Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
ChemMedChem ; : e202400038, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38818625

RESUMEN

Hearing loss is a significant disability that often goes under recognised, largely due to poor identification, prevention, and treatment. Steps are being made to amend these pitfalls in the investigation of hearing loss, however, the development of a cure to reverse advanced forms remains distant. This review details some current advances in the treatment of hearing loss, with a particular focus on genetic-based nanotechnology and how it may provide a useful avenue for further research. This review presents a broad background on the pathophysiology of hearing loss and some current interventions. We also highlight some potential genes that may be useful in the amelioration of hearing loss. Pathways of cellular differentiation from stem or supporting cell to functional hair cell are covered in detail, as this mechanism represents a key means of regenerating these cell types. Overall, we believe that polymer-based nanotechnology coupled with novel excipients represents a useful area of further research in the treatment of hearing loss, although further studies in this area are required.

2.
Biomedicines ; 11(12)2023 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-38137568

RESUMEN

Up to 1.5 billion people worldwide suffer from various forms of hearing loss, with an additional 1.1 billion people at risk from various insults such as increased consumption of recreational noise-emitting devices and ageing. The most common type of hearing impairment is sensorineural hearing loss caused by the degeneration or malfunction of cochlear hair cells or spiral ganglion nerves in the inner ear. There is currently no cure for hearing loss. However, emerging frontier technologies such as gene, drug or cell-based therapies offer hope for an effective cure. In this review, we discuss the current therapeutic progress for the treatment of hearing loss. We describe and evaluate the major therapeutic approaches being applied to hearing loss and summarize the key trials and studies.

3.
Front Pharmacol ; 14: 1207141, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37927600

RESUMEN

Despite significant advances in the development of therapeutics for hearing loss, drug delivery to the middle and inner ear remains a challenge. As conventional oral or intravascular administration are ineffective due to poor bioavailability and impermeability of the blood-labyrinth-barrier, localized delivery is becoming a preferable approach for certain drugs. Even then, localized delivery to the ear precludes continual drug delivery due to the invasive and potentially traumatic procedures required to access the middle and inner ear. To address this, the preclinical development of controlled release therapeutics and drug delivery devices have greatly advanced, with some now showing promise clinically. This review will discuss the existing challenges in drug development for treating the most prevalent and damaging hearing disorders, in particular otitis media, perforation of the tympanic membrane, cholesteatoma and sensorineural hearing loss. We will then address novel developments in drug delivery that address these including novel controlled release therapeutics such as hydrogel and nanotechnology and finally, novel device delivery approaches such as microfluidic systems and cochlear prosthesis-mediated delivery. The aim of this review is to investigate how drugs can reach the middle and inner ear more efficiently and how recent innovations could be applied in aiding drug delivery in certain pathologic contexts.

4.
J Drug Target ; 31(6): 585-595, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37211674

RESUMEN

Gene therapies are becoming more abundantly researched for use in a multitude of potential treatments, including for hearing loss. Hearing loss is a condition which impacts an increasing number of the population each year, with significant burdens associated. As such, this review will present the concept that delivering a gene effectively to the inner ear may assist in expanding novel treatment options and improving patient outcomes. Historically, several drawbacks have been associated with the use of gene therapies, some of which may be overcome via targeted delivery. Targeted delivery has the potential to alleviate off-target effects and permit a safer delivery profile. Viral vectors have often been described as a delivery method, however, there is an emerging depiction of the potential for nanotechnology to be used. Resulting nanoparticles may also be tuned to allow for targeted delivery. Therefore, this review will focus on hearing loss, gene delivery techniques and inner ear targets, including highlighting promising research. Targeted delivery is a key concept to permitting gene delivery in a safe effective manner, however, further research is required, both in the determination of genes to use in functional hearing recovery and formulating nanoparticles for targeted delivery.


Asunto(s)
Oído Interno , Pérdida Auditiva , Humanos , Pérdida Auditiva/genética , Pérdida Auditiva/terapia , Terapia Genética/métodos , Técnicas de Transferencia de Gen , Vectores Genéticos
5.
Small ; 19(8): e2204986, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36538754

RESUMEN

Hearing loss impacts a large proportion of the global population. Damage to the inner ear, in particular the sensitive hair cells, can impact individuals for the rest of their lives. There are very limited options for interventions after damage to these cells has occurred. Targeted gene delivery may provide an effective means to trigger appropriate differentiation of progenitor cells for effective replacement of these sensitive hair cells. There are several hurdles that need to be overcome to effectively deliver these genes. Nanoencapsulation technology has previously been used for the delivery of pharmaceuticals, proteins and nucleic acids, and may provide an effective means of delivering genes to trigger appropriate differentiation. This review investigates the background of hearing loss, current advancements and pitfalls of gene delivery, and how nanoencapsulation may be useful.


Asunto(s)
Oído Interno , Pérdida Auditiva , Humanos , Ácidos y Sales Biliares , Oído Interno/metabolismo , Pérdida Auditiva/genética , Pérdida Auditiva/metabolismo , Pérdida Auditiva/terapia , Técnicas de Transferencia de Gen , Terapia Genética
6.
J Pharm Sci ; 112(3): 700-707, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36150468

RESUMEN

The biocompatibility and effects on cells' bioactivity of developed pharmaceuticals are crucial properties, required to permit their safe delivery. Nanogel matrices offer a promising role in emerging pharmaceutics; however, it is crucial that they and their excipients do not demonstrate detrimental effects on the cells to which they interact. This study investigated the use of Teflon and the secondary bile acid deoxycholic acid in the formation of novel nanogel matrices. Each has properties which may be of benefit for the nanogels created and their use in the pharmaceutical industry. Rheological parameters and scanning electron microscopy studies were conducted. In order to assess the developed nanogels' impacts on cellular bioactivity, studies using Seahorse assays were conducted on three cell types, hepatic, muscle and pancreatic beta cells. Results demonstrated the addition of Teflon did not alter the morphological characteristics of resulting nanogels or the metabolic profiles of the cell lines. Interestingly, pancreatic beta cells highlighted the potential of Teflon to exert a protective profile from mitochondrial damage. Overall, the developed nanogels showed potentially promising profiles in certain studies conducted which may lead to future research.


Asunto(s)
Polietilenglicoles , Politetrafluoroetileno , Nanogeles , Polietileneimina
7.
Stem Cell Res ; 50: 102129, 2020 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-33360097

RESUMEN

Mutations in the USH2A gene are the most common cause of Usher syndrome and autosomal recessive non-syndromic retinitis pigmentosa. Here, we describe the generation of three induced pluripotent stem cell lines from dermal fibroblasts derived from a patient carrying biallelic c.949C > A and c.1256G > T variants in the USH2A gene, using episomal reprogramming plasmids expressing OCT4, SOX2, KLF4, MYCL, LIN28, mir302/367 and shRNA targeting TP53. All three lines expressed pluripotency markers, displayed unaltered karyotypes as well as trilineage differentiation potential, and were negative for reprogramming episomes and mycoplasma.

8.
Front Genet ; 11: 565216, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33193648

RESUMEN

Usher syndrome (USH) is an autosomal recessive (AR) disorder that permanently and severely affects the senses of hearing, vision, and balance. Three clinically distinct types of USH have been identified, decreasing in severity from Type 1 to 3, with symptoms of sensorineural hearing loss (SNHL), retinitis pigmentosa (RP), and vestibular dysfunction. There are currently nine confirmed and two suspected USH-causative genes, and a further three candidate loci have been mapped. The proteins encoded by these genes form complexes that play critical roles in the development and maintenance of cellular structures within the inner ear and retina, which have minimal capacity for repair or regeneration. In the cochlea, stereocilia are located on the apical surface of inner ear hair cells (HC) and are responsible for transducing mechanical stimuli from sound pressure waves into chemical signals. These signals are then detected by the auditory nerve fibers, transmitted to the brain and interpreted as sound. Disease-causing mutations in USH genes can destabilize the tip links that bind the stereocilia to each other, and cause defects in protein trafficking and stereocilia bundle morphology, thereby inhibiting mechanosensory transduction. This review summarizes the current knowledge on Usher syndrome with a particular emphasis on mutations in USH genes, USH protein structures, and functional analyses in animal models. Currently, there is no cure for USH. However, the genetic therapies that are rapidly developing will benefit from this compilation of detailed genetic information to identify the most effective strategies for restoring functional USH proteins.

9.
Nucleic Acids Res ; 48(6): 2880-2896, 2020 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-31956913

RESUMEN

The transcription factor Six1 is essential for induction of sensory cell fate and formation of auditory sensory epithelium, but how it activates gene expression programs to generate distinct cell-types remains unknown. Here, we perform genome-wide characterization of Six1 binding at different stages of auditory sensory epithelium development and find that Six1-binding to cis-regulatory elements changes dramatically at cell-state transitions. Intriguingly, Six1 pre-occupies enhancers of cell-type-specific regulators and effectors before their expression. We demonstrate in-vivo cell-type-specific activity of Six1-bound novel enhancers of Pbx1, Fgf8, Dusp6, Vangl2, the hair-cell master regulator Atoh1 and a cascade of Atoh1's downstream factors, including Pou4f3 and Gfi1. A subset of Six1-bound sites carry consensus-sequences for its downstream factors, including Atoh1, Gfi1, Pou4f3, Gata3 and Pbx1, all of which physically interact with Six1. Motif analysis identifies RFX/X-box as one of the most significantly enriched motifs in Six1-bound sites, and we demonstrate that Six1-RFX proteins cooperatively regulate gene expression through binding to SIX:RFX-motifs. Six1 targets a wide range of hair-bundle regulators and late Six1 deletion disrupts hair-bundle polarity. This study provides a mechanistic understanding of how Six1 cooperates with distinct cofactors in feedforward loops to control lineage-specific gene expression programs during progressive differentiation of the auditory sensory epithelium.


Asunto(s)
Diferenciación Celular/genética , Linaje de la Célula/genética , Epitelio/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Ciliadas Auditivas/citología , Células Ciliadas Auditivas/metabolismo , Proteínas de Homeodominio/metabolismo , Secuencia de Bases , Polaridad Celular , Secuencia de Consenso , ADN/metabolismo , Fosfatasa 6 de Especificidad Dual/genética , Fosfatasa 6 de Especificidad Dual/metabolismo , Elementos de Facilitación Genéticos/genética , Factor 8 de Crecimiento de Fibroblastos/genética , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Sitios Genéticos , Genoma , Células Ciliadas Auditivas/ultraestructura , Humanos , Complejos Multiproteicos/metabolismo , Motivos de Nucleótidos/genética , Unión Proteica , Transducción de Señal/genética
10.
Stem Cell Res ; 36: 101420, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30904819

RESUMEN

The human iPSC lines LEIi010-A and LEIi010-B were generated from the dermal fibroblasts of a patient with Usher syndrome using episomal plasmids containing OCT4, SOX2, KLF4, L-MYC, LIN28, mir302/367 microRNA and shRNA for p53. These iPSC lines carry compound heterozygous mutations (c.949C > A and c.1256G > T) in USH2A. LEIi010-A and LEIi010-B expressed pluripotent stem cell markers, had a normal karyotype and could be differentiated into endoderm, mesoderm and ectodermal lineages.


Asunto(s)
Línea Celular , Proteínas de la Matriz Extracelular/genética , Células Madre Pluripotentes Inducidas , Síndromes de Usher/genética , Diferenciación Celular , Técnicas de Reprogramación Celular , Fibroblastos , Heterocigoto , Humanos , Cariotipo , Factor 4 Similar a Kruppel , Piel
11.
FASEB J ; 33(6): 6838-6851, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30808210

RESUMEN

The channel that governs mechanotransduction (MT) by hair cells in the inner ear has been investigated intensively for 4 decades, but its precise molecular composition remains enigmatic. Transmembrane channel-like protein 1 (TMC1) was recently identified as a component of the MT channel, and lipoma HMGIC fusion partner-like 5 (LHFPL5) is considered to be part of the MT complex and may functionally couple the tip link to the MT channel. As components of the MT complex, TMC1 and LHFPL5 are expected to localize at the lower end of the tip link in hair cells, a notion generally supported by previous studies on neonatal mice. However, the localization of these 2 proteins, particularly in the hair cells of adult mice, remains incompletely elucidated. Because determination of TMC1 and LHFPL5 localization at distinct developmental stages is essential for understanding their function and regulation, we used several approaches to examine the localization of these proteins in neonatal and adult hair cells in the mouse. We report several notable findings: 1) TMC1 and LHFPL5 predominantly localize at the tip of the shorter rows of stereocilia in neonatal hair cells, which largely verifies the previously published findings in neonatal hair cells; 2) LHFPL5 persists in the hair bundle of hair cells after postnatal day (P)7, which clarifies the previously reported unexpected absence of LHFPL5 after P7 and supports the view that LHFPL5 is a permanent component in the MT complex; and 3) TMC1 and LHFPL5 remain at the tip of the shorter rows of stereocilia in adult outer hair cells, but in adult inner hair cells, TMC1 is uniformly distributed in both the tallest row and the shorter rows of stereocilia, whereas LHFPL5 is uniformly distributed in the shorter rows of stereocilia. These findings raise intriguing questions regarding the turnover rate, regulation, additional functions, and functional interaction of TMC1 and LHFPL5. Our study confirms the previous findings in neonatal hair cells and reveals several previously unidentified aspects of TMC1 and LHFPL5 localization in more mature hair cells.-Li, X., Yu, X., Chen, X., Liu, Z., Wang, G., Li, C., Wong, E. Y. M., Sham, M. H., Tang, J., He, J., Xiong, W., Liu, Z., Huang, P. Localization of TMC1 and LHFPL5 in auditory hair cells in neonatal and adult mice.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Auditivas/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Animales Recién Nacidos , Sistemas CRISPR-Cas , Mecanotransducción Celular , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados
12.
Mol Cell Biol ; 37(1)2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27795300

RESUMEN

EYA1 is known to be overexpressed in human breast cancer, in which the Myc protein is also accumulated in association with decreased phospho-T58 (pT58) levels. We have recently reported that EYA1 functions as a unique protein phosphatase to dephosphorylate Myc at pT58 to regulate Myc levels. However, it remains unclear whether EYA1-mediated Myc dephosphorylation on T58 is a critical function in regulating Myc protein stability in breast cancer. Furthermore, EYA1's substrate specificity has remained elusive. In this study, we have investigated these questions, and here, we report that depletion of EYA1 using short hairpin RNA (shRNA) in breast cancer cells destabilizes the Myc protein and increases pT58 levels, leading to an increase in the doubling time and impairment of cell cycle progression. In correlation with EYA1-mediated stabilization of cMyc and reduced levels of pT58, EYA1 greatly reduced cMyc-FBW7 binding and cMyc ubiquitination, thus providing novel insight into how EYA1 acts to regulate the FBW7-mediated Myc degradation machinery. We found that the conserved C-terminal haloacid dehalogenase domain of EYA1, which has been reported to have only tyrosine phosphatase activity, has dual phosphatase activities, and both the N- and C-terminal domains interact with substrates to increase the catalytic activity of EYA1. Enzymatic assay and nuclear magnetic resonance (NMR) analysis demonstrated that EYA1 has a striking conformation preference for phospho-T58 of Myc. Together, our results not only provide novel structural evidence about the conformation specificity of EYA1 in dephosphorylating phosphothreonine in Myc but also reveal an important mechanism contributing to Myc deregulation in human breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas F-Box/metabolismo , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Proteínas Tirosina Fosfatasas/química , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ciclo Celular , Línea Celular Tumoral , Proteína 7 que Contiene Repeticiones F-Box-WD , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Modelos Moleculares , Fosforilación , Unión Proteica , Conformación Proteica , Estabilidad Proteica , Proteínas Proto-Oncogénicas c-myc/química , Treonina/metabolismo
13.
PLoS One ; 11(5): e0154984, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27171474

RESUMEN

Mouse N-ethyl-N-nitrosourea (ENU) mutagenesis has generated many useful animal models for human diseases. Here we describe the identification of a novel ENU-induced mouse mutant strain Turner (Tur) that displays circling and headtossing behavior and progressive hearing loss. Tur/Tur homozygous animals lack Preyer and righting reflexes and display severe headtossing and reaching response defect. We mapped the Tur mutation to a critical region of 11 cM on chromosome 9 that includes myosin VI. Direct sequence analysis revealed a c.820A>T substitution in exon 8 of the Myo6 gene that changes amino acid Asn200 to Ile (p.N200I) in the motor domain. Analysis of inner ear hair cells by immunohistochemistry, scanning electron microscopy and histology revealed degeneration of hair cells in the inner ear and structural malformation of the stereocilia in the cochlea of Turner homozygous mutant mice. Our data indicate that this novel mouse strain provides a useful model for future studies on the function of myosin VI in mammalian auditory and non-auditory systems and in human syndromes.


Asunto(s)
Sordera/genética , Sordera/fisiopatología , Mutación/genética , Cadenas Pesadas de Miosina/genética , Vestíbulo del Laberinto/fisiopatología , Animales , Umbral Auditivo , Secuencia de Bases , Conducta Animal , Cromosomas de los Mamíferos/genética , Epitelio/patología , Epitelio/ultraestructura , Etilnitrosourea , Potenciales Evocados Auditivos del Tronco Encefálico , Genes Dominantes , Células Ciliadas Auditivas/patología , Células Ciliadas Auditivas/ultraestructura , Masculino , Ratones Endogámicos C57BL , Ratones Mutantes , Penetrancia , Vestíbulo del Laberinto/ultraestructura
14.
Dev Cell ; 31(4): 434-47, 2014 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-25458011

RESUMEN

Self-renewal and proliferation of nephron progenitor cells and the decision to initiate nephrogenesis are crucial events directing kidney development. Despite recent advancements in defining lineage and regulators for the progenitors, fundamental questions about mechanisms driving expansion of the progenitors remain unanswered. Here we show that Eya1 interacts with Six2 and Myc to control self-renewing cell activity. Cell fate tracing reveals a developmental restriction of the Eya1(+) population within the intermediate mesoderm to nephron-forming cell fates and a common origin shared between caudal mesonephric and metanephric nephrons. Conditional inactivation of Eya1 leads to loss of Six2 expression and premature epithelialization of the progenitors. Six2 mediates translocation of Eya1 to the nucleus, where Eya1 uses its threonine phosphatase activity to control Myc phosphorylation/dephosphorylation and function in the progenitor cells. Our results reveal a functional link between Eya1, Six2, and Myc in driving the expansion and maintenance of the multipotent progenitors during nephrogenesis.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Nefronas/citología , Nefronas/metabolismo , Proteínas Nucleares/metabolismo , Organogénesis/fisiología , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Células Madre/citología , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular/fisiología , Mesodermo/metabolismo , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Madre/metabolismo
15.
Mol Cell Biol ; 33(5): 927-36, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23263983

RESUMEN

The phosphatase and transactivator EYA family proteins are overexpressed in many cancer cell lines and are abundantly distributed in undifferentiated cells during development. Loss-of-function studies have shown that EYA1 is required for cell proliferation and survival during mammalian organogenesis. However, how EYA1 is regulated during development is unknown. Here, we report that EYA1 is regulated throughout the cell cycle via ubiquitin-mediated proteolysis. The level of EYA1 protein fluctuates in the cell cycle, peaking during mitosis and dropping drastically as cells exit into G(1). We found that EYA1 is efficiently degraded during mitotic exit in a Cdh1-dependent manner and that these two proteins physically interact. Overexpression of Cdh1 reduces the protein levels of ectopically expressed or endogenous EYA1, whereas depletion of Cdh1 by RNA interference stabilizes the EYA1 protein. Together, our results indicate that anaphase-promoting complex/cyclosome (APC/C)-Cdh1 specifically targets EYA1 for degradation during M-to-G(1) transition, failure of which may compromise cell proliferation and survival.


Asunto(s)
División Celular , Fase G1 , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Ciclosoma-Complejo Promotor de la Anafase , Animales , Ciclo Celular , Línea Celular , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Proteínas Nucleares/genética , Proteínas Tirosina Fosfatasas/genética , Interferencia de ARN , Complejos de Ubiquitina-Proteína Ligasa/genética , Ubiquitinación , Regulación hacia Arriba , Xenopus
16.
Hear Res ; 297: 13-9, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23104013

RESUMEN

The phosphatase-transactivator EYA1 interacts with the homeodomain protein SIX1 to form transcriptional activation complexes, which play essential roles in regulating cell proliferation, survival and induction of sensory and neuronal differentiation programs during inner ear development. Mutations of the Eya1 and Six1 genes cause profound developmental auditory defects in mice and humans. The molecular mechanisms and developmental processes controlled by the EYA1 and SIX1 complex in inner ear development and neurosensory fate induction are the focus of this review.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Mutación , Neuronas/metabolismo , Proteínas Nucleares/fisiología , Proteínas Tirosina Fosfatasas/fisiología , Animales , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Supervivencia Celular , Electroporación , Células Ciliadas Auditivas/citología , Humanos , Ratones , Neuronas/fisiología , Proteómica/métodos , Transcripción Genética
17.
Dev Cell ; 22(2): 377-90, 2012 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-22340499

RESUMEN

Inner-ear hair cell differentiation requires Atoh1 function, while Eya1, Six1, and Sox2 are coexpressed in sensory progenitors and mutations in these genes cause sensorineural hearing loss. However, how these genes are linked functionally and the transcriptional networks controlling hair cell induction remain unclear. Here, we show (1) that Eya1/Six1 are necessary for hair cell development, and their coexpression in mouse cochlear explants is sufficient to induce hair cell fate in the nonsensory epithelium expressing low-level Sox2 by activating not only Atoh1-dependent but also Atoh1-independent pathways and (2) that both pathways induce Pou4f3 to promote hair cell differentiation. Sox2 cooperates with Eya1/Six1 to synergistically activate Atoh1 transcription via direct binding to the conserved Sox- and Six-binding sites in Atoh1 enhancers, and these proteins physically interact. Our findings demonstrate that direct and cooperative interactions between the Sox2, Six1, and Eya1 proteins coordinate Atoh1 expression to specify hair cell fate.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Cóclea/metabolismo , Células Ciliadas Auditivas/metabolismo , Proteínas de Homeodominio/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Factores de Transcripción SOXB1/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Western Blotting , Diferenciación Celular , Inmunoprecipitación de Cromatina , Cóclea/citología , Ensayo de Cambio de Movilidad Electroforética , Electroporación , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Técnicas para Inmunoenzimas , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Mutación/genética , Proteínas Nucleares/genética , Fosforilación , Proteínas Tirosina Fosfatasas/genética , Factores de Transcripción SOXB1/genética
18.
Dev Biol ; 352(2): 382-92, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21320481

RESUMEN

The spatial regulation of combinatorial expression of Hox genes is critical for determining hindbrain rhombomere (r) identities. To address the cross-regulatory relationship between Hox genes in hindbrain neuronal specification, we have generated a gain-of-function transgenic mouse mutant Hoxb3(Tg) using the Hoxb2 r4-specific enhancer element. Interestingly, in r4 of the Hoxb3(Tg) mutant where Hoxb3 was ectopically expressed, the expression of Hoxb1 was specifically abolished. The hindbrain neuronal defects of the Hoxb3(Tg) mutant mice were similar to those of Hoxb1(-/-) mutants. Therefore, we hypothesized that Hoxb3 could directly suppress Hoxb1 expression. We first identified a novel Hoxb3 binding site S3 on the Hoxb1 locus and confirmed protein binding to this site by EMSA, and by in vivo ChIP analysis using P19 cells and hindbrain tissues from the Hoxb3(Tg) mutant. We further showed that Hoxb3 could suppress Hoxb1 transcriptional activity by chick in ovo luciferase reporter assay. Moreover, in E10.5 wildtype caudal hindbrain, where Hoxb1 is not expressed, we showed by in vivo ChIP that Hoxb3 was consistently bound to the S3 site on the Hoxb1 gene. This study reveals a novel negative regulatory mechanism by which Hoxb3 as a posterior gene serves to restrict Hoxb1 expression in r4 by direct transcriptional repression to maintain the rhombomere identity.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Rombencéfalo/embriología , Rombencéfalo/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas Aviares/genética , Proteínas Aviares/metabolismo , Secuencia de Bases , Sitios de Unión/genética , Tipificación del Cuerpo , Embrión de Pollo , Anomalías Craneofaciales/embriología , Anomalías Craneofaciales/genética , Anomalías Craneofaciales/metabolismo , Cartilla de ADN/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Ratones , Ratones Mutantes , Ratones Transgénicos , Modelos Neurológicos , Neurogénesis/genética , Neurogénesis/fisiología
19.
Biol Reprod ; 70(3): 775-84, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14627543

RESUMEN

VCY2 is a testis-specific protein that locates in a frequently deleted azoospermia factor c region on chromosome Yq. Although its genomic structure has been characterized, the function of VCY2 is still unknown. To gain insight regarding the likely function of VCY2, we investigated the proteins that interact with VCY2 using the yeast two-hybrid system. We identified a novel VCY2 interaction partner, named VCY2IP-1, that encodes an open reading frame of 1059 amino acids. The amino acid sequence of VCY2IP-1 shows 59.3% and 41.9% homology to two human microtubule-associated proteins (MAPs), MAP1B and MAP1A, respectively. VCY2IP-1 has an extensive homology to the N-terminus and C-terminus regions of MAP1B and MAP1A, placing it within a large family of MAPs. We mapped VCY2IP-1 to chromosome 19p13.11. The VCY2IP-1 gene spans 15 kilobases (kb) and consists of seven exons. Northern blot analysis identified a single, intense band of approximately 3.2-kb VCY2IP-1 transcript, predominantly expressed in human testis. In situ hybridization of human testicular sections showed the localization of VCY2IP-1 transcripts in germ cells, and reverse transcription-polymerase chain reaction analysis demonstrated the presence of VCY2 and VCY2IP-1 transcripts in human ejaculated spermatozoa. Our expression data support the involvement of VCY2 and VCY2IP-1 in spermatogenesis. Based on the high homology of VCY2IP-1 with MAPs, we propose the involvement of VCY2 in the cytoskeletal network via interaction with VCY2IP-1.


Asunto(s)
Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Testículo/fisiología , Secuencia de Aminoácidos , Secuencia de Bases , Cromosomas Humanos Par 19 , Biblioteca de Genes , Humanos , Inmunoprecipitación , Masculino , Datos de Secuencia Molecular , Proteínas/metabolismo , ARN Mensajero/análisis , Homología de Secuencia de Aminoácido , Espermatozoides/fisiología , Técnicas del Sistema de Dos Híbridos , Levaduras/fisiología
20.
Biochem Biophys Res Commun ; 296(5): 1104-11, 2002 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-12207887

RESUMEN

VCY2 locates in the AZFc region on chromosome Yq and is frequently deleted in infertile men with severe oligozoospermia or azoospermia. VCY2 is a testis-specific protein with unknown function. This study was to identify the protein that interacts with VCY2. We used the full-length VCY2 as bait to screen the human testis cDNA library using yeast two-hybrid approach. We identified a number of positive-interacting clones that encode ubiquitin-protein ligase E3A (UBE3A). UBE3A contains a HECT domain that binds VCY2. The specificity of the interaction was confirmed by co-immunoprecipitation and yeast mating. Northern blot analyses revealed two UBE3A transcripts 1.4 and 2kb that were abundantly expressed in human testis. We also showed that both VCY2 and UBE3A mRNAs were expressed in ejaculated human spermatozoa, indicating that both genes localize in the germ cell compartment. These data suggest that UBE3A ubiquitination may be required for VCY2 function.


Asunto(s)
Ligasas/química , Ligasas/metabolismo , Proteínas/metabolismo , Secuencia de Aminoácidos , Humanos , Infertilidad Masculina/genética , Ligasas/genética , Masculino , Datos de Secuencia Molecular , Pruebas de Precipitina , Estructura Terciaria de Proteína , Proteínas/genética , Proteínas/fisiología , ARN Mensajero/biosíntesis , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crecimiento & desarrollo , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Espermatozoides/metabolismo , Testículo/metabolismo , Distribución Tisular , Técnicas del Sistema de Dos Híbridos , Ubiquitina-Proteína Ligasas , Ubiquitinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...