratones Noqueados , Extinción Psicológica/fisiología
2.
PLoS Genet ; 12(10): e1006335, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27760138

RESUMEN

Left-ventricular outflow tract obstructions (LVOTO) encompass a wide spectrum of phenotypically heterogeneous heart malformations which frequently cluster in families. We performed family based whole-exome and targeted re-sequencing on 182 individuals from 51 families with multiple affected members. Central to our approach is the family unit which serves as a reference to identify causal genotype-phenotype correlations. Screening a multitude of 10 overlapping phenotypes revealed disease associated and co-segregating variants in 12 families. These rare or novel protein altering mutations cluster predominantly in genes (NOTCH1, ARHGAP31, MAML1, SMARCA4, JARID2, JAG1) along the Notch signaling cascade. This is in line with a significant enrichment (Wilcoxon, p< 0.05) of variants with a higher pathogenicity in the Notch signaling pathway in patients compared to controls. The significant enrichment of novel protein truncating and missense mutations in NOTCH1 highlights the allelic and phenotypic heterogeneity in our pediatric cohort. We identified novel co-segregating pathogenic mutations in NOTCH1 associated with left and right-sided cardiac malformations in three independent families with a total of 15 affected individuals. In summary, our results suggest that a small but highly pathogenic fraction of family specific mutations along the Notch cascade are a common cause of LVOTO.


Asunto(s)
Constricción Patológica/genética , Cardiopatías Congénitas/genética , Receptor Notch1/genética , Obstrucción del Flujo Ventricular Externo/genética , Válvula Aórtica/fisiopatología , Codón sin Sentido , Constricción Patológica/fisiopatología , Exoma/genética , Femenino , Estudios de Asociación Genética , Ligamiento Genético , Genoma Humano , Cardiopatías Congénitas/fisiopatología , Humanos , Masculino , Linaje , Receptores Notch/genética , Eliminación de Secuencia , Transducción de Señal/genética , Obstrucción del Flujo Ventricular Externo/fisiopatología
3.
J Hypertens ; 42(7): 1256-1268, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38704218

RESUMEN

OBJECTIVES: γδ T-lymphocytes play a role in angiotensin II (AngII)-induced hypertension, vascular injury and T-cell infiltration in perivascular adipose tissue (PVAT) in mice. Mesenteric arteries of hypertensive mice and subcutaneous arteries from obese humans present similar remodeling. We hypothesized that γδ T-cell subtypes in mesenteric vessels with PVAT (MV/PVAT) from hypertensive mice and subcutaneous adipose tissue (SAT) from obese humans, who are prone to develop hypertension, would be similar. METHODS: Mice were infused with AngII for 14 days. MV/PVAT T-cells were used for single-cell RNA-sequencing (scRNA-seq). scRNA-seq data (GSE155960) of SAT CD45 + cells from three lean and three obese women were downloaded from the Gene Expression Omnibus database. RESULTS: δ T-cell subclustering identified six δ T-cell subtypes. AngII increased T-cell receptor δ variable 4 ( Trdv4 ) + γδ T-effector memory cells and Cd28high δ T EM -cells, changes confirmed by flow cytometry. δ T-cell subclustering identified nine δ T-cell subtypes in human SAT. CD28 expressing δ T-cell subclustering demonstrated similar δ T-cell subpopulations in murine MV/PVAT and human SAT. Cd28+ γδ NKT EM and Cd28high δ T EM -cells increased in MV/PVAT from hypertensive mice and CD28high δ T EM -cells in SAT from obese women compared to the lean women. CONCLUSION: Similar CD28 + δ T-cells were identified in murine MV/PVAT and human SAT. CD28 high δ T EM -cells increased in MV/PVAT in hypertensive mice and in SAT from humans with obesity, a prehypertensive condition. CD28 + δ T-lymphocytes could have a pathogenic role in human hypertension associated with obesity, and could be a potential target for therapy.


Asunto(s)
Antígenos CD28 , Hipertensión , Obesidad , Grasa Subcutánea , Animales , Humanos , Hipertensión/inmunología , Hipertensión/metabolismo , Ratones , Grasa Subcutánea/metabolismo , Antígenos CD28/metabolismo , Femenino , Masculino , Angiotensina II , Linfocitos Intraepiteliales/inmunología , Linfocitos Intraepiteliales/metabolismo , Tejido Adiposo/metabolismo
4.
J Immunother Cancer ; 12(5)2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38754915

RESUMEN

BACKGROUND: Allogeneic hematopoietic stem cell transplantation (HSCT) remains the standard of care for chemotherapy-refractory leukemia patients, but cure rates are still dismal. To prevent leukemia relapse following HSCT, we aim to improve the early graft-versus-leukemia effect mediated by natural killer (NK) cells. Our approach is based on the adoptive transfer of Therapeutic Inducers of Natural Killer cell Killing (ThINKK). ThINKK are expanded and differentiated from HSC, and exhibit blood plasmacytoid dendritic cell (pDC) features. We previously demonstrated that ThINKK stimulate NK cells and control acute lymphoblastic leukemia (ALL) development in a preclinical mouse model of HSCT for ALL. Here, we assessed the cellular identity of ThINKK and investigated their potential to activate allogeneic T cells. We finally evaluated the effect of immunosuppressive drugs on ThINKK-NK cell interaction. METHODS: ThINKK cellular identity was explored using single-cell RNA sequencing and flow cytometry. Their T-cell activating potential was investigated by coculture of allogeneic T cells and antigen-presenting cells in the presence or the absence of ThINKK. A preclinical human-to-mouse xenograft model was used to evaluate the impact of ThINKK injections on graft-versus-host disease (GvHD). Finally, the effect of immunosuppressive drugs on ThINKK-induced NK cell cytotoxicity against ALL cells was tested. RESULTS: The large majority of ThINKK shared the key characteristics of canonical blood pDC, including potent type-I interferon (IFN) production following Toll-like receptor stimulation. A minor subset expressed some, although not all, markers of other dendritic cell populations. Importantly, while ThINKK were not killed by allogeneic T or NK cells, they did not increase T cell proliferation induced by antigen-presenting cells nor worsened GvHD in vivo. Finally, tacrolimus, sirolimus or mycophenolate did not decrease ThINKK-induced NK cell activation and cytotoxicity. CONCLUSION: Our results indicate that ThINKK are type I IFN producing cells with low T cell activation capacity. Therefore, ThINKK adoptive immunotherapy is not expected to increase the risk of GvHD after allogeneic HSCT. Furthermore, our data predict that the use of tacrolimus, sirolimus or mycophenolate as anti-GvHD prophylaxis regimen will not decrease ThINKK therapeutic efficacy. Collectively, these preclinical data support the testing of ThINKK immunotherapy in a phase I clinical trial.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Asesinas Naturales , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/efectos de los fármacos , Humanos , Trasplante de Células Madre Hematopoyéticas/métodos , Animales , Ratones , Trasplante Homólogo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Enfermedad Injerto contra Huésped/prevención & control
5.
Cell Rep ; 43(1): 113616, 2024 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-38150367

RESUMEN

Macrophages populate the embryo early in gestation, but their role in development is not well defined. In particular, specification and function of macrophages in intestinal development remain little explored. To study this event in the human developmental context, we derived and combined human intestinal organoid and macrophages from pluripotent stem cells. Macrophages migrate into the organoid, proliferate, and occupy the emerging microanatomical niches of epithelial crypts and ganglia. They also acquire a transcriptomic profile similar to that of fetal intestinal macrophages and display tissue macrophage behaviors, such as recruitment to tissue injury. Using this model, we show that macrophages reduce glycolysis in mesenchymal cells and limit tissue growth without affecting tissue architecture, in contrast to the pro-growth effect of enteric neurons. In short, we engineered an intestinal tissue model populated with macrophages, and we suggest that resident macrophages contribute to the regulation of metabolism and growth of the developing intestine.


Asunto(s)
Macrófagos , Células Madre Pluripotentes , Humanos , Diferenciación Celular , Macrófagos/metabolismo , Intestinos , Células Madre Pluripotentes/metabolismo , Intestino Delgado , Organoides/metabolismo
6.
Nat Commun ; 14(1): 4033, 2023 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-37468473

RESUMEN

Muscle stem cells, the engine of muscle repair, are affected in myotonic dystrophy type 1 (DM1); however, the underlying molecular mechanism and the impact on the disease severity are still elusive. Here, we show using patients' samples that muscle stem cells/myoblasts exhibit signs of cellular senescence in vitro and in situ. Single cell RNAseq uncovers a subset of senescent myoblasts expressing high levels of genes related to the senescence-associated secretory phenotype (SASP). We show that the levels of interleukin-6, a prominent SASP cytokine, in the serum of DM1 patients correlate with muscle weakness and functional capacity limitations. Drug screening revealed that the senolytic BCL-XL inhibitor (A1155463) can specifically remove senescent DM1 myoblasts by inducing their apoptosis. Clearance of senescent cells reduced the expression of SASP, which rescued the proliferation and differentiation capacity of DM1 myoblasts in vitro and enhanced their engraftment following transplantation in vivo. Altogether, this study identifies the pathogenic mechanism associated with muscle stem cell defects in DM1 and opens a therapeutic avenue that targets these defective cells to restore myogenesis.


Asunto(s)
Distrofia Miotónica , Células Satélite del Músculo Esquelético , Humanos , Distrofia Miotónica/tratamiento farmacológico , Distrofia Miotónica/genética , Distrofia Miotónica/metabolismo , Senoterapéuticos , Fibras Musculares Esqueléticas/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Desarrollo de Músculos/genética
7.
Proc Natl Acad Sci U S A ; 105(9): 3449-54, 2008 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-18296642

RESUMEN

Neural tube defects (NTDs) are very frequent congenital abnormalities in humans. Recently, we have documented independent association of Vangl1 and Vangl2 gene mutations with NTDs. In the Looptail mouse, homozygosity (but not heterozygosity) for loss-of-function alleles at Vangl2 causes the severe NTD craniorachischisis, whereas heterozygosity for mutant variants of VANGL1 is associated with NTDs in a human cohort of sporadic and familial cases. To understand the role of Vangl1 in normal development, we created a mouse mutant with an inactivating mutation at Vangl1 (Vangl1(gt)). Vangl1 shows a dynamic pattern of expression in the developing neural tube and notochord at the time of neural tube closure. Vangl1(gt/+) heterozygotes and Vangl1(gt/gt) homozygotes are viable and fertile, although Vangl1(gt/gt) display subtle alterations in polarity of inner hair cells of the cochlea. Remarkably, and as opposed to healthy Vangl1(gt/+) and Vangl2(lp/+) heterozygotes, Vangl1(gt/+);Vangl2(lp/+) double heterozygotes show profound developmental defects that include severe craniorachischisis, inner ear defects (disorganization of the stereociliary bundles of hair cells of the organ of Corti), and cardiac abnormality (aberrant right subclavian artery). These results show that genetic interaction between Vangl1 and Vangl2 genes causes neural tube defects and raise the possibility that interaction between individual Vangl genes and other genetic loci and/or environmental factors may additionally contribute to the etiology of NTDs.


Asunto(s)
Proteínas Portadoras/genética , Heterocigoto , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Defectos del Tubo Neural/genética , Animales , Cardiopatías , Enfermedades del Laberinto , Ratones , Ratones Mutantes , Ratones Mutantes Neurológicos , Defectos del Tubo Neural/etiología , Notocorda , Fenotipo
8.
Dev Cell ; 56(15): 2237-2251.e6, 2021 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-34273276

RESUMEN

Endothelial tip cells guiding tissue vascularization are primary targets for angiogenic therapies. Whether tip cells require differential signals to develop their complex branching patterns remained unknown. Here, we show that diving tip cells invading the mouse neuroretina (D-tip cells) are distinct from tip cells guiding the superficial retinal vascular plexus (S-tip cells). D-tip cells have a unique transcriptional signature, including high TGF-ß signaling, and they begin to acquire blood-retina barrier properties. Endothelial deletion of TGF-ß receptor I (Alk5) inhibits D-tip cell identity acquisition and deep vascular plexus formation. Loss of endothelial ALK5, but not of the canonical SMAD effectors, leads to aberrant contractile pericyte differentiation and hemorrhagic vascular malformations. Oxygen-induced retinopathy vasculature exhibits S-like tip cells, and Alk5 deletion impedes retina revascularization. Our data reveal stage-specific tip cell heterogeneity as a requirement for retinal vascular development and suggest that non-canonical-TGF-ß signaling could improve retinal revascularization and neural function in ischemic retinopathy.


Asunto(s)
Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Retina/fisiología , Neovascularización Retiniana/metabolismo , Animales , Células Endoteliales/metabolismo , Endotelio Vascular , Ratones , Ratones Noqueados , Neovascularización Fisiológica/fisiología , Receptor Tipo I de Factor de Crecimiento Transformador beta/genética , Retina/citología , Retina/metabolismo , Neovascularización Retiniana/patología , Vasos Retinianos , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
9.
Science ; 369(6506)2020 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-32820093

RESUMEN

In developed countries, the leading causes of blindness such as diabetic retinopathy are characterized by disorganized vasculature that can become fibrotic. Although many such pathological vessels often naturally regress and spare sight-threatening complications, the underlying mechanisms remain unknown. Here, we used orthogonal approaches in human patients with proliferative diabetic retinopathy and a mouse model of ischemic retinopathies to identify an unconventional role for neutrophils in vascular remodeling during late-stage sterile inflammation. Senescent vasculature released a secretome that attracted neutrophils and triggered the production of neutrophil extracellular traps (NETs). NETs ultimately cleared diseased endothelial cells and remodeled unhealthy vessels. Genetic or pharmacological inhibition of NETosis prevented the regression of senescent vessels and prolonged disease. Thus, clearance of senescent retinal blood vessels leads to reparative vascular remodeling.


Asunto(s)
Envejecimiento/patología , Retinopatía Diabética/patología , Trampas Extracelulares/inmunología , Vasos Retinianos/patología , Animales , Senescencia Celular , Retinopatía Diabética/inmunología , Modelos Animales de Enfermedad , Células Endoteliales/inmunología , Células Endoteliales/patología , Humanos , Inflamación/inmunología , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Neutrófilos/inmunología , Vasos Retinianos/inmunología
10.
Nat Genet ; 52(1): 40-47, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31844321

RESUMEN

Valvular heart disease is observed in approximately 2% of the general population1. Although the initial observation is often localized (for example, to the aortic or mitral valve), disease manifestations are regularly observed in the other valves and patients frequently require surgery. Despite the high frequency of heart valve disease, only a handful of genes have so far been identified as the monogenic causes of disease2-7. Here we identify two consanguineous families, each with two affected family members presenting with progressive heart valve disease early in life. Whole-exome sequencing revealed homozygous, truncating nonsense alleles in ADAMTS19 in all four affected individuals. Homozygous knockout mice for Adamts19 show aortic valve dysfunction, recapitulating aspects of the human phenotype. Expression analysis using a lacZ reporter and single-cell RNA sequencing highlight Adamts19 as a novel marker for valvular interstitial cells; inference of gene regulatory networks in valvular interstitial cells positions Adamts19 in a highly discriminatory network driven by the transcription factor lymphoid enhancer-binding factor 1 downstream of the Wnt signaling pathway. Upregulation of endocardial Krüppel-like factor 2 in Adamts19 knockout mice precedes hemodynamic perturbation, showing that a tight balance in the Wnt-Adamts19-Klf2 axis is required for proper valve maturation and maintenance.


Asunto(s)
Proteínas ADAMTS/metabolismo , Regulación del Desarrollo de la Expresión Génica , Enfermedades de las Válvulas Cardíacas/etiología , Proteínas ADAMTS/genética , Animales , Familia , Femenino , Enfermedades de las Válvulas Cardíacas/patología , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Ratones , Ratones Noqueados , Linaje , Análisis de la Célula Individual , Vía de Señalización Wnt
11.
Cell Mol Gastroenterol Hepatol ; 7(2): 411-431, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30739867

RESUMEN

BACKGROUND & AIMS: A generalized human pacemaking syndrome, chronic atrial and intestinal dysrhythmia (CAID) (OMIM 616201), is caused by a homozygous SGO1 mutation (K23E), leading to chronic intestinal pseudo-obstruction and arrhythmias. Because CAID patients do not show phenotypes consistent with perturbation of known roles of SGO1, we hypothesized that noncanonical roles of SGO1 drive the clinical manifestations observed. METHODS: To identify a molecular signature for CAID syndrome, we achieved unbiased screens in cell lines and gut tissues from CAID patients vs wild-type controls. We performed RNA sequencing along with stable isotope labeling with amino acids in cell culture. In addition, we determined the genome-wide DNA methylation and chromatin accessibility signatures using reduced representative bisulfite sequencing and assay for transposase-accessible chromatin with high-throughput sequencing. Functional studies included patch-clamp, quantitation of transforming growth factor-ß (TGF-ß) signaling, and immunohistochemistry in CAID patient gut biopsy specimens. RESULTS: Proteome and transcriptome studies converge on cell-cycle regulation, cardiac conduction, and smooth muscle regulation as drivers of CAID syndrome. Specifically, the inward rectifier current, an important regulator of cellular function, was disrupted. Immunohistochemistry confirmed overexpression of Budding Uninhibited By Benzimidazoles 1 (BUB1) in patients, implicating the TGF-ß pathway in CAID pathogenesis. Canonical TGF-ß signaling was up-regulated and uncoupled from noncanonical signaling in CAID patients. Reduced representative bisulfite sequencing and assay for transposase-accessible chromatin with high-throughput sequencing experiments showed significant changes of chromatin states in CAID, pointing to epigenetic regulation as a possible pathologic mechanism. CONCLUSIONS: Our findings point to impaired inward rectifier potassium current, dysregulation of canonical TGF-ß signaling, and epigenetic regulation as potential drivers of intestinal and cardiac manifestations of CAID syndrome. Transcript profiling and genomics data are as follows: repository URL: https://www.ncbi.nlm.nih.gov/geo; SuperSeries GSE110612 was composed of the following subseries: GSE110309, GSE110576, and GSE110601.


Asunto(s)
Anomalías Múltiples/genética , Proteínas de Ciclo Celular/metabolismo , Epigenómica , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Anomalías Múltiples/patología , Anomalías Múltiples/fisiopatología , Adulto , Metilación de ADN/genética , Dermis/patología , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Ontología de Genes , Humanos , Canales de Potasio/metabolismo , Proteoma/metabolismo , Reproducibilidad de los Resultados , Síndrome
12.
Data Brief ; 13: 731-737, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28748208

RESUMEN

The data shown in this article are related to the research article entitled "Characterization of Sgo1 expression pattern in developing and adult mouse" (Song et al., 2017) [3]. The article provides novel data on Sgo1 gene expression pattern utilizing Sgo1_LacZ_Knock in mouse line and immunohistochemistry in wild type mice. The data presents Sgo1 expression pattern during development, and in post-developmental proliferative and quiescent tissue. The article describes following tissues: developing heart, neural tube, adult colon, cerebellum, cerebral cortex, liver, and testis.

13.
Gene Expr Patterns ; 25-26: 36-45, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28465207

RESUMEN

SGO1 has been characterized in its function in correct cell division and its role in centrosome cohesion in the nucleus. However, its organ-specific maturation-related expression pattern in vivo remains largely uncharacterized. Here, we show clear SGO1 expression in post-developmental neuronal cells and cytoplasmic localisation in nucleated cells with a transgenic mice model and immunohistochemistry of wild type mice. We demonstrate extranuclear expression of Sgo1 in the developing heart and gut, which have been shown to be dysregulated in humans with homozygous SGO1 mutation. Additionally, we show Sgo1 expression in select population of retinal cells in developing and post-developmental retina. Our expression analysis strongly suggests that the function of SGO1 goes beyond its well characterized role in cell division.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Citoplasma/metabolismo , Embrión de Mamíferos/metabolismo , Animales , División Celular , Citoplasma/genética , Tracto Gastrointestinal/crecimiento & desarrollo , Tracto Gastrointestinal/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células HeLa , Corazón/crecimiento & desarrollo , Humanos , Ratones , Ratones Transgénicos , Miocardio/metabolismo , Neuronas/metabolismo , Especificidad de Órganos , Retina/crecimiento & desarrollo , Retina/metabolismo , Distribución Tisular
14.
Can J Cardiol ; 32(1): 135.e1-7, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26148450

RESUMEN

BACKGROUND: We report a 13-year-old female patient followed since birth for multiple rare congenital defects, including hypotrichosis, telangiectasia, and severe dilatation of the ascending aorta. METHODS: Comprehensive phenotype assessment throughout childhood included repeated echocardiographic measurements, evaluation of renal function, and immunohistochemical analysis of skin biopsy samples. Whole-exome sequencing was performed for the patient and both unaffected parents. RESULTS: We identified a novel de novo mutation in the transcription factor SOX18 (c.481C>T:p.Gln161*) in the patient, which was absent in all unaffected family members. Echocardiography revealed early onset and progressive dilatation of the ascending aorta. Skin biopsy results confirmed the defects of the blood vasculature in the presence of intact lymphatic vessels. Assessment of renal function did not show any signs of renal problems or renal failure in the patient. CONCLUSIONS: The genetic finding of a pathogenic SOX18 mutation enabled the diagnosis of the rare hypotrichosis-lymphedema-telangiectasia syndrome in our patient. The identification of a novel stop gain mutation in the SOX18 gene in association with dilatation of the aorta highlights the importance of this gene during the development of the circulatory system. Our study highlights the importance of whole-exome sequencing in the rapid identification of genes and gene mutations involved in rare conditions and thus expanding the knowledge and spectrum of clinical manifestations associated with them.


Asunto(s)
Aneurisma de la Aorta Torácica/genética , ADN/genética , Hipotricosis/genética , Linfedema/genética , Mutación , Factores de Transcripción SOXF/genética , Telangiectasia/genética , Adolescente , Aneurisma de la Aorta Torácica/diagnóstico , Aneurisma de la Aorta Torácica/metabolismo , Biopsia , Análisis Mutacional de ADN , Ecocardiografía , Femenino , Humanos , Hipotricosis/diagnóstico , Hipotricosis/metabolismo , Linfedema/diagnóstico , Linfedema/metabolismo , Fenotipo , Reacción en Cadena de la Polimerasa , Factores de Transcripción SOXF/metabolismo , Telangiectasia/diagnóstico , Telangiectasia/metabolismo
15.
Brain Res Mol Brain Res ; 107(2): 201-13, 2002 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-12425948

RESUMEN

The aim of the present study was to examine the glucuronidation of a series of odorant molecules by homogenates prepared either with rat olfactory mucosa, olfactory bulb or brain. Most of the odorant molecules tested were efficiently conjugated by olfactory mucosa, whereas olfactory bulb and brain homogenates displayed lower activities and glucuronidated only a few molecules. Important age-related changes in glucuronidation efficiency were observed in olfactory mucosa and bulb. Therefore, we studied changes in expression of two UDP-glucuronosyltransferase isoforms, UGT1A6 and UGT2A1, in 1-day, 1- and 2-week-, 3-, 12- and 24-month-old rats. UGT1A6 was expressed at the same transcriptional level in the olfactory mucosa, bulb and brain, throughout the life period studied. UGT2A1 mRNA was expressed in both olfactory mucosa and olfactory bulb, in accordance with previous results [Mol. Brain Res. 90 (2001) 83], but UGT2A1 transcriptional level was 400-4000 times higher than that of UGT1A6. Moreover, age-dependent variations in UGT2A1 mRNA expression were observed. As it has been suggested that drug metabolizing enzymes could participate in olfactory function, mitral cell electrical activity was recorded during exposure to different odorant molecules in young, adult and old animals. Age-related changes in the amplitude of response after stimulation with several odorant molecules were observed, and the highest responses were obtained with molecules that were not efficiently glucuronidated by olfactory mucosa. In conclusion, the present work presents new evidence of the involvement of UGT activity in some steps of the olfactory process.


Asunto(s)
Glucuronosiltransferasa/metabolismo , Proteínas de Transporte de Monosacáridos , Neuronas/metabolismo , Vías Olfatorias/enzimología , Receptores Odorantes/metabolismo , Olfato/fisiología , Telencéfalo/enzimología , Uridina Difosfato Ácido Glucurónico/metabolismo , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Envejecimiento/metabolismo , Animales , Animales Recién Nacidos , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/enzimología , Regulación Enzimológica de la Expresión Génica/fisiología , Glucuronosiltransferasa/efectos de los fármacos , Masculino , Neuronas/efectos de los fármacos , Odorantes , Bulbo Olfatorio/efectos de los fármacos , Bulbo Olfatorio/enzimología , Vías Olfatorias/efectos de los fármacos , Neuronas Receptoras Olfatorias/efectos de los fármacos , Neuronas Receptoras Olfatorias/enzimología , Isoformas de Proteínas/efectos de los fármacos , Isoformas de Proteínas/metabolismo , Ratas , Ratas Wistar , Receptores Odorantes/efectos de los fármacos , Olfato/efectos de los fármacos , Telencéfalo/efectos de los fármacos
16.
J Drug Target ; 10(4): 285-96, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12164377

RESUMEN

It is generally accepted that the rate of entry into and distribution of drugs and other xenobiotics within the central nervous system (CNS) depends on the particular anatomy of the brain microvessels forming the blood-brain barrier (BBB), and of the choroid plexus forming the blood-cerebrospinal fluid barrier (CSF), which possess tight junctions preventing the passage of most polar substances. Drug entry to the CNS also depends on the physicochemical properties of the substances, which can be metabolised during this transport to pharmacologically inactive, non-penetrating polar products. Finally, the entry of drugs may be prevented by multiple complex specialized carriers, which are able to catalyse the active transport of numerous drugs and xenobiotics out of the CNS. Nasal delivery is currently considered as an efficient tool for systemic administration of drugs that are poorly absorbed via the oral route, and increasing evidence suggests that numerous drugs and potentially toxic xenobiotics can reach the CNS by this route. This short review summarizes recent knowledge on factors controlling the nasal pathway, focusing on drug metabolising enzymes in olfactory mucosa, olfactory bulb and brain, which should constitute a CNS metabolic barrier.


Asunto(s)
Barrera Hematoencefálica/fisiología , Encéfalo/metabolismo , Cavidad Nasal/metabolismo , Administración Intranasal , Animales , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Barrera Hematoencefálica/efectos de los fármacos , Humanos , Cavidad Nasal/efectos de los fármacos
17.
Nat Genet ; 46(11): 1245-9, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25282101

RESUMEN

The pacemaking activity of specialized tissues in the heart and gut results in lifelong rhythmic contractions. Here we describe a new syndrome characterized by Chronic Atrial and Intestinal Dysrhythmia, termed CAID syndrome, in 16 French Canadians and 1 Swede. We show that a single shared homozygous founder mutation in SGOL1, a component of the cohesin complex, causes CAID syndrome. Cultured dermal fibroblasts from affected individuals showed accelerated cell cycle progression, a higher rate of senescence and enhanced activation of TGF-ß signaling. Karyotypes showed the typical railroad appearance of a centromeric cohesion defect. Tissues derived from affected individuals displayed pathological changes in both the enteric nervous system and smooth muscle. Morpholino-induced knockdown of sgol1 in zebrafish recapitulated the abnormalities seen in humans with CAID syndrome. Our findings identify CAID syndrome as a novel generalized dysrhythmia, suggesting a new role for SGOL1 and the cohesin complex in mediating the integrity of human cardiac and gut rhythm.


Asunto(s)
Anomalías Múltiples/genética , Arritmias Cardíacas/genética , Proteínas de Ciclo Celular/genética , Proteínas Cromosómicas no Histona/genética , Enfermedades Intestinales/genética , Contracción Muscular/fisiología , Transducción de Señal/genética , Animales , Arritmias Cardíacas/patología , Ciclo Celular/genética , Sistema Nervioso Entérico/patología , Fibroblastos , Efecto Fundador , Tracto Gastrointestinal/fisiopatología , Técnicas de Silenciamiento del Gen , Humanos , Enfermedades Intestinales/fisiopatología , Cariotipificación , Contracción Muscular/genética , Músculo Liso Vascular/patología , Mutación/genética , Quebec , Síndrome , Factor de Crecimiento Transformador beta/metabolismo , Pez Cebra , Cohesinas
18.
Clin Sci (Lond) ; 110(6): 645-54, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16417466

RESUMEN

In the present study, we have investigated the effect of (i) ET-1 (endothelin-1) and its precursor, big ET-1, on MMP (matrix metalloproteinase)-2 and MMP-9 synthesis and activity in osteosarcoma tissue, and (ii) ET-1 receptor antagonists on cell invasion. Using Western blotting, zymography, RT-PCR (reverse transcription-PCR), immunohistochemistry, immunofluorescence and Northern blotting, we have shown that ET-1 and ET-1 receptors (ET(A) and ET(B)) were expressed in these cells. Additionally, we have demonstrated that ET-1 markedly induced the synthesis and activity of MMP-2, which was significantly increased when compared with MMP-9. Furthermore, inhibition of NF-kappaB (nuclear factor kappaB) activation blocked MMP-2 production and activity, indicating the involvement of NF-kappaB, a ubiquitous transcription factor playing a central role in the differentiation, proliferation and malignant transformation. Since ET-1 acts as an autocrine mediator through gelatinase induction and because inhibition of ET(A) receptor is beneficial for reducing both basal and ET-1-induced osteosarcoma cell invasion, targeting this receptor could be an attractive therapeutic alternative for the successful treatment of osteosarcoma.


Asunto(s)
Neoplasias Óseas/metabolismo , Endotelina-1/farmacología , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , FN-kappa B/metabolismo , Osteosarcoma/metabolismo , Adolescente , Adulto , Antioxidantes/farmacología , Northern Blotting/métodos , Western Blotting/métodos , Línea Celular Tumoral , Niño , Activación Enzimática , Femenino , Humanos , Inmunohistoquímica , Masculino , Metaloproteinasa 2 de la Matriz/análisis , Metaloproteinasa 9 de la Matriz/análisis , Persona de Mediana Edad , FN-kappa B/análisis , Pirrolidinas/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estadísticas no Paramétricas , Tiocarbamatos/farmacología , Inhibidor Tisular de Metaloproteinasa-1/análisis , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Inhibidor Tisular de Metaloproteinasa-2/análisis , Inhibidor Tisular de Metaloproteinasa-2/metabolismo , Células Tumorales Cultivadas
19.
Dev Dyn ; 233(4): 1525-34, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15965983

RESUMEN

Urodele amphibians (e.g., axolotls) have the unique ability, among vertebrates, to regenerate perfectly many parts of their body after amputation. The limb has been the most widely studied regenerating structure in these organisms and provides an ideal model in which to understand how vertebrates can regenerate complex tissues. The present study focuses on Hsp-70, a member of the stress-related heat-shock protein family. This protein is normally induced after a stress or trauma such as heat-shock, ultraviolet irradiation, or wounding. Thus, studying its expression during axolotl limb regeneration, a response to an important traumatic event (amputation), is of great interest to further understand how the regenerative process is mediated. Using molecular biology and biochemical techniques, we have characterized both the spatiotemporal and quantitative expression patterns of Hsp-70 in axolotl development and regeneration. Our results show that Hsp-70 is expressed and regulated during axolotl development as in other vertebrates. Our data also demonstrate an up-regulation of the RNA transcript for Hsp-70 during limb regeneration as early as 24 hr after amputation that is maintained up to early differentiation. We also demonstrate a similar pattern of expression for the protein during regeneration. Finally, we show that axolotl Hsp-70 is induced threefold after heat-shock as observed in other vertebrates.


Asunto(s)
Extremidades/crecimiento & desarrollo , Proteínas HSP70 de Choque Térmico/genética , Regeneración/fisiología , Ambystoma mexicanum , Animales , Línea Celular , Proteínas HSP70 de Choque Térmico/biosíntesis , Calor , Larva
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA