Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(1): e2213099120, 2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36577057

RESUMEN

The cochlea's ability to discriminate sound frequencies is facilitated by a special topography along its longitudinal axis known as tonotopy. Auditory hair cells located at the base of the cochlea respond to high-frequency sounds, whereas hair cells at the apex respond to lower frequencies. Gradual changes in morphological and physiological features along the length of the cochlea determine each region's frequency selectivity, but it remains unclear how tonotopy is established during cochlear development. Recently, sonic hedgehog (SHH) was proposed to initiate the establishment of tonotopy by conferring regional identity to the primordial cochlea. Here, using mouse genetics, we provide in vivo evidence that regional identity in the embryonic cochlea acts as a framework upon which tonotopy-specific properties essential for frequency selectivity in the mature cochlea develop. We found that follistatin (FST) is required for the maintenance of apical cochlear identity, but dispensable for its initial induction. In a fate-mapping analysis, we found that FST promotes expansion of apical cochlear cells, contributing to the formation of the apical cochlear domain. SHH, in contrast, is required both for the induction and maintenance of apical identity. In the absence of FST or SHH, mice produce a short cochlea lacking its apical domain. This results in the loss of apex-specific anatomical and molecular properties and low-frequency-specific hearing loss.


Asunto(s)
Folistatina , Proteínas Hedgehog , Animales , Ratones , Folistatina/genética , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Cóclea/fisiología , Audición/fisiología , Mamíferos/metabolismo
2.
Proc Natl Acad Sci U S A ; 117(20): 11109-11117, 2020 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-32358189

RESUMEN

Outer hair cells (OHCs) play an essential role in hearing by acting as a nonlinear amplifier which helps the cochlea detect sounds with high sensitivity and accuracy. This nonlinear sound processing generates distortion products, which can be measured as distortion-product otoacoustic emissions (DPOAEs). The OHC stereocilia that respond to sound vibrations are connected by three kinds of extracellular links: tip links that connect the taller stereocilia to shorter ones and convey force to the mechanoelectrical transduction channels, tectorial membrane-attachment crowns (TM-ACs) that connect the tallest stereocilia to one another and to the overlying TM, and horizontal top connectors (HTCs) that link adjacent stereocilia. While the tip links have been extensively studied, the roles that the other two types of links play in hearing are much less clear, largely because of a lack of suitable animal models. Here, while analyzing genetic combinations of tubby mice, we encountered models missing both HTCs and TM-ACs or HTCs alone. We found that the tubby mutation causes loss of both HTCs and TM-ACs due to a mislocalization of stereocilin, which results in OHC dysfunction leading to severe hearing loss. Intriguingly, the addition of the modifier allele modifier of tubby hearing 1 in tubby mice selectively rescues the TM-ACs but not the HTCs. Hearing is significantly rescued in these mice with robust DPOAE production, indicating an essential role of the TM-ACs but not the HTCs in normal OHC function. In contrast, the HTCs are required for the resistance of hearing to damage caused by noise stress.


Asunto(s)
Células Ciliadas Auditivas Externas/fisiología , Ruido , Emisiones Otoacústicas Espontáneas/fisiología , Sonido , Estimulación Acústica , Animales , Células Ciliadas Auditivas Externas/citología , Pérdida Auditiva , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/genética , Modelos Animales , Emisiones Otoacústicas Espontáneas/genética , Estereocilios/fisiología , Membrana Tectoria
3.
Development ; 146(2)2019 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-30630826

RESUMEN

Defects in the middle ear ossicles - malleus, incus and stapes - can lead to conductive hearing loss. During development, neural crest cells (NCCs) migrate from the dorsal hindbrain to specific locations in pharyngeal arch (PA) 1 and 2, to form the malleus-incus and stapes, respectively. It is unclear how migratory NCCs reach their proper destination in the PA and initiate mesenchymal condensation to form specific ossicles. We show that secreted molecules sonic hedgehog (SHH) and bone morphogenetic protein 4 (BMP4) emanating from the pharyngeal endoderm are important in instructing region-specific NCC condensation to form malleus-incus and stapes, respectively, in mouse. Tissue-specific knockout of Shh in the pharyngeal endoderm or Smo (a transducer of SHH signaling) in NCCs causes the loss of malleus-incus condensation in PA1 but only affects the maintenance of stapes condensation in PA2. By contrast, knockout of Bmp4 in the pharyngeal endoderm or Smad4 (a transducer of TGFß/BMP signaling) in the NCCs disrupts NCC migration into the stapes region in PA2, affecting stapes formation. These results indicate that region-specific endodermal signals direct formation of specific middle ear ossicles.


Asunto(s)
Osículos del Oído/embriología , Endodermo/embriología , Endodermo/metabolismo , Cresta Neural/citología , Transducción de Señal , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Movimiento Celular , Supervivencia Celular , Eliminación de Gen , Proteínas Hedgehog , Yunque/embriología , Yunque/metabolismo , Martillo/embriología , Martillo/metabolismo , Ratones , Modelos Biológicos , Cresta Neural/embriología , Cresta Neural/metabolismo , Especificidad de Órganos , Faringe/embriología , Fenotipo , Estribo/embriología , Estribo/metabolismo , Factores de Tiempo , Factor de Crecimiento Transformador beta/metabolismo
4.
IUBMB Life ; 68(6): 436-44, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27080371

RESUMEN

Modulation of chromatin structure has been proposed as a molecular mechanism underlying the spatiotemporal collinear expression of Hox genes during development. CCCTC-binding factor (CTCF)-mediated chromatin organization is now recognized as a crucial epigenetic mechanism for transcriptional regulation. Thus, we examined whether CTCF-mediated chromosomal conformation is involved in Hoxc gene expression by comparing wild-type mouse embryonic fibroblast (MEF) cells expressing anterior Hoxc genes with Akt1 null MEFs expressing anterior as well as posterior Hoxc genes. We found that CTCF binding between Hoxc11 and -c12 is important for CTCF-mediated chromosomal loop formation and concomitant posterior Hoxc gene expression. Hypomethylation at this site increased CTCF binding and recapitulated the chromosomal conformation and posterior Hoxc gene expression patterns observed in Akt1 null MEFs. From this work we found that CTCF at the C12|11 does not function as a barrier/boundary, instead let the posterior Hoxc genes switch their interaction from inactive centromeric to active telomeric genomic niche, and concomitant posterior Hoxc gene expression. Although it is not clear whether CTCF affects Hoxc gene expression solely through its looping activity, CTCF-mediated chromatin structural modulation could be an another tier of Hox gene regulation during development. © 2016 IUBMB Life, 68(6):436-444, 2016.


Asunto(s)
Cromatina/genética , Proteínas de Homeodominio/genética , Proteínas Represoras/metabolismo , Animales , Factor de Unión a CCCTC , Células Cultivadas , Cromatina/metabolismo , ADN/química , ADN/metabolismo , Metilación de ADN , Femenino , Fibroblastos/fisiología , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Masculino , Ratones Endogámicos C57BL , Familia de Multigenes , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Represoras/genética
5.
Mol Brain ; 15(1): 80, 2022 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-36104704

RESUMEN

Tubby mice exhibit hearing impairment due to the loss of stereocilin from the tip regions that connect the tallest stereocilia of the outer hair cells (OHCs) to the tectorial membrane. Stereocilin is an essential stereociliary protein in the OHCs, the mutation of which in humans causes autosomal recessive non-syndromic deafness. Map1a is a modifier of tubby hearing (moth1), and its wild-type allele, rather than the moth1 allele from the C57BL/6 J strain, restores stereocilin localization to the stereocilia and rescues the hearing impairment of tubby mice. The mechanism by which MAP1A accomplishes this is unclear, partly due to ambiguity regarding whether the tubby mutation is a true null. We therefore generated Tub-null (Tub-/-) mice by deleting exon 3 and found that they exhibit hearing impairment like that of tubby mice, suggesting the tubby mutation is a loss-of-function mutation with regard to hearing. When we crossed Tub-/- mice with AKR mice that have wild-type Map1a alleles, we found that wild-type MAP1A restores stereocilin localization to the tips of stereocilia and rescues hearing impairment. These data suggest MAP1A does not require interaction with tubby protein in maintaining stereocilin at the tips of stereocilia and that OHCs use two independent molecules-MAP1A and tubby-to doubly ensure proper stereocilin localization.


Asunto(s)
Pérdida Auditiva , Estereocilios , Proteínas Adaptadoras Transductoras de Señales , Animales , Células Ciliadas Auditivas Internas , Pérdida Auditiva/genética , Pérdida Auditiva/metabolismo , Pérdida Auditiva Sensorineural , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/metabolismo , Proteína Estafilocócica A/metabolismo , Estereocilios/metabolismo
6.
EBioMedicine ; 82: 104184, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35870427

RESUMEN

BACKGROUND: Cryopyrin-associated periodic syndrome (CAPS) is an inherited autoinflammatory disease caused by a gain-of-function mutation in NLRP3. Although CAPS patients frequently suffer from sensorineural hearing loss, it remains unclear whether CAPS-associated mutation in NLRP3 is associated with the progression of hearing loss. METHODS: We generated a mice with conditional expression of CAPS-associated NLRP3 mutant (D301N) in cochlea-resident CX3CR1 macrophages and examined the susceptibility of CAPS mice to inflammation-mediated hearing loss in a local and systemic inflammation context. FINDINGS: Upon lipopolysaccharide (LPS) injection into middle ear cavity, NLRP3 mutant mice exhibited severe cochlear inflammation, inflammasome activation and hearing loss. However, this middle ear injection model induced a considerable hearing loss in control mice and inevitably caused an inflammation-independent hearing loss possibly due to ear tissue damages by injection procedure. Subsequently, we optimized a systemic LPS injection model, which induced a significant hearing loss in NLRP3 mutant mice but not in control mice. Peripheral inflammation induced by a repetitive low dose of LPS injection caused a blood-labyrinth barrier disruption, macrophage infiltration into cochlea and cochlear inflammasome activation in an NLRP3-dependent manner. Interestingly, both cochlea-infiltrating and -resident macrophages contribute to peripheral inflammation-mediated hearing loss of CAPS mice. Furthermore, NLRP3-specific inhibitor, MCC950, as well as an interleukin-1 receptor antagonist significantly alleviated systemic LPS-induced hearing loss and inflammatory phenotypes in NLRP3 mutant mice. INTERPRETATION: Our findings reveal that CAPS-associated NLRP3 mutation is critical for peripheral inflammation-induced hearing loss in our CAPS mice model, and an NLRP3-specific inhibitor can be used to treat inflammation-mediated sensorineural hearing loss. FUNDING: National Research Foundation of Korea Grant funded by the Korean Government and the Team Science Award of Yonsei University College of Medicine.


Asunto(s)
Síndromes Periódicos Asociados a Criopirina , Sordera , Pérdida Auditiva Sensorineural , Pérdida Auditiva , Animales , Síndromes Periódicos Asociados a Criopirina/etiología , Síndromes Periódicos Asociados a Criopirina/genética , Modelos Animales de Enfermedad , Pérdida Auditiva/etiología , Pérdida Auditiva/genética , Pérdida Auditiva Sensorineural/etiología , Pérdida Auditiva Sensorineural/genética , Humanos , Inflamasomas/metabolismo , Inflamación/metabolismo , Lipopolisacáridos/toxicidad , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/genética
7.
Biochem Biophys Res Commun ; 392(4): 543-7, 2010 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-20097160

RESUMEN

Hoxc8 is a member of Hox family transcription factors that play crucial roles in spatiotemporal body patterning during embryogenesis. Hox proteins contain a conserved 61 amino acid homeodomain, which is responsible for recognition and binding of the proteins onto Hox-specific DNA binding motifs and regulates expression of their target genes. Previously, using proteome analysis, we identified Proliferating cell nuclear antigen (Pcna) as one of the putative target genes of Hoxc8. Here, we asked whether Hoxc8 regulates Pcna expression by directly binding to the regulatory sequence of Pcna. In mouse embryos at embryonic day 11.5, the expression pattern of Pcna was similar to that of Hoxc8 along the anteroposterior body axis. Moreover, Pcna transcript levels as well as cell proliferation rate were increased by overexpression of Hoxc8 in C3H10T1/2 mouse embryonic fibroblast cells. Characterization of 2.3kb genomic sequence upstream of Pcna coding region revealed that the upstream sequence contains several Hox core binding sequences and one Hox-Pbx binding sequence. Direct binding of Hoxc8 proteins to the Pcna regulatory sequence was verified by chromatin immunoprecipitation assay. Taken together, our data suggest that Pcna is a direct downstream target of Hoxc8.


Asunto(s)
Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Antígeno Nuclear de Célula en Proliferación/genética , Animales , Secuencia de Bases , Células Cultivadas , Inmunoprecipitación de Cromatina , Proteínas de Homeodominio/genética , Ratones , Ratones Endogámicos ICR , Datos de Secuencia Molecular , Transcripción Genética
8.
J Biomed Biotechnol ; 2010: 231708, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20508826

RESUMEN

Using a ChIP-cloning technique, we identified a Zinc finger protein 804a (Zfp804a) as one of the putative Hoxc8 downstream target genes. We confirmed binding of Hoxc8 to an intronic region of Zfp804a by ChIP-PCR in F9 cells as well as in mouse embryos. Hoxc8 upregulated Zfp804a mRNA levels and augmented minimal promoter activity in vitro. In E11.5 mouse embryos, Zfp804a and Hoxc8 were coexpressed. Recent genome-wide studies identified Zfp804a (or ZNF804A in humans) as a plausible marker for schizophrenia, leading us to hypothesize that this embryogenic regulatory control might also exert influence in development of complex traits such as psychosis.

9.
Elife ; 92020 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-33382037

RESUMEN

Defective primary cilia cause a range of diseases known as ciliopathies, including hearing loss. The etiology of hearing loss in ciliopathies, however, remains unclear. We analyzed cochleae from three ciliopathy mouse models exhibiting different ciliogenesis defects: Intraflagellar transport 88 (Ift88), Tbc1d32 (a.k.a. bromi), and Cilk1 (a.k.a. Ick) mutants. These mutants showed multiple developmental defects including shortened cochlear duct and abnormal apical patterning of the organ of Corti. Although ciliogenic defects in cochlear hair cells such as misalignment of the kinocilium are often associated with the planar cell polarity pathway, our results showed that inner ear defects in these mutants are primarily due to loss of sonic hedgehog signaling. Furthermore, an inner ear-specific deletion of Cilk1 elicits low-frequency hearing loss attributable to cellular changes in apical cochlear identity that is dedicated to low-frequency sound detection. This type of hearing loss may account for hearing deficits in some patients with ciliopathies.


Asunto(s)
Polaridad Celular/fisiología , Ciliopatías/fisiopatología , Regulación del Desarrollo de la Expresión Génica/genética , Pérdida Auditiva/fisiopatología , Proteínas Hedgehog/metabolismo , Animales , Cilios/metabolismo , Modelos Animales de Enfermedad , Embrión de Mamíferos/metabolismo , Células Ciliadas Auditivas/metabolismo , Ratones , Vía de Señalización Wnt/fisiología
10.
Am J Transl Res ; 11(9): 6102-6109, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31632578

RESUMEN

Increasing evidence implicates chromatin structure and epigenetic regulation in various human developmental disorders, including facial abnormalities and intellectual disability. Mutations in CCCTC-binding factor (CTCF) demonstrate its role in craniofacial development, but early lethality precludes the use of Ctcf mutant mice for phenotypic investigations. In this study, we deleted Ctcf specifically in neural crest cells, the multipotent cells that give rise to many structures of the skeleton and connective tissues in the developing head. Although the pharyngeal arches were initially morphologically normal, many of the neural crest cell-derived skeletal and non-skeletal components were truncated in the Wnt1-Cre; Ctcffl/fl mutant mice. The expression level of chondrogenic and osteogenic-related genes were significantly decreased. Our results implicate CTCF in two distinct events in craniofacial development; first, in the regulation of outgrowth and morphogenesis by cell survival and proliferation, and second, in the differentiation of the facial skeleton. Our findings highlight the important contribution of CTCF to craniofacial pathologies.

12.
Mol Cells ; 41(7): 695-702, 2018 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-30008200

RESUMEN

The inner ear is a complex sensory organ responsible for hearing and balance. Formation of the inner ear is dependent on tight regulation of spatial and temporal expression of genes that direct a series of developmental processes. Recently, epigenetic regulation has emerged as a crucial regulator of the development of various organs. However, what roles higher-order chromatin organization and its regulator molecules play in inner ear development are unclear. CCCTC-binding factor (CTCF) is a highly conserved 11-zinc finger protein that regulates the three-dimensional architecture of chromatin, and is involved in various gene regulation processes. To delineate the role of CTCF in inner ear development, the present study investigated inner ear-specific Ctcf knockout mouse embryos (Pax2-Cre; Ctcffl/fl ). The loss of Ctcf resulted in multiple defects of inner ear development and severely compromised otic neurogenesis, which was partly due to a loss of Neurog1 expression. Furthermore, reduced Neurog1 gene expression by CTCF knockdown was found to be associated with changes in histone modification at the gene's promoter, as well as its upstream enhancer. The results of the present study demonstrate that CTCF plays an essential role in otic neurogenesis by modulating histone modification in the Neurog1 locus.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factor de Unión a CCCTC/metabolismo , Oído Interno/inervación , Sitios Genéticos , Histonas/metabolismo , Proteínas del Tejido Nervioso/genética , Neurogénesis , Procesamiento Proteico-Postraduccional , Acetilación , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Oído Interno/embriología , Oído Interno/patología , Embrión de Mamíferos/metabolismo , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Lisina/metabolismo , Ratones , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/efectos de los fármacos , Neurogénesis/genética , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Ganglio Espiral de la Cóclea/efectos de los fármacos , Ganglio Espiral de la Cóclea/metabolismo , Tretinoina/farmacología
13.
Mol Neurobiol ; 55(8): 6518-6532, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29327200

RESUMEN

The exocyst, an octameric protein complex consisting of Exoc1 through Exoc8, was first determined to regulate exocytosis by targeting vesicles to the plasma membrane in yeast to mice. In addition to this fundamental role, the exocyst complex has been implicated in other cellular processes. In this study, we investigated the role of the exocyst in cochlear development and hearing by targeting EXOC5, a central exocyst component. Deleting Exoc5 in the otic epithelium with widely used Cre lines resulted in early lethality. Thus, we generated two different inner ear-specific Exoc5 knockout models by crossing Gfi1Cre mice with Exoc5f/f mice for hair cell-specific deletion (Gfi1Cre/+;Exoc5f/f) and by in utero delivery of rAAV-iCre into the otocyst of embryonic day 12.5 for deletion throughout the otic epithelium (rAAV2/1-iCre;Exoc5f/f). Gfi1Cre/+;Exoc5f/f mice showed relatively normal hair cell morphology until postnatal day 20, after which hair cells underwent apoptosis accompanied by disorganization of stereociliary bundles, resulting in progressive hearing loss. rAAV2/1-iCre;Exoc5f/f mice exhibited abnormal neurite morphology, followed by apoptotic degeneration of spiral ganglion neurons (SGNs) and hair cells, which led to profound and early-onset hearing loss. These results demonstrate that Exoc5 is essential for the normal development and survival of cochlear hair cells and SGNs, as well as the functional maintenance of hearing.


Asunto(s)
Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patología , Audición , Neuronas/patología , Ganglio Espiral de la Cóclea/patología , Proteínas de Transporte Vesicular/metabolismo , Animales , Apoptosis , Supervivencia Celular , Proteínas de Unión al ADN/metabolismo , Dependovirus/metabolismo , Epitelio/patología , Células Ciliadas Auditivas/ultraestructura , Pérdida Auditiva/metabolismo , Pérdida Auditiva/patología , Integrasas/metabolismo , Ratones Endogámicos C57BL , Degeneración Nerviosa/patología , Neuritas/metabolismo , Neuronas/metabolismo , Órgano Espiral/metabolismo , Órgano Espiral/ultraestructura , Estereocilios/metabolismo , Estereocilios/ultraestructura , Factores de Transcripción/metabolismo , Proteínas de Transporte Vesicular/deficiencia
14.
Mol Cells ; 40(4): 314, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28486794

RESUMEN

[This corrects the article on p. 395 in vol. 39.].

15.
Mol Cells ; 39(5): 395-402, 2016 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-27025388

RESUMEN

Identifying Hoxc8 target genes is at the crux of understanding the Hoxc8-mediated regulatory networks underlying its roles during development. However, identification of these genes remains difficult due to intrinsic factors of Hoxc8, such as low DNA binding specificity, context-dependent regulation, and unknown cofactors. Therefore, as an alternative, the present study attempted to test whether the roles of Hoxc8 could be inferred by simply analyzing genes frequently coexpressed with Hoxc8, and whether these genes include putative target genes. Using archived gene expression datasets in which Hoxc8 was differentially expressed, we identified a total of 567 genes that were positively coexpressed with Hoxc8 in at least four out of eight datasets. Among these, 23 genes were coexpressed in six datasets. Gene sets associated with extracellular matrix and cell adhesion were most significantly enriched, followed by gene sets for skeletal system development, morphogenesis, cell motility, and transcriptional regulation. In particular, transcriptional regulators, including paralogs of Hoxc8, known Hox co-factors, and transcriptional remodeling factors were enriched. We randomly selected Adam19, Ptpn13, Prkd1, Tgfbi, and Aldh1a3, and validated their coexpression in mouse embryonic tissues and cell lines following TGF-ß2 treatment or ectopic Hoxc8 expression. Except for Aldh1a3, all genes showed concordant expression with that of Hoxc8, suggesting that the coexpressed genes might include direct or indirect target genes. Collectively, we suggest that the coexpressed genes provide a resource for constructing Hoxc8-mediated regulatory networks.


Asunto(s)
Expresión Génica , Proteínas de Homeodominio/genética , Factores de Transcripción/genética , Animales , Línea Celular , Bases de Datos Genéticas , Femenino , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Masculino , Ratones , Células 3T3 NIH , Análisis de Secuencia por Matrices de Oligonucleótidos
16.
Int J Biol Sci ; 9(9): 960-5, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24155669

RESUMEN

The Hox genes, which are organized into clusters on different chromosomes, are key regulators of embryonic anterior-posterior (A-P) body pattern formation and are expressed at specific times and in specific positions in developing vertebrate embryos. Previously, we have shown that histone methylation patterns are closely correlated with collinear Hox gene expression patterns along the A-P axis of E14.5 mouse embryos. Since histone modification is thought to play a crucial mechanistic role in the highly coordinated pattern of collinear Hox gene expression, we examined the maintenance of the spatial collinear expression pattern of Hoxc genes and the corresponding histone modifications during embryogenesis and in early postnatal mice. Hox expression patterns and histone modifications were analyzed by semi-quantitative RT-PCR and chromatin immunoprecipitation (ChIP)-PCR analyses, respectively. The spatiotemporal expression patterns of Hoxc genes in a cluster were maintained until the early postnatal stage (from E8.5 through P5). Examination of histone modifications in E14.5 and P5 tissues revealed that level of H3K27me3 is only a weak correlation with collinear Hoxc gene expression in the trunk regions although diminished in general, however the enrichment of H3K4me3 is strongly correlated with the gene expression in both stages. In summary, the initial spatiotemporal collinear expression pattern of Hoxc genes and epigenetic modifications are maintained after birth, likely contributing to the establishment of the gene expression code for position in the anatomic body axis throughout the entire life of the organism.


Asunto(s)
Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Genes Homeobox/fisiología , Histonas/metabolismo , Animales , Tipificación del Cuerpo/genética , Ensamble y Desensamble de Cromatina , Inmunoprecipitación de Cromatina , Desarrollo Embrionario/genética , Femenino , Perfilación de la Expresión Génica , Histonas/química , Metilación , Ratones , Ratones Endogámicos ICR , Reacción en Cadena en Tiempo Real de la Polimerasa
17.
Int J Biol Sci ; 8(6): 802-10, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22719220

RESUMEN

Hox genes are organized as clusters and specify regional identity along the anteroposterior body axis by sequential expression at a specific time and region during embryogenesis. However, the precise mechanisms underlying the sequential spatio-temporal, collinear expression pattern of Hox genes are not fully understood. Since epigenetic modifications such as chromatin architecture and histone modifications have become crucial mechanisms for highly coordinated gene expressions, we examined such modifications. E14.5 mouse embryos were dissected into three parts along the anteroposterior axis: brain, trunk-anterior, and trunk-posterior. Then, structural changes and epigenetic modifications were analyzed along the Hoxc cluster using chromosome conformation capture and chromatin immunoprecipitation-PCR methods. Hox non-expressing brain tissues had more compact, heterochromatin-like structures together with the strong repressive mark H3K27me3 than trunk tissues. In the trunk, however, a more loose euchromatin-like topology with a reduced amount of H3K27me3 modifications were observed along the whole cluster, regardless of their potency in gene activation. The active mark H3K4me3 was rather closely associated with the collinear expression of Hoxc genes; at trunk-anterior tissues, only 3' anterior Hoxc genes were marked by H3K4me3 upon gene activation, whereas whole Hoxc genes were marked by H3K4me3 and showed expression in trunk-posterior tissues. Altogether, these results indicated that loosening of the chromatin architecture and removing H3K27me3 were not sufficient for, but rather the concomitant acquisition of H3K4me3 drove the collinear expression of Hoxc genes.


Asunto(s)
Embrión de Mamíferos/metabolismo , Epigénesis Genética/genética , Proteínas de Homeodominio/genética , Animales , Cromatina/metabolismo , Inmunoprecipitación de Cromatina , Regulación del Desarrollo de la Expresión Génica/genética , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/genética
18.
Mol Cells ; 33(2): 111-6, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22228179

RESUMEN

AK007836 encodes a noncoding RNA (ncRNA) consisting of 2 exons. Since AK007836 is located just upstream of Pcna and transcribed in the opposite direction to that of Pcna, we analyzed its expression pattern. Both ncRNA and Pcna expressions were detected in in vitro and in vivo cells, showing a positive correlation. A 177 bp region separating the first exons of Pcna and AK007836 has a bidirectional promoter activity. When the expression of ncRNA was reduced by siRNA, Pcna expression was also reduced in normal cells, but not in cancer cells. These results suggest that the ncRNA is divergently transcribed from the bidirectional promoter, positively regulating the neighboring protein-coding Pcna gene transcription, and this regulatory function is somehow disrupted in cancer cells.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Antígeno Nuclear de Célula en Proliferación/genética , ARN no Traducido/metabolismo , Animales , Secuencia de Bases , Carcinoma Pulmonar de Lewis , Fibroblastos/metabolismo , Sitios Genéticos/genética , Melanoma Experimental , Ratones , Datos de Secuencia Molecular , Células 3T3 NIH , Regiones Promotoras Genéticas/genética , ARN Interferente Pequeño/genética , ARN no Traducido/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética
19.
Anat Cell Biol ; 43(1): 78-85, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21190008

RESUMEN

Spatially and temporally programmed expression of the Hox genes along the antero-posterior (A-P) axis is essential for correct pattern formation during embryonic development. An accumulating body of evidence indicates the pivotal role of spatial chromatin organization for the coordination of gene regulation. Recently, chromosome conformation capture (3C) technique has been developed and opened a new way to study chromosomal interactions in the nucleus. In this study, we describe 3C method we applied in F9 embryonic teratocarcinoma cells and demonstrate that the chromosomal interactions at Hox loci are successfully detected. Interestingly, at Hoxc loci, the abundance of intrachromosomal interactions with neighboring fragments was drastically decreased when the genes are expressed. These results indicate the possibility of the dynamic pattern of chromosomal interaction in association with the transcriptional regulation of Hox genes.

20.
Appl Biochem Biotechnol ; 160(3): 891-900, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19214787

RESUMEN

Hoxc8 has multiple roles in normal skeletal development. In this paper, a MC3T3-E1 subclone 4 osteogenic cell differentiation model was used to examine expression of Hoxc8 at multiple stages of osteogenesis. We found that Hoxc8 expression levels do not change in the early stage but increase in the middle stage and decrease in the late stage of osteogenesis. A knockdown of Hoxc8 by small-interfering RNA transfection in C2C12 cells indicated that Hoxc8 is a negative regulator of osteogenesis. Similarly, expression of Hoxc8 in C2C12 cells decreases alkaline phosphatase levels induced by bone morphogenetic protein-2 (BMP-2). The results of this study showed that Hoxc8 is involved in BMP-2-induced osteogenesis, and osteoblast differentiation in vitro is negatively regulated by Hoxc8, suggesting that Hoxc8 regulation is essential for osteoblast differentiation.


Asunto(s)
Diferenciación Celular , Proteínas de Homeodominio/metabolismo , Osteoblastos/citología , Animales , Proteínas Morfogenéticas Óseas/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular , Células Clonales , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Proteínas de Homeodominio/genética , Humanos , Ratones , Osteoblastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , ARN Interferente Pequeño/genética , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA