Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-38826421

RESUMEN

Monogenic syndromes are associated with neurodevelopmental changes that result in cognitive impairments, neurobehavioral phenotypes including autism and attention deficit hyperactivity disorder (ADHD), and seizures. Limited studies and resources are available to make meaningful headway into the underlying molecular mechanisms that result in these symptoms. One such example is DeSanto-Shinawi Syndrome (DESSH), a rare disorder caused by pathogenic variants in the WAC gene. Individuals with DESSH syndrome exhibit a recognizable craniofacial gestalt, developmental delay/intellectual disability, neurobehavioral symptoms that include autism, ADHD, behavioral difficulties and seizures. However, no thorough studies from a vertebrate model exist to understand how these changes occur. To overcome this, we developed both murine and zebrafish Wac/wac deletion mutants and studied whether their phenotypes recapitulate those described in individuals with DESSH syndrome. We show that the two Wac models exhibit craniofacial and behavioral changes, reminiscent of abnormalities found in DESSH syndrome. In addition, each model revealed impacts to GABAergic neurons and further studies showed that the mouse mutants are susceptible to seizures, changes in brain volumes that are different between sexes and relevant behaviors. Finally, we uncovered transcriptional impacts of Wac loss of function that will pave the way for future molecular studies into DESSH. These studies begin to uncover some biological underpinnings of DESSH syndrome and elucidate the biology of Wac, with advantages in each model.

2.
Elife ; 102021 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-34605404

RESUMEN

Enhancers are cis-regulatory elements that play critical regulatory roles in modulating developmental transcription programs and driving cell-type-specific and context-dependent gene expression in the brain. The development of massively parallel reporter assays (MPRAs) has enabled high-throughput functional screening of candidate DNA sequences for enhancer activity. Tissue-specific screening of in vivo enhancer function at scale has the potential to greatly expand our understanding of the role of non-coding sequences in development, evolution, and disease. Here, we adapted a self-transcribing regulatory element MPRA strategy for delivery to early postnatal mouse brain via recombinant adeno-associated virus (rAAV). We identified and validated putative enhancers capable of driving reporter gene expression in mouse forebrain, including regulatory elements within an intronic CACNA1C linkage disequilibrium block associated with risk in neuropsychiatric disorder genetic studies. Paired screening and single enhancer in vivo functional testing, as we show here, represents a powerful approach towards characterizing regulatory activity of enhancers and understanding how enhancer sequences organize gene expression in the brain.


Asunto(s)
Encéfalo/metabolismo , Elementos de Facilitación Genéticos , Animales , Encéfalo/crecimiento & desarrollo , Secuenciación de Nucleótidos de Alto Rendimiento , Ratones
3.
Genome Med ; 13(1): 69, 2021 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-33910599

RESUMEN

BACKGROUND: Genes with multiple co-active promoters appear common in brain, yet little is known about functional requirements for these potentially redundant genomic regulatory elements. SCN1A, which encodes the NaV1.1 sodium channel alpha subunit, is one such gene with two co-active promoters. Mutations in SCN1A are associated with epilepsy, including Dravet syndrome (DS). The majority of DS patients harbor coding mutations causing SCN1A haploinsufficiency; however, putative causal non-coding promoter mutations have been identified. METHODS: To determine the functional role of one of these potentially redundant Scn1a promoters, we focused on the non-coding Scn1a 1b regulatory region, previously described as a non-canonical alternative transcriptional start site. We generated a transgenic mouse line with deletion of the extended evolutionarily conserved 1b non-coding interval and characterized changes in gene and protein expression, and assessed seizure activity and alterations in behavior. RESULTS: Mice harboring a deletion of the 1b non-coding interval exhibited surprisingly severe reductions of Scn1a and NaV1.1 expression throughout the brain. This was accompanied by electroencephalographic and thermal-evoked seizures, and behavioral deficits. CONCLUSIONS: This work contributes to functional dissection of the regulatory wiring of a major epilepsy risk gene, SCN1A. We identified the 1b region as a critical disease-relevant regulatory element and provide evidence that non-canonical and seemingly redundant promoters can have essential function.


Asunto(s)
Epilepsia/genética , Regulación de la Expresión Génica , Canal de Sodio Activado por Voltaje NAV1.1/genética , Eliminación de Secuencia/genética , Animales , Atención , Secuencia de Bases , Encéfalo/metabolismo , Encéfalo/patología , Cromatina/metabolismo , Secuencia Conservada/genética , Modelos Animales de Enfermedad , Electroencefalografía , Epilepsia/diagnóstico por imagen , Evolución Molecular , Femenino , Células HEK293 , Heterocigoto , Homocigoto , Humanos , Masculino , Aprendizaje por Laberinto , Trastornos de la Memoria/genética , Ratones Endogámicos C57BL , Neuronas/metabolismo , Prueba de Campo Abierto , Fenotipo , Unión Proteica , Secuencias Reguladoras de Ácidos Nucleicos/genética , Análisis de Supervivencia , Temperatura , Transactivadores/metabolismo
4.
Elife ; 102021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33666173

RESUMEN

In utero exposure to maternal immune activation (MIA) is an environmental risk factor for neurodevelopmental and neuropsychiatric disorders. Animal models provide an opportunity to identify mechanisms driving neuropathology associated with MIA. We performed time-course transcriptional profiling of mouse cortical development following induced MIA via poly(I:C) injection at E12.5. MIA-driven transcriptional changes were validated via protein analysis, and parallel perturbations to cortical neuroanatomy were identified via imaging. MIA-induced acute upregulation of genes associated with hypoxia, immune signaling, and angiogenesis, by 6 hr following exposure. This acute response was followed by changes in proliferation, neuronal and glial specification, and cortical lamination that emerged at E14.5 and peaked at E17.5. Decreased numbers of proliferative cells in germinal zones and alterations in neuronal and glial populations were identified in the MIA-exposed cortex. Overall, paired transcriptomic and neuroanatomical characterization revealed a sequence of perturbations to corticogenesis driven by mid-gestational MIA.


Asunto(s)
Encéfalo/embriología , Neurogénesis , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones Endogámicos C57BL , Trastornos del Neurodesarrollo , Poli I-C/inmunología , Embarazo , Transcriptoma
5.
Proc Natl Acad Sci U S A ; 117(37): 23073-23084, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32873638

RESUMEN

The small GTPase ARL4C participates in the regulation of cell migration, cytoskeletal rearrangements, and vesicular trafficking in epithelial cells. The ARL4C signaling cascade starts by the recruitment of the ARF-GEF cytohesins to the plasma membrane, which, in turn, bind and activate the small GTPase ARF6. However, the role of ARL4C-cytohesin-ARF6 signaling during hippocampal development remains elusive. Here, we report that the E3 ubiquitin ligase Cullin 5/RBX2 (CRL5) controls the stability of ARL4C and its signaling effectors to regulate hippocampal morphogenesis. Both RBX2 knockout and Cullin 5 knockdown cause hippocampal pyramidal neuron mislocalization and development of multiple apical dendrites. We used quantitative mass spectrometry to show that ARL4C, Cytohesin-1/3, and ARF6 accumulate in the RBX2 mutant telencephalon. Furthermore, we show that depletion of ARL4C rescues the phenotypes caused by Cullin 5 knockdown, whereas depletion of CYTH1 or ARF6 exacerbates overmigration. Finally, we show that ARL4C, CYTH1, and ARF6 are necessary for the dendritic outgrowth of pyramidal neurons to the superficial strata of the hippocampus. Overall, we identified CRL5 as a key regulator of hippocampal development and uncovered ARL4C, CYTH1, and ARF6 as CRL5-regulated signaling effectors that control pyramidal neuron migration and dendritogenesis.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Proteínas Cullin/metabolismo , Hipocampo/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Morfogénesis/fisiología , Factor 6 de Ribosilación del ADP , Animales , Membrana Celular/metabolismo , Movimiento Celular/fisiología , Dendritas/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Ratones , Neurogénesis/fisiología , Células Piramidales/metabolismo , Transducción de Señal/fisiología , Ubiquitina-Proteína Ligasas/metabolismo
6.
J Vis Exp ; (114)2016 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-27584943

RESUMEN

The skin is essential for our survival. The outer epidermal layer consists of the interfollicular epidermis, which is a stratified squamous epithelium covering most of our body, and epidermal appendages such as the hair follicles and sweat glands. The epidermis undergoes regeneration throughout life and in response to injury. This is enabled by K14-expressing basal epidermal stem/progenitor cell populations that are tightly regulated by multiple regulatory mechanisms active within the epidermis and between epidermis and dermis. This article describes a simple method to clarify full thickness mouse skin biopsies, and visualize K14 protein expression patterns, Ki67 labeled proliferating cells, Nile Red labeled sebocytes, and DAPI nuclear labeling at single cell resolution in 3D. This method enables accurate assessment and quantification of skin anatomy and pathology, and of abnormal epidermal phenotypes in genetically modified mouse lines. The CUBIC protocol is the best method available to date to investigate molecular and cellular interactions in full thickness skin biopsies at single cell resolution.


Asunto(s)
Microscopía Confocal/métodos , Análisis de la Célula Individual/métodos , Piel/citología , Piel/metabolismo , Animales , Biopsia/métodos , Folículo Piloso/citología , Folículo Piloso/diagnóstico por imagen , Folículo Piloso/metabolismo , Queratinocitos/citología , Queratinocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Regeneración/fisiología , Glándulas Sebáceas/citología , Glándulas Sebáceas/diagnóstico por imagen , Glándulas Sebáceas/metabolismo , Piel/diagnóstico por imagen , Células Madre/citología , Células Madre/metabolismo , Técnicas de Cultivo de Tejidos/métodos
7.
BMC Genomics ; 17: 450, 2016 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-27295951

RESUMEN

BACKGROUND: Williams-Beuren Syndrome (WBS) is a genetic disorder associated with multisystemic abnormalities, including craniofacial dysmorphology and cognitive defects. It is caused by a hemizygous microdeletion involving up to 28 genes in chromosome 7q11.23. Genotype/phenotype analysis of atypical microdeletions implicates two evolutionary-related transcription factors, GTF2I and GTF2IRD1, as prime candidates for the cause of the facial dysmorphology. RESULTS: Using a targeted Gtf2ird1 knockout mouse, we employed massively-parallel sequencing of mRNA (RNA-Seq) to understand changes in the transcriptional landscape associated with inactivation of Gtf2ird1 in lip tissue. We found widespread dysregulation of genes including differential expression of 78 transcription factors or coactivators, several involved in organ development including Hey1, Myf6, Myog, Dlx2, Gli1, Gli2, Lhx2, Pou3f3, Sox2, Foxp3. We also found that the absence of GTF2IRD1 is associated with increased expression of genes involved in cellular proliferation, including growth factors consistent with the observed phenotype of extreme thickening of the epidermis. At the same time, there was a decrease in the expression of genes involved in other signalling mechanisms, including the Wnt pathway, indicating dysregulation in the complex networks necessary for epidermal differentiation and facial skin patterning. Several of the differentially expressed genes have known roles in both tissue development and neurological function, such as the transcription factor Lhx2 which regulates several genes involved in both skin and brain development. CONCLUSIONS: Gtf2ird1 inactivation results in widespread gene dysregulation, some of which may be due to the secondary consequences of gene regulatory network disruptions involving several transcription factors and signalling molecules. Genes involved in growth factor signalling and cell cycle progression were identified as particularly important for explaining the skin dysmorphology observed in this mouse model. We have noted that a number of the dysregulated genes have known roles in brain development as well as epidermal differentiation and maintenance. Therefore, this study provides clues as to the underlying mechanisms that may be involved in the broader profile of WBS.


Asunto(s)
Epidermis/metabolismo , Estudios de Asociación Genética , Proteínas Musculares/genética , Proteínas Nucleares/genética , Transactivadores/genética , Síndrome de Williams/genética , Animales , Análisis por Conglomerados , Biología Computacional/métodos , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ontología de Genes , Secuenciación de Nucleótidos de Alto Rendimiento , Ratones , Ratones Noqueados , Modelos Biológicos , Proteínas Musculares/deficiencia , Proteínas Musculares/metabolismo , Proteínas Nucleares/deficiencia , Proteínas Nucleares/metabolismo , Fenotipo , Reproducibilidad de los Resultados , Transducción de Señal , Transactivadores/deficiencia , Transactivadores/metabolismo , Síndrome de Williams/diagnóstico , Síndrome de Williams/metabolismo
8.
Hum Genet ; 134(10): 1099-115, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26275350

RESUMEN

GTF2IRD1 is one of the three members of the GTF2I gene family, clustered on chromosome 7 within a 1.8 Mb region that is prone to duplications and deletions in humans. Hemizygous deletions cause Williams-Beuren syndrome (WBS) and duplications cause WBS duplication syndrome. These copy number variations disturb a variety of developmental systems and neurological functions. Human mapping data and analyses of knockout mice show that GTF2IRD1 and GTF2I underpin the craniofacial abnormalities, mental retardation, visuospatial deficits and hypersociability of WBS. However, the cellular role of the GTF2IRD1 protein is poorly understood due to its very low abundance and a paucity of reagents. Here, for the first time, we show that endogenous GTF2IRD1 has a punctate pattern in the nuclei of cultured human cell lines and neurons. To probe the functional relationships of GTF2IRD1 in an unbiased manner, yeast two-hybrid libraries were screened, isolating 38 novel interaction partners, which were validated in mammalian cell lines. These relationships illustrate GTF2IRD1 function, as the isolated partners are mostly involved in chromatin modification and transcriptional regulation, whilst others indicate an unexpected role in connection with the primary cilium. Mapping of the sites of protein interaction also indicates key features regarding the evolution of the GTF2IRD1 protein. These data provide a visual and molecular basis for GTF2IRD1 nuclear function that will lead to an understanding of its role in brain, behaviour and human disease.


Asunto(s)
Núcleo Celular/metabolismo , Cromatina/metabolismo , Proteínas Musculares/metabolismo , Proteínas Nucleares/metabolismo , Transactivadores/metabolismo , Ensamble y Desensamble de Cromatina , Cilios/metabolismo , Proteínas de Unión al ADN/metabolismo , Células HEK293 , Células HeLa , Humanos , Proteínas Musculares/química , Proteínas Nucleares/química , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas , Transactivadores/química , Técnicas del Sistema de Dos Híbridos
9.
Br J Med Med Res ; 5(10): 1198-2012, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-34012910

RESUMEN

RATIONALE: Mutations in Transient Receptor Potential Channel 6 (TRPC6) gene are associated with autosomal dominant focal and segmental glomerulosclerosis (FSGS). The majority of the identified mutations affect the ion channel function. Since calcium channels are promising candidate drug targets, there is an an urgent need for a mouse model to assess new therapeutic drugs and to help delineate the pathogenic process leading to FSGS. We have previously reported the generation of three independent transgenic mouse lines carrying different Trpc6 mutations that display a glomerular disease comparable to the phenotype presented by individuals with FSGS. However, the utility of these models for drug testing is dampened by the late-onset of the presentation and the mild phenotypic manifestations. METHODOLOGY: In order to obtain a time-effective mouse model for Trpc6-associated FSGS we generated a new transgenic mutant Trpc6 mouse model emulating the amino acid change carried by the first pediatric patient of FSGS associated with a TRPC6 mutation: M132T. RESULTS: Mice carrying the orthologous Trpc6 M131T transgene showed early onset proteinuria and early signs of FSGS. When exploring molecular consequences of the overexpression of this mutated form of Trpc6 in podocytes, differences in expression levels of Axin2 and ß-catenin were found in glomeruli from transgenic Trpc6 M131T mice. These data supports the proposed molecular mechanisms related to the activation of calcineurin-NFAT/Wnt signaling, as outcome of the increased calcium influx caused by the mutated form of Trpc6. CONCLUSION: Given that the Trpc6 M131T mouse develops an early onset of FSGS-like phenotypes it represents a promising model for studying the pathogenesis of FSGS caused by TRpC6, facilitating the assessment of new drugs as treatments and allowing further studies to understand underlying molecular pathways involved in the development of the TRPC6 mediated disease.

10.
Eur J Hum Genet ; 23(6): 774-80, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25248400

RESUMEN

Williams-Beuren Syndrome (WBS) is a rare genetic condition caused by a hemizygous deletion involving up to 28 genes within chromosome 7q11.23. Among the spectrum of physical and neurological defects in WBS, it is common to find a distinctive response to sound stimuli that includes extreme adverse reactions to loud, or sudden sounds and a fascination with certain sounds that may manifest as strengths in musical ability. However, hearing tests indicate that sensorineural hearing loss (SNHL) is frequently found in WBS patients. The functional and genetic basis of this unusual auditory phenotype is currently unknown. Here, we investigated the potential involvement of GTF2IRD1, a transcription factor encoded by a gene located within the WBS deletion that has been implicated as a contributor to the WBS assorted neurocognitive profile and craniofacial abnormalities. Using Gtf2ird1 knockout mice, we have analysed the expression of the gene in the inner ear and examined hearing capacity by evaluating the auditory brainstem response (ABR) and the distortion product of otoacoustic emissions (DPOAE). Our results show that Gtf2ird1 is expressed in a number of cell types within the cochlea, and Gtf2ird1 null mice showed higher auditory thresholds (hypoacusis) in both ABR and DPOAE hearing assessments. These data indicate that the principal hearing deficit in the mice can be traced to impairments in the amplification process mediated by the outer hair cells and suggests that similar mechanisms may underpin the SNHL experienced by WBS patients.


Asunto(s)
Umbral Auditivo , Proteínas Musculares/genética , Proteínas Nucleares/genética , Transactivadores/genética , Síndrome de Williams/genética , Animales , Cóclea/citología , Cóclea/metabolismo , Cóclea/fisiología , Potenciales Evocados Auditivos del Tronco Encefálico , Ratones , Ratones Endogámicos C57BL , Proteínas Musculares/metabolismo , Proteínas Nucleares/metabolismo , Emisiones Otoacústicas Espontáneas , Transactivadores/metabolismo , Síndrome de Williams/fisiopatología
11.
PLoS One ; 7(9): e45155, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23028815

RESUMEN

Smith-Magenis Syndrome (SMS) is a complex genomic disorder mostly caused by the haploinsufficiency of the Retinoic Acid Induced 1 gene (RAI1), located in the chromosomal region 17p11.2. In a subset of SMS patients, heterozygous mutations in RAI1 are found. Here we investigate the molecular properties of these mutated forms and their relationship with the resulting phenotype. We compared the clinical phenotype of SMS patients carrying a mutation in RAI1 coding region either in the N-terminal or the C-terminal half of the protein and no significant differences were found. In order to study the molecular mechanism related to these two groups of RAI1 mutations first we analyzed those mutations that result in the truncated protein corresponding to the N-terminal half of RAI1 finding that they have cytoplasmic localization (in contrast to full length RAI1) and no ability to activate the transcription through an endogenous target: the BDNF enhancer. Similar results were found in lymphoblastoid cells derived from a SMS patient carrying RAI1 c.3103insC, where both mutant and wild type products of RAI1 were detected. The wild type form of RAI1 was found in the chromatin bound and nuclear matrix subcellular fractions while the mutant product was mainly cytoplasmic. In addition, missense mutations at the C-terminal half of RAI1 presented a correct nuclear localization but no activation of the endogenous target. Our results showed for the first time a correlation between RAI1 mutations and abnormal protein function plus they suggest that a reduction of total RAI1 transcription factor activity is at the heart of the SMS clinical presentation.


Asunto(s)
Mutación/genética , Síndrome de Smith-Magenis/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Cromatina/metabolismo , Elementos de Facilitación Genéticos/genética , Genes Reporteros , Humanos , Linfocitos/metabolismo , Ratones , Proteínas Mutantes/metabolismo , Fenotipo , Estructura Terciaria de Proteína , Transporte de Proteínas , Síndrome de Smith-Magenis/patología , Fracciones Subcelulares/metabolismo , Transactivadores , Factores de Transcripción/química , Transcripción Genética , Activación Transcripcional/genética
12.
Am J Med Genet A ; 158A(7): 1579-88, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22639462

RESUMEN

Potocki-Lupski syndrome is a genomic disorder caused by duplication of 17p11.2. It is characterized by failure to thrive, intellectual disability, hypotonia, and behavioral difficulties. Structural renal anomalies have been observed in <10% of affected individuals. We present detailed clinical and molecular data on six patients with Potocki-Lupski syndrome, two of whom had renal abnormalities, and investigate the prevalence of kidney abnormalities in the mouse model for the syndrome. In contrast to affected humans, the mouse model does not demonstrate a renal phenotype. Comparison of the duplicated segment in patients with Potocki-Lupski syndrome and the renal phenotype and the syntenic duplicated region in the mouse model allowed us to suggest a 0.285 Mb critical region, including the FLCN gene that may be important for development of renal abnormalities in patients with this duplication.


Asunto(s)
Riñón/anomalías , Síndrome de Smith-Magenis/genética , Anomalías Múltiples , Adolescente , Animales , Niño , Preescolar , Bandeo Cromosómico , Trastornos de los Cromosomas , Duplicación Cromosómica , Mapeo Cromosómico , Cromosomas Humanos Par 17 , Modelos Animales de Enfermedad , Femenino , Duplicación de Gen , Humanos , Lactante , Riñón/patología , Masculino , Ratones , Fenotipo , Síndrome de Smith-Magenis/complicaciones , Sistema Urinario/anomalías
13.
EMBO Mol Med ; 3(1): 1-4, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21204264

RESUMEN

Copy number variations (CNV) within the genome are extremely abundant. In this closeup, Canales and Walz discuss how CNV are associated with normal variation, genomic disorders, genome evolution, adaptive traits and how the use of a novel screen described by Ermakova et al in this issue that is designed to identify human diseaserelevant phenotypes associated with CNV in the mouse can help elucidating susceptibility or predisposition to diseases loci.


Asunto(s)
Dosificación de Gen , Predisposición Genética a la Enfermedad , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Genoma Humano , Genotipo , Humanos , Ratones , Proteínas de la Mielina/genética , Proteínas de la Mielina/metabolismo , Fenotipo , Proteínas/genética , Proteínas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
14.
PLoS One ; 5(9): e12859, 2010 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-20877463

RESUMEN

Mutations in the TRPC6 calcium channel (Transient receptor potential channel 6) gene have been associated with familiar forms of Focal and Segmental Glomerulosclerosis (FSGS) affecting children and adults. In addition, acquired glomerular diseases are associated with increased expression levels of TRPC6. However, the exact role of TRPC6 in the pathogenesis of FSGS remains to be elucidated. In this work we describe the generation and phenotypic characterization of three different transgenic mouse lines with podocyte-specific overexpression of the wild type or any of two mutant forms of Trpc6 (P111Q and E896K) previously related to FSGS. Consistent with the human phenotype a non-nephrotic range of albuminuria was detectable in almost all transgenic lines. The histological analysis demonstrated that the transgenic mice developed a kidney disease similar to human FSGS. Differences of 2-3 folds in the presence of glomerular lesions were found between the non transgenic and transgenic mice expressing Trpc6 in its wild type or mutant forms specifically in podocytes. Electron microscopy of glomerulus from transgenic mice showed extensive podocyte foot process effacement. We conclude that overexpression of Trpc6 (wild type or mutated) in podocytes is sufficient to cause a kidney disease consistent with FSGS. Our results contribute to reinforce the central role of podocytes in the etiology of FSGS. These mice constitute an important new model in which to study future therapies and outcomes of this complex disease.


Asunto(s)
Expresión Génica , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Podocitos/metabolismo , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/metabolismo , Animales , Línea Celular , Modelos Animales de Enfermedad , Femenino , Glomeruloesclerosis Focal y Segmentaria/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Mutación , Especificidad de Órganos , Canal Catiónico TRPC6
15.
BMC Mol Biol ; 11: 63, 2010 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-20738874

RESUMEN

BACKGROUND: Smith-Magenis Syndrome is a contiguous gene syndrome in which the dosage sensitive gene has been identified: the Retinoic Acid Induced 1 (RAI1). Little is known about the function of human RAI1. RESULTS: We generated the full-length cDNA of the wild type protein and five mutated forms: RAI1-HA 2687delC, RAI1-HA 3103delC, RAI1 R960X, RAI1-HA Q1562R, and RAI1-HA S1808N. Four of them have been previously associated with SMS clinical phenotype. Molecular weight, subcellular localization and transcription factor activity of the wild type and mutant forms were studied by western blot, immunofluorescence and luciferase assays respectively. The wild type protein and the two missense mutations presented a higher molecular weight than expected, localized to the nucleus and activated transcription of a reporter gene. The frameshift mutations generated a truncated polypeptide with transcription factor activity but abnormal subcellular localization, and the same was true for the 1-960aa N-terminal half of RAI1. Two different C-terminal halves of the RAI1 protein (1038aa-end and 1229aa-end) were able to localize into the nucleus but had no transactivation activity. CONCLUSION: Our results indicate that transcription factor activity and subcellular localization signals reside in two separate domains of the protein and both are essential for the correct functionality of RAI1. The pathogenic outcome of some of the mutated forms can be explained by the dissociation of these two domains.


Asunto(s)
Mutación , Síndrome de Smith-Magenis/genética , Factores de Transcripción/genética , Animales , Línea Celular , Mapeo Cromosómico , Análisis Mutacional de ADN , Dosificación de Gen , Genes Reporteros , Humanos , Ratones , Datos de Secuencia Molecular , Síndrome de Smith-Magenis/fisiopatología , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/metabolismo
16.
Hum Mol Genet ; 17(16): 2486-95, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18469339

RESUMEN

The Potocki-Lupski syndrome (PTLS) is associated with a microduplication of 17p11.2. Clinical features include multiple congenital and neurobehavioral abnormalities and autistic features. We have generated a PTLS mouse model, Dp(11)17/+, that recapitulates some of the physical and neurobehavioral phenotypes present in patients. Here, we investigated the social behavior and gene expression pattern of this mouse model in a pure C57BL/6-Tyr(c-Brd) genetic background. Dp(11)17/+ male mice displayed normal home-cage behavior but increased anxiety and increased dominant behavior in specific tests. A subtle impairment in the preference for a social target versus an inanimate target and abnormal preference for social novelty (the preference to explore an unfamiliar mouse versus a familiar one) was also observed. Our results indicate that these animals could provide a valuable model to identify the specific gene(s) that confer abnormal social behaviors and that map within this delimited genomic deletion interval. In a first attempt to identify candidate genes and for elucidating the mechanisms of regulation of these important phenotypes, we directly assessed the relative transcription of genes within and around this genomic interval. In this mouse model, we found that candidates genes include not only most of the duplicated genes, but also normal-copy genes that flank the engineered interval; both categories of genes showed altered expression levels in the hippocampus of Dp(11)17/+ mice.


Asunto(s)
Trastorno Autístico/fisiopatología , Modelos Animales de Enfermedad , Expresión Génica , Animales , Trastorno Autístico/genética , Conducta Animal , Encéfalo/crecimiento & desarrollo , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Tamaño de los Órganos , Fenotipo , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA