Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Am J Hum Genet ; 111(3): 614-617, 2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38330941

RESUMEN

Age-related hearing loss (ARHL) is a major health concern among the elderly population. It is hoped that increasing our understanding of its underlying pathophysiological processes will lead to the development of novel therapies. Recent genome-wide association studies (GWASs) discovered a few dozen genetic variants in association with elevated risk for ARHL. Integrated analysis of GWAS results and transcriptomics data is a powerful approach for elucidating specific cell types that are involved in disease pathogenesis. Intriguingly, recent studies that applied such bioinformatics approaches to ARHL resulted in disagreeing findings as for the key cell types that are most strongly linked to the genetic pathogenesis of ARHL. These conflicting studies pointed either to cochlear sensory epithelial or to stria vascularis cells as the cell types most prominently involved in the genetic basis of ARHL. Seeking to resolve this discrepancy, we integrated the analysis of four ARHL GWAS datasets with four independent inner-ear single-cell RNA-sequencing datasets. Our analysis clearly points to the cochlear sensory epithelial cells as the key cells for the genetic predisposition to ARHL. We also explain the limitation of the bioinformatics analysis performed by previous studies that led to missing the enrichment for ARHL GWAS signal in sensory epithelial cells. Collectively, we show that cochlear epithelial cells, not stria vascularis cells, are the main inner-ear cells related to the genetic pathogenesis of ARHL.


Asunto(s)
Presbiacusia , Estría Vascular , Anciano , Humanos , Estría Vascular/patología , Estudio de Asociación del Genoma Completo , Cóclea/patología , Presbiacusia/genética , Presbiacusia/patología , Epitelio/patología
2.
Cell Rep ; 42(11): 113421, 2023 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-37952154

RESUMEN

We explore the changes in chromatin accessibility and transcriptional programs for cochlear hair cell differentiation from postmitotic supporting cells using organoids from postnatal cochlea. The organoids contain cells with transcriptional signatures of differentiating vestibular and cochlear hair cells. Construction of trajectories identifies Lgr5+ cells as progenitors for hair cells, and the genomic data reveal gene regulatory networks leading to hair cells. We validate these networks, demonstrating dynamic changes both in expression and predicted binding sites of transcription factors (TFs) during organoid differentiation. We identify known regulators of hair cell development, Atoh1, Pou4f3, and Gfi1, and the analysis predicts the regulatory factors Tcf4, an E-protein and heterodimerization partner of Atoh1, and Ddit3, a CCAAT/enhancer-binding protein (C/EBP) that represses Hes1 and activates transcription of Wnt-signaling-related genes. Deciphering the signals for hair cell regeneration from mammalian cochlear supporting cells reveals candidates for hair cell (HC) regeneration, which is limited in the adult.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Cóclea , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Diferenciación Celular/genética , Organoides/metabolismo , Mamíferos/metabolismo
3.
iScience ; 26(10): 107769, 2023 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-37720106

RESUMEN

The cochlea consists of diverse cellular populations working in harmony to convert mechanical stimuli into electrical signals for the perception of sound. Otic mesenchyme cells (OMCs), often considered a homogeneous cell type, are essential for normal cochlear development and hearing. Despite being the most numerous cell type in the developing cochlea, OMCs are poorly understood. OMCs are known to differentiate into spatially and functionally distinct cell types, including fibrocytes of the lateral wall and spiral limbus, modiolar osteoblasts, and specialized tympanic border cells of the basilar membrane. Here, we show that OMCs are transcriptionally and functionally heterogeneous and can be divided into four distinct populations that spatially correspond to OMC-derived cochlear structures. We also show that this heterogeneity and complexity of OMCs commences during early phases of cochlear development. Finally, we describe the cell-cell communication network of the developing cochlea, inferring a major role for OMC in outgoing signaling.

4.
Otol Neurotol ; 44(9): 956-963, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37641232

RESUMEN

HYPOTHESIS: Metformin treatment will protect mice from noise-induced hearing loss (NIHL). BACKGROUND: We recently identified metformin as the top-ranking, Food and Drug Administration-approved drug to counter inner ear molecular changes induced by permanent threshold shift-inducing noise. This study is designed to functionally test metformin as a potential otoprotective drug against NIHL. METHODS: Male and female B6CBAF1/J mice were obtained at 7 to 8 weeks of age. A cohort of the females underwent ovariectomy to simulate menopause and eliminate the effect of ovarian-derived estrogens. At 10 weeks of age, mice underwent a permanent threshold shift-inducing noise exposure (102.5 or 105 dB SPL, 8-16 kHz, 2 h). Auditory brainstem response (ABR) thresholds were obtained at baseline, 24 h after noise exposure, and 1 week after noise exposure. Mice were administered metformin (200 mg/kg/d) or a saline control in their drinking water after the baseline ABR and for the remainder of the study. After the 1-week ABR, mice were euthanized and cochlear tissue was analyzed. RESULTS: Metformin treatment reduced the 1-week ABR threshold shift at 16 kHz ( p < 0.01; d = 1.20) and 24 kHz ( p < 0.01; d = 1.15) as well as outer hair cell loss in the 32-45.5 kHz range ( p < 0.0001; d = 2.37) in male mice. In contrast, metformin treatment did not prevent hearing loss or outer hair cell loss in the intact or ovariectomized female mice. CONCLUSIONS: Metformin exhibits sex-dependent efficacy as a therapeutic for NIHL. These data compel continued investigation into metformin's protective effects and demonstrate the importance of evaluating the therapeutic efficacy of drugs in subjects of both sexes.


Asunto(s)
Sordera , Oído Interno , Pérdida Auditiva Provocada por Ruido , Metformina , Femenino , Masculino , Estados Unidos , Animales , Ratones , Pérdida Auditiva Provocada por Ruido/prevención & control , Cóclea , Metformina/farmacología , Metformina/uso terapéutico
5.
Int J Mol Sci ; 22(22)2021 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-34830090

RESUMEN

Recent studies have identified sex-differences in auditory physiology and in the susceptibility to noise-induced hearing loss (NIHL). We hypothesize that 17ß-estradiol (E2), a known modulator of auditory physiology, may underpin sex-differences in the response to noise trauma. Here, we gonadectomized B6CBAF1/J mice and used a combination of electrophysiological and histological techniques to study the effects of estrogen replacement on peripheral auditory physiology in the absence of noise exposure and on protection from NIHL. Functional analysis of auditory physiology in gonadectomized female mice revealed that E2-treatment modulated the peripheral response to sound in the absence of changes to the endocochlear potential compared to vehicle-treatment. E2-replacement in gonadectomized female mice protected against hearing loss following permanent threshold shift (PTS)- and temporary threshold shift (TTS)-inducing noise exposures. Histological analysis of the cochlear tissue revealed that E2-replacement mitigated outer hair cell loss and cochlear synaptopathy following noise exposure compared to vehicle-treatment. Lastly, using fluorescent in situ hybridization, we demonstrate co-localization of estrogen receptor-2 with type-1C, high threshold spiral ganglion neurons, suggesting that the observed protection from cochlear synaptopathy may occur through E2-mediated preservation of these neurons. Taken together, these data indicate the estrogen signaling pathways may be harnessed for the prevention and treatment of NIHL.


Asunto(s)
Cóclea , Estradiol/farmacología , Potenciales Evocados Auditivos/efectos de los fármacos , Pérdida Auditiva Provocada por Ruido , Animales , Cóclea/metabolismo , Cóclea/patología , Cóclea/fisiopatología , Femenino , Pérdida Auditiva Provocada por Ruido/metabolismo , Pérdida Auditiva Provocada por Ruido/patología , Pérdida Auditiva Provocada por Ruido/fisiopatología , Pérdida Auditiva Provocada por Ruido/prevención & control , Ratones , Ovariectomía
6.
PLoS Biol ; 19(11): e3001445, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34758021

RESUMEN

Cochlear supporting cells (SCs) are glia-like cells critical for hearing function. In the neonatal cochlea, the greater epithelial ridge (GER) is a mitotically quiescent and transient organ, which has been shown to nonmitotically regenerate SCs. Here, we ablated Lgr5+ SCs using Lgr5-DTR mice and found mitotic regeneration of SCs by GER cells in vivo. With lineage tracing, we show that the GER houses progenitor cells that robustly divide and migrate into the organ of Corti to replenish ablated SCs. Regenerated SCs display coordinated calcium transients, markers of the SC subtype inner phalangeal cells, and survive in the mature cochlea. Via RiboTag, RNA-sequencing, and gene clustering algorithms, we reveal 11 distinct gene clusters comprising markers of the quiescent and damaged GER, and damage-responsive genes driving cell migration and mitotic regeneration. Together, our study characterizes GER cells as mitotic progenitors with regenerative potential and unveils their quiescent and damaged translatomes.


Asunto(s)
Linaje de la Célula/genética , Cóclea/citología , Estudios de Asociación Genética , Mitosis , Biosíntesis de Proteínas , Regeneración/genética , Células Madre/citología , Células Madre/metabolismo , Animales , Diferenciación Celular , Supervivencia Celular , Células Epiteliales/citología , Regulación de la Expresión Génica , Integrasas/metabolismo , Ratones , Familia de Multigenes , Receptores Acoplados a Proteínas G/metabolismo
7.
Cell Rep ; 36(13): 109758, 2021 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-34592158

RESUMEN

Noise-induced hearing loss (NIHL) results from a complex interplay of damage to the sensory cells of the inner ear, dysfunction of its lateral wall, axonal retraction of type 1C spiral ganglion neurons, and activation of the immune response. We use RiboTag and single-cell RNA sequencing to survey the cell-type-specific molecular landscape of the mouse inner ear before and after noise trauma. We identify induction of the transcription factors STAT3 and IRF7 and immune-related genes across all cell-types. Yet, cell-type-specific transcriptomic changes dominate the response. The ATF3/ATF4 stress-response pathway is robustly induced in the type 1A noise-resilient neurons, potassium transport genes are downregulated in the lateral wall, mRNA metabolism genes are downregulated in outer hair cells, and deafness-associated genes are downregulated in most cell types. This transcriptomic resource is available via the Gene Expression Analysis Resource (gEAR; https://umgear.org/NIHL) and provides a blueprint for the rational development of drugs to prevent and treat NIHL.


Asunto(s)
Oído Interno/metabolismo , Células Ciliadas Auditivas/metabolismo , Pérdida Auditiva Provocada por Ruido/metabolismo , Pérdida Auditiva Provocada por Ruido/fisiopatología , Ganglio Espiral de la Cóclea/metabolismo , Animales , Cóclea/metabolismo , Cóclea/fisiopatología , Oído Interno/fisiopatología , Potenciales Evocados Auditivos del Tronco Encefálico/fisiología , Pérdida Auditiva Provocada por Ruido/genética , Ratones , Neuronas/metabolismo , Ruido , Ganglio Espiral de la Cóclea/citología , Ganglio Espiral de la Cóclea/fisiopatología
9.
Laryngoscope ; 131 Suppl 5: S1-S16, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-32579737

RESUMEN

OBJECTIVE: The cellular diversity of the inner ear has presented a technical challenge in obtaining molecular insight into its development and function. The application of technological advancements in cell type-specific expression enable clinicians and researchers to leap forward from classic genetics to obtaining mechanistic understanding of congenital and acquired hearing loss. This understanding is essential for development of therapeutics to prevent and reverse diseases of the inner ear, including hearing loss. The objective of this study is to describe and compare the available tools for cell type-specific analysis of the ear, as a means to support decision making in study design. STUDY DESIGN: Three major approaches for cell type-specific analysis of the ear including fluorescence-activated cell sorting (FACS), ribosomal and RNA pulldown techniques, and single cell RNA-seq (scRNA-seq) are compared and contrasted using both published and original data. RESULTS: We demonstrate the strength and weaknesses of these approaches leading to the inevitable conclusion that to maximize the utility of these approaches, it is important to match the experimental approach with the tissue of origin, cell type of interest, and the biological question. Often, a combined approach (eg, cell sorting and scRNA-seq or expression analysis using 2 separate approaches) is required. Finally, new tools for visualization and analysis of complex expression data, such as the gEAR platform (umgear.org), collate cell type-specific gene expression from the ear field and provide unprecedented access to both clinicians and researchers. LEVEL OF EVIDENCE: N/A Laryngoscope, 131:S1-S16, 2021.


Asunto(s)
Oído Interno/citología , Citometría de Flujo/métodos , Perfilación de la Expresión Génica , ARN/aislamiento & purificación , Análisis de Secuencia de ARN/métodos , Animales , Toma de Decisiones , Colorantes Fluorescentes , Expresión Génica , Pérdida Auditiva/congénito , Pérdida Auditiva/genética , Humanos , Ratones , Ratones Transgénicos , Órgano Espiral/citología , Compuestos de Piridinio , Compuestos de Amonio Cuaternario , Ribosomas/metabolismo , Análisis de la Célula Individual/métodos , Uniones Estrechas
10.
PLoS Genet ; 16(9): e1009025, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32986727

RESUMEN

Age-related hearing impairment (ARHI), one of the most common medical conditions, is strongly heritable, yet its genetic causes remain largely unknown. We conducted a meta-analysis of GWAS summary statistics from multiple hearing-related traits in the UK Biobank (n = up to 330,759) and identified 31 genome-wide significant risk loci for self-reported hearing difficulty (p < 5x10-8), of which eight have not been reported previously in the peer-reviewed literature. We investigated the regulatory and cell specific expression for these loci by generating mRNA-seq, ATAC-seq, and single-cell RNA-seq from cells in the mouse cochlea. Risk-associated genes were most strongly enriched for expression in cochlear epithelial cells, as well as for genes related to sensory perception and known Mendelian deafness genes, supporting their relevance to auditory function. Regions of the human genome homologous to open chromatin in epithelial cells from the mouse were strongly enriched for heritable risk for hearing difficulty, even after adjusting for baseline effects of evolutionary conservation and cell-type non-specific regulatory regions. Epigenomic and statistical fine-mapping most strongly supported 50 putative risk genes. Of these, 39 were expressed robustly in mouse cochlea and 16 were enriched specifically in sensory hair cells. These results reveal new risk loci and risk genes for hearing difficulty and suggest an important role for altered gene regulation in the cochlear sensory epithelium.


Asunto(s)
Cóclea/citología , Sitios Genéticos , Predisposición Genética a la Enfermedad , Pérdida Auditiva/genética , Adulto , Animales , Bancos de Muestras Biológicas , Cromatina/genética , Estudios de Cohortes , Epigenoma , Células Epiteliales/fisiología , Femenino , Estudio de Asociación del Genoma Completo , Células Ciliadas Auditivas/citología , Células Ciliadas Auditivas/fisiología , Humanos , Ratones Endogámicos ICR , Ratones Endogámicos , Polimorfismo de Nucleótido Simple , Análisis de la Célula Individual , Reino Unido
11.
Development ; 147(17)2020 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-32917668

RESUMEN

Despite the known importance of the transcription factors ATOH1, POU4F3 and GFI1 in hair cell development and regeneration, their downstream transcriptional cascades in the inner ear remain largely unknown. Here, we have used Gfi1cre;RiboTag mice to evaluate changes to the hair cell translatome in the absence of GFI1. We identify a systematic downregulation of hair cell differentiation genes, concomitant with robust upregulation of neuronal genes in the GFI1-deficient hair cells. This includes increased expression of neuronal-associated transcription factors (e.g. Pou4f1) as well as transcription factors that serve dual roles in hair cell and neuronal development (e.g. Neurod1, Atoh1 and Insm1). We further show that the upregulated genes are consistent with the NEUROD1 regulon and are normally expressed in hair cells prior to GFI1 onset. Additionally, minimal overlap of differentially expressed genes in auditory and vestibular hair cells suggests that GFI1 serves different roles in these systems. From these data, we propose a dual mechanism for GFI1 in promoting hair cell development, consisting of repression of neuronal-associated genes as well as activation of hair cell-specific genes required for normal functional maturation.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Células Ciliadas Auditivas Internas/metabolismo , Factores de Transcripción/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Unión al ADN/genética , Células Ciliadas Auditivas Internas/citología , Ratones , Ratones Transgénicos , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factor de Transcripción Brn-3A/genética , Factor de Transcripción Brn-3A/metabolismo , Factores de Transcripción/genética
12.
Nature ; 563(7733): 696-700, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30464345

RESUMEN

The sensory cells that are responsible for hearing include the cochlear inner hair cells (IHCs) and outer hair cells (OHCs), with the OHCs being necessary for sound sensitivity and tuning1. Both cell types are thought to arise from common progenitors; however, our understanding of the factors that control the fate of IHCs and OHCs remains limited. Here we identify Ikzf2 (which encodes Helios) as an essential transcription factor in mice that is required for OHC functional maturation and hearing. Helios is expressed in postnatal mouse OHCs, and in the cello mouse model a point mutation in Ikzf2 causes early-onset sensorineural hearing loss. Ikzf2cello/cello OHCs have greatly reduced prestin-dependent electromotile activity, a hallmark of OHC functional maturation, and show reduced levels of crucial OHC-expressed genes such as Slc26a5 (which encodes prestin) and Ocm. Moreover, we show that ectopic expression of Ikzf2 in IHCs: induces the expression of OHC-specific genes; reduces the expression of canonical IHC genes; and confers electromotility to IHCs, demonstrating that Ikzf2 can partially shift the IHC transcriptome towards an OHC-like identity.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Células Ciliadas Auditivas Externas/citología , Células Ciliadas Auditivas Externas/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Transcriptoma/genética , Animales , Secuencia de Bases , Biomarcadores/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL
13.
BMC Genomics ; 19(1): 696, 2018 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-30241496

RESUMEN

BACKGROUND: Cell type-specific ribosome-pulldown has become an increasingly popular method for analysis of gene expression. It allows for expression analysis from intact tissues and monitoring of protein synthesis in vivo. However, while its utility has been assessed, technical aspects related to sequencing of these samples, often starting with a smaller amount of RNA, have not been reported. In this study, we evaluated the performance of five library prep protocols for ribosome-associated mRNAs when only 250 pg-4 ng of total RNA are used. RESULTS: We obtained total and RiboTag-IP RNA, in three biological replicates. We compared 5 methods of library preparation for Illumina Next Generation sequencing: NuGEN Ovation RNA-Seq system V2 Kit, TaKaRa SMARTer Stranded Total RNA-Seq Kit, TaKaRa SMART-Seq v4 Ultra Low Input RNA Kit, Illumina TruSeq RNA Library Prep Kit v2 and NEBNext® Ultra™ Directional RNA Library Prep Kit using slightly modified protocols each with 4 ng of total RNA. An additional set of samples was processed using the TruSeq kit with 70 ng, as a 'gold standard' control and the SMART-Seq v4 with 250 pg of total RNA. TruSeq-processed samples had the best metrics overall, with similar results for the 4 ng and 70 ng samples. The results of the SMART-Seq v4 processed samples were similar to TruSeq (Spearman correlation > 0.8) despite using lower amount of input RNA. All RiboTag-IP samples had an increase in the intronic reads compared with the corresponding whole tissue, suggesting that the IP captures some immature mRNAs. The SMARTer-processed samples had a higher representation of ribosomal and non-coding RNAs leading to lower representation of protein coding mRNA. The enrichment or depletion of IP samples compared to corresponding input RNA was similar across all kits except for SMARTer kit. CONCLUSION: RiboTag-seq can be performed successfully with as little as 250 pg of total RNA when using the SMART-Seq v4 kit and 4 ng when using the modified protocols of other library preparation kits. The SMART-Seq v4 and TruSeq kits resulted in the highest quality libraries. RiboTag IP RNA contains some immature transcripts.


Asunto(s)
Biblioteca de Genes , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Biosíntesis de Proteínas , ARN Mensajero/metabolismo , Ribosomas/metabolismo , Análisis de Secuencia de ARN/veterinaria , Transcriptoma , Animales , Inmunoprecipitación , Ratones , Control de Calidad , ARN Mensajero/genética , ARN Mensajero/aislamiento & purificación , Ribosomas/genética
14.
Biol Sex Differ ; 9(1): 12, 2018 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-29530094

RESUMEN

BACKGROUND: Noise-induced hearing loss (NIHL) is the most prevalent form of acquired hearing loss and affects about 40 million US adults. Among the suggested therapeutics tested in rodents, suberoylanilide hydroxamic acid (SAHA) has been shown to be otoprotective from NIHL; however, these results were limited to male mice. METHODS: Here we tested the effect of SAHA on the hearing of 10-week-old B6CBAF1/J mice of both sexes, which were exposed to 2 h of octave-band noise (101 dB SPL centered at 11.3 kHz). Hearing was assessed by measuring auditory brainstem responses (ABR) at 8, 16, 24, and 32 kHz, 1 week before, as well as at 24 h and 15-21 days following exposure (baseline, compound threshold shift (CTS) and permanent threshold shift (PTS), respectively), followed by histologic analyses. RESULTS: We found significant differences in the CTS and PTS of the control (vehicle injected) mice to noise, where females had a significantly smaller CTS at 16 and 24 kHz (p < 0.0001) and PTS at 16, 24, and 32 kHz (16 and 24 kHz p < 0.001, 32 kHz p < 0.01). This sexual dimorphic effect could not be explained by a differential loss of sensory cells or synapses but was reflected in the amplitude and amplitude progression of wave I of the ABR, which correlates with outer hair cell (OHC) function. Finally, the frequency of the protective effect of SAHA differed significantly between males (PTS, 24 kHz, p = 0.002) and females (PTS, 16 kHz, p = 0.003), and the magnitude of the protection was smaller in females than in males. Importantly, the magnitude of the protection by SAHA was smaller than the effect of sex as a biological factor in the vehicle-injected mice. CONCLUSIONS: These results indicate that female mice are significantly protected from NIHL in comparison to males and that therapeutics for NIHL may have a different effect in males and females. The data highlight the importance of analyzing NIHL experiments from males and females, separately. Finally, these data also raise the possibility of effectors in the estrogen signaling pathway as novel therapeutics for NIHL.


Asunto(s)
Pérdida Auditiva Provocada por Ruido/prevención & control , Ruido/efectos adversos , Sustancias Protectoras/farmacología , Caracteres Sexuales , Vorinostat/farmacología , Animales , Potenciales Evocados Auditivos del Tronco Encefálico , Femenino , Masculino , Ratones
15.
Sci Rep ; 7: 42079, 2017 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-28181545

RESUMEN

Studies of developmental and functional biology largely rely on conditional expression of genes in a cell type-specific manner. Therefore, the importance of specificity and lack of inherent phenotypes for Cre-driver animals cannot be overemphasized. The Gfi1Cre mouse is commonly used for conditional hair cell-specific gene deletion/reporter gene activation in the inner ear. Here, using immunofluorescence and flow cytometry, we show that the Gfi1Cre mice produce a pattern of recombination that is not strictly limited to hair cells within the inner ear. We observe a broad expression of Cre recombinase in the Gfi1Cre mouse neonatal inner ear, primarily in inner ear resident macrophages, which outnumber the hair cells. We further show that heterozygous Gfi1Cre mice exhibit an early onset progressive hearing loss as compared with their wild-type littermates. Importantly, vestibular function remains intact in heterozygotes up to 10 months, the latest time point tested. Finally, we detect minor, but statistically significant, changes in expression of hair cell-enriched transcripts in the Gfi1Cre heterozygous mice cochleae compared with their wild-type littermate controls. Given the broad use of the Gfi1Cre mice, both for gene deletion and reporter gene activation, these data are significant and necessary for proper planning and interpretation of experiments.


Asunto(s)
Proteínas de Unión al ADN/genética , Oído Interno/patología , Pérdida Auditiva/genética , Pérdida Auditiva/patología , Integrasas/genética , Recombinación Genética , Factores de Transcripción/genética , Animales , Proteínas de Unión al ADN/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Integrasas/metabolismo , Ratones , Factores de Transcripción/metabolismo
16.
Nat Commun ; 6: 8549, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26469318

RESUMEN

Sensorineural hearing loss is a common and currently irreversible disorder, because mammalian hair cells (HCs) do not regenerate and current stem cell and gene delivery protocols result only in immature HC-like cells. Importantly, although the transcriptional regulators of embryonic HC development have been described, little is known about the postnatal regulators of maturating HCs. Here we apply a cell type-specific functional genomic analysis to the transcriptomes of auditory and vestibular sensory epithelia from early postnatal mice. We identify RFX transcription factors as essential and evolutionarily conserved regulators of the HC-specific transcriptomes, and detect Rfx1,2,3,5 and 7 in the developing HCs. To understand the role of RFX in hearing, we generate Rfx1/3 conditional knockout mice. We show that these mice are deaf secondary to rapid loss of initially well-formed outer HCs. These data identify an essential role for RFX in hearing and survival of the terminally differentiating outer HCs.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Células Ciliadas Auditivas/metabolismo , Audición/fisiología , Factores de Transcripción/metabolismo , Animales , Animales Recién Nacidos , Evolución Biológica , Inmunoprecipitación de Cromatina , Femenino , Regulación de la Expresión Génica , Células Ciliadas Auditivas/ultraestructura , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Familia de Multigenes , Factores de Transcripción del Factor Regulador X , Factor Regulador X1 , Análisis de Secuencia de ADN , Transcriptoma , Pez Cebra
17.
BMC Genomics ; 15: 988, 2014 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-25407537

RESUMEN

BACKGROUND: Chromatin compactness has been considered a major determinant of gene activity and has been associated with specific chromatin modifications in studies on a few individual genetic loci. At the same time, genome-wide patterns of open and closed chromatin have been understudied, and are at present largely predicted from chromatin modification and gene expression data. However the universal applicability of such predictions is not self-evident, and requires experimental verification. RESULTS: We developed and implemented a high-throughput analysis for general chromatin sensitivity to DNase I which provides a comprehensive epigenomic assessment in a single assay. Contiguous domains of open and closed chromatin were identified by computational analysis of the data, and correlated to other genome annotations including predicted chromatin "states", individual chromatin modifications, nuclear lamina interactions, and gene expression. While showing that the widely trusted predictions of chromatin structure are correct in the majority of cases, we detected diverse "exceptions" from the conventional rules. We found a profound paucity of chromatin modifications in a major fraction of closed chromatin, and identified a number of loci where chromatin configuration is opposite to that expected from modification and gene expression patterns. Further, we observed that chromatin of large introns tends to be closed even when the genes are expressed, and that a significant proportion of active genes including their promoters are located in closed chromatin. CONCLUSIONS: These findings reveal limitations of the existing predictive models, indicate novel mechanisms of epigenetic regulation, and provide important insights into genome organization and function.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , Cromatina/genética , Mapeo Cromosómico , Drosophila/genética , Genoma de los Insectos , Animales , Sitios de Unión , Cromatina/metabolismo , Biología Computacional/métodos , Desoxirribonucleasa I/metabolismo , Unión Proteica
18.
PLoS One ; 7(11): e49692, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23226217

RESUMEN

Theoretical models suggest that gene silencing at the nuclear periphery may involve "closing" of chromatin by transcriptional repressors, such as histone deacetylases (HDACs). Here we provide experimental evidence confirming these predictions. Histone acetylation, chromatin compactness, and gene repression in lamina-interacting multigenic chromatin domains were analyzed in Drosophila S2 cells in which B-type lamin, diverse HDACs, and lamina-associated proteins were downregulated by dsRNA. Lamin depletion resulted in decreased compactness of the repressed multigenic domain associated with its detachment from the lamina and enhanced histone acetylation. Our data reveal the major role for HDAC1 in mediating deacetylation, chromatin compaction, and gene silencing in the multigenic domain, and an auxiliary role for HDAC3 that is required for retention of the domain at the lamina. These findings demonstrate the manifold and central involvement of class I HDACs in regulation of lamina-associated genes, illuminating a mechanism by which these enzymes can orchestrate normal and pathological development.


Asunto(s)
Cromatina/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Regulación de la Expresión Génica , Histona Desacetilasa 1/genética , Histona Desacetilasas/genética , Histonas/genética , Lámina Nuclear/genética , Acetilación , Animales , Western Blotting , Línea Celular , Cromatina/enzimología , Inmunoprecipitación de Cromatina , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/enzimología , Silenciador del Gen , Histona Desacetilasa 1/metabolismo , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Lamina Tipo B/antagonistas & inhibidores , Lamina Tipo B/genética , Lamina Tipo B/metabolismo , Familia de Multigenes , Lámina Nuclear/enzimología , ARN Bicatenario/genética , Transcripción Genética
19.
Prostate ; 71(14): 1518-24, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21360563

RESUMEN

BACKGROUND: A marked decrease in the level of zinc is a consistent characteristic of prostate cancer; which results from down-regulation of ZIP1 zinc transporter. The aim of this study was to determine if RREB-1 transcription is involved in the down-regulation of ZIP1 gene expression; and to determine the expression of RREB-1 in benign and cancerous prostate in situ. METHODS: Overexpression and siRNA knock down of RREB-1 were used to determine the effect of RREB-1 on hZIP1 abundance in PC-3 cells. Immunohistochemistry with tissue microarrays (TMAs) and tissue sections was used to determine the levels of RREB-1 expression in prostate in situ. RESULTS: Overexpression of RREB-1 resulted in a decrease in the abundance of hZIP1 in the plasma membrane of PC-3 cells; whereas siRNA knock down significantly increased hZIP1 expression. Prostate TMAs and tissue sections showed an inverse relationship between RREB-1 and hZIP1 staining. CONCLUSIONS: RREB-1 overexpression results in down-regulation of hZIP1 and contributes to the loss of hZIP1 expression and zinc in prostate cancer. This is an early event in prostate carcinogenesis.


Asunto(s)
Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Proteínas de Transporte de Catión/metabolismo , Proteínas de Unión al ADN/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Factores de Transcripción/genética , Adenocarcinoma/patología , Adulto , Anciano , Biopsia , Proteínas de Transporte de Catión/genética , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo/fisiología , Epigénesis Genética/fisiología , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Neoplasias de la Próstata/patología , ARN Interferente Pequeño/genética , Factores de Transcripción/metabolismo , Zinc/metabolismo
20.
Prostate ; 70(3): 288-96, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19802870

RESUMEN

BACKGROUND: Normal prostate accumulates extremely high levels of zinc compared to other soft tissues. In contrast, the level of zinc in the prostate decreases significantly in prostate cancer. We have shown that down-regulation of the expression of the zinc transporter hZIP1 in prostate cancer is an important event that is responsible for the decrease of zinc levels. However, the mechanism of hZIP1 down-regulation is not known. We have hypothesized that hZIP1 is down-regulated through transcriptional regulation. METHODS: The hZIP1 promoter was studied using luciferase reporter assays, site-directed mutagenesis, gel shift, and ChIP assay. RESULTS: We have characterized a promoter region, downstream of the transcription start site, responsible for repression of hZIP1 transcription. We demonstrate that this region contains a binding site for the Ras-Responsive Element Binding protein 1 (RREB-1) and that the binding of RREB-1 to the hZIP1 promoter is involved in the decrease of hZIP1 transcription in PC-3 cells. CONCLUSION: The Ras pathway and activation of RREB-1 are involved in hZIP1 down-regulation and may play a role in the decrease of the transporter expression in prostate cancer.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Proteínas de Unión al ADN/metabolismo , Neoplasias de la Próstata/metabolismo , Factores de Transcripción/metabolismo , Secuencia de Bases , Sitios de Unión/genética , Proteínas de Transporte de Catión/genética , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Regulación hacia Abajo , Humanos , Luciferasas/metabolismo , Masculino , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , ARN Interferente Pequeño/farmacología , Factores de Transcripción/genética , Sitio de Iniciación de la Transcripción , Transcripción Genética , Proteínas ras/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...