Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 275
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 43(4): 114083, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38602877

RESUMO

A common cause of deafness in humans is dysregulation of the endocochlear potential generated by the stria vascularis (SV). Thus, proper formation of the SV is critical for hearing. Using single-cell transcriptomics and a series of Shh signaling mutants, we discovered that the Shh receptor Patched1 (Ptch1) is essential for marginal cell (MC) differentiation and SV formation. Single-cell RNA sequencing analyses revealed that the cochlear roof epithelium is already specified into discrete domains with distinctive gene expression profiles at embryonic day 14, with Gsc as a marker gene of the MC lineage. Ptch1 deficiency leads to defective specification of MC precursors along the cochlear basal-apical regions. We demonstrated that elevated Gli2 levels impede MC differentiation through sustaining Otx2 expression and maintaining the progenitor state of MC precursors. Our results uncover an early specification of cochlear non-sensory epithelial cells and establish a crucial role of the Ptch1-Gli2 axis in regulating the development of SV.


Assuntos
Diferenciação Celular , Cóclea , Receptor Patched-1 , Estria Vascular , Receptor Patched-1/metabolismo , Receptor Patched-1/genética , Animais , Camundongos , Estria Vascular/metabolismo , Estria Vascular/citologia , Cóclea/metabolismo , Cóclea/embriologia , Cóclea/citologia , Transdução de Sinais , Proteína Gli2 com Dedos de Zinco/metabolismo , Proteína Gli2 com Dedos de Zinco/genética , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/genética
2.
Acta Otolaryngol ; 144(2): 96-99, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38511591

RESUMO

BACKGROUND: Water homeostasis is essential for inner ear function. Several aquaporins (AQPs), which are water transport proteins in the cell or plasma membrane, have been reported in the lateral wall of the rat inner ear (cochlea). However, the presence of AQP-10, -11 and -12 has not been reported in the rat stria vascularis (SV) to date. AIMS/OBJECTIVES: We have aimed to clarify the expression of AQP-10, -11 and -12 in the cochlea lateral wall. MATERIALS AND METHODS: Using Wistar rats, we examined the expression of AQP-10, -11 and -12 in the cochlea lateral wall using molecular approaches and immunohistochemistry. RESULTS: AQP-11 was molecular biologically expressed, but the expression of AQP-10 and -12 was not observed. Immunohistochemically, AQP-11 was diffusely localized in the basal cells and marginal cells of the rat SV but was not expressed at the apical site of marginal cells with double staining. The expression of AQP-10 and -12 was not observed. CONCLUSIONS AND SIGNIFICANCE: Only AQP-11 was expressed in the basal cells and marginal cells, but it was not expressed at the apical site of marginal cells. Based on this study, AQP-11 may not have an important role in water flux between the perilymph and endolymph.


Assuntos
Aquaporinas , Ratos Wistar , Estria Vascular , Animais , Ratos , Aquaporinas/metabolismo , Imuno-Histoquímica , Estria Vascular/metabolismo
3.
BMC Genomics ; 25(1): 213, 2024 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-38413848

RESUMO

BACKGROUND: The stria vascularis (SV), located in the lateral wall of the cochlea, maintains cochlear fluid homeostasis and mechanoelectrical transduction (MET) activity required for sound wave conduction. The pathogenesis of a number of human inheritable deafness syndromes, age related hearing loss, drug-induced ototoxicity and noise-induced hearing loss results from the morphological changes and functional impairments in the development of the SV. In this study, we investigate the implications of intercellular communication within the SV in the pathogenesis of sensorineural hearing loss (SNHL). We aim to identify commonly regulated signaling pathways using publicly available single-cell transcriptomic sequencing (scRNA-seq) datasets. METHODS: We analyzed scRNA-seq data, which was derived from studying the cochlear SV in mice with SNHL compared to normal adult mice. After quality control and filtering, we obtained the major cellular components of the mouse cochlear SV and integrated the data. Using Seurat's FindAllMarkers and FindMarkers packages, we searched for novel conservative genes and differential genes. We employed KEGG and GSEA to identify molecular pathways that are commonly altered among different types of SNHL. We utilized pySCENIC to discover new specific regulatory factors in SV subpopulation cells. With the help of CellChat, we identified changes in subpopulation cells showing similar trends across different SNHL types and their alterations in intercellular communication pathways. RESULTS: Through the analysis of the integrated data, we discovered new conserved genes to SV specific cells and identified common downregulated pathways in three types of SNHL. The enriched genes for these pathways showing similar trends are primarily associated with the Electron Transport Chain, related to mitochondrial energy metabolism. Using the CellChat package, we further found that there are shared pathways in the incoming signaling of specific intermediate cells in SNHL, and these pathways have common upstream regulatory transcription factor of Nfe2l2. Combining the results from pySCENIC and CellChat, we predicted the transcription factor Nfe2l2 as an upstream regulatory factor for multiple shared cellular pathways in IC. Additionally, it serves as an upstream factor for several genes within the Electron Transport Chain. CONCLUSION: Our bioinformatics analysis has revealed that downregulation of the mitochondrial electron transport chain have been observed in various conditions of SNHL. E2f1, Esrrb, Runx1, Yy1, and Gata2 could serve as novel important common TFs regulating the electron transport chain. Adm has emerged as a potential new marker gene for intermediate cells, while Itgb5 and Tesc show promise as potential new marker genes for marginal cells in the SV. These findings offer a new perspective on SV lesions in SNHL and provide additional theoretical evidence for the same drug treatment and prevention of different pathologies of SNHL.


Assuntos
Perda Auditiva Neurossensorial , Estria Vascular , Adulto , Humanos , Animais , Camundongos , Estria Vascular/metabolismo , Estria Vascular/patologia , Análise da Expressão Gênica de Célula Única , Perda Auditiva Neurossensorial/genética , Perda Auditiva Neurossensorial/patologia , Cóclea , Fatores de Transcrição/metabolismo
4.
Sci Rep ; 14(1): 3038, 2024 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-38321040

RESUMO

The stria vascularis (SV) is a stratified epithelium in the lateral wall of the mammalian cochlea, responsible for both endolymphatic ion homeostasis and generation of the endocochlear potential (EP) critical for normal hearing. The SV has three layers consisting predominantly of basal, intermediate, and marginal cells. Intermediate and marginal cells form an intricate interdigitated network of cell projections making discrimination of the cells challenging. To enable intermediate cell visualization, we engineered by BAC transgenesis, reporter mouse lines expressing ZsGreen fluorescent protein under the control of Kcnj10 promoter and regulatory sequences. Kcnj10 encodes KCNJ10 protein (also known as Kir4.1 or Kir1.2), an ATP-sensitive inwardly-rectifying potassium channel critical to EP generation, highly expressed in SV intermediate cells. In these transgenic mice, ZsGreen fluorescence mimics Kcnj10 endogenous expression in the cochlea and was detected in the intermediate cells of the SV, in the inner phalangeal cells, Hensen's, Deiters' and pillar cells, in a subset of spiral ganglion neurons, and in glial cells. We show that expression of the transgene in hemizygous mice does not alter auditory function, nor EP. These transgenic Tg(Kcnj10-ZsGreen) mice allow live and fixed tissue visualization of ZsGreen-expressing intermediate cells and will facilitate future studies of stria vascularis cell function.


Assuntos
Orelha Interna , Canais de Potássio Corretores do Fluxo de Internalização , Animais , Camundongos , Estria Vascular/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Cóclea/metabolismo , Orelha Interna/metabolismo , Camundongos Transgênicos , Mamíferos/metabolismo
5.
Int J Mol Sci ; 24(11)2023 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-37298241

RESUMO

TRPC channels are critical players in cochlear hair cells and sensory neurons, as demonstrated in animal experiments. However, evidence for TRPC expression in the human cochlea is still lacking. This reflects the logistic and practical difficulties in obtaining human cochleae. The purpose of this study was to detect TRPC6, TRPC5 and TRPC3 in the human cochlea. Temporal bone pairs were excised from ten body donors, and the inner ear was first assessed based on computed tomography scans. Decalcification was then performed using 20% EDTA solutions. Immunohistochemistry with knockout-tested antibodies followed. The organ of Corti, the stria vascularis, the spiral lamina, spiral ganglion neurons and cochlear nerves were specifically stained. This unique report of TRPC channels in the human cochlea supports the hypothesis of the potentially critical role of TRPC channels in human cochlear health and disease which has been suggested in previous rodent experiments.


Assuntos
Cóclea , Orelha Interna , Animais , Humanos , Imuno-Histoquímica , Cóclea/metabolismo , Orelha Interna/metabolismo , Estria Vascular/metabolismo , Audição
6.
BMC Med Genomics ; 16(1): 133, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37322474

RESUMO

BACKGROUND: The primary pathological alterations of Pendred syndrome are endolymphatic pH acidification and luminal enlargement of the inner ear. However, the molecular contributions of specific cell types remain poorly characterized. Therefore, we aimed to identify pH regulators in pendrin-expressing cells that may contribute to the homeostasis of endolymph pH and define the cellular pathogenic mechanisms that contribute to the dysregulation of cochlear endolymph pH in Slc26a4-/- mice. METHODS: We used single-cell RNA sequencing to identify both Slc26a4-expressing cells and Kcnj10-expressing cells in wild-type (WT, Slc26a4+/+) and Slc26a4-/- mice. Bioinformatic analysis of expression data confirmed marker genes defining the different cell types of the stria vascularis. In addition, specific findings were confirmed at the protein level by immunofluorescence. RESULTS: We found that spindle cells, which express pendrin, contain extrinsic cellular components, a factor that enables cell-to-cell communication. In addition, the gene expression profile informed the pH of the spindle cells. Compared to WT, the transcriptional profiles in Slc26a4-/- mice showed downregulation of extracellular exosome-related genes in spindle cells. Immunofluorescence studies in spindle cells of Slc26a4-/- mice validated the increased expression of the exosome-related protein, annexin A1, and the clathrin-mediated endocytosis-related protein, adaptor protein 2. CONCLUSION: Overall, cell isolation of stria vascularis from WT and Slc26a4-/- samples combined with cell type-specific transcriptomic analyses revealed pH-dependent alternations in spindle cells and intermediate cells, inspiring further studies into the dysfunctional role of stria vascularis cells in SLC26A4-related hearing loss.


Assuntos
Surdez , Estria Vascular , Camundongos , Animais , Estria Vascular/metabolismo , Estria Vascular/patologia , Proteínas de Transporte de Ânions/genética , Proteínas de Transporte de Ânions/metabolismo , Cóclea/metabolismo , Cóclea/patologia , Surdez/genética , Transportadores de Sulfato/genética , RNA/metabolismo
7.
Int J Mol Sci ; 23(24)2022 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-36555432

RESUMO

The stria vascularis (SV) contributes to cochlear homeostasis and consists of three layers, one of which contains the blood-labyrinthic barrier (BLB), with a large number of bovine cochlear pericytes (BCPs). Cisplatin is a chemotherapeutic drug that can damage the SV and cause hearing loss. In this study, cell viability, proliferation rate, cytotoxicity and reactive oxygen species production were evaluated. The protein content of phospho-extracellular signal-regulated kinases (ERK) 1/2, total ERK 1/2, phospho-cytosolic phospholipase A2 (cPLA2), total cPLA2 and cyclooxygenase 2 (COX-2) and the release of prostaglandin E2 (PGE2) and vascular endothelial growth factor (VEGF) from BCPs were analyzed. Finally, the protective effect of platelet-derived growth factor (PDGF-BB) on BCPs treated with cisplatin was investigated. Cisplatin reduced viability and proliferation, activated ERK 1/2, cPLA2 and COX-2 expression and increased PGE2 and VEGF release; these effects were reversed by Dexamethasone. The presence of PDGF-BB during the treatment with cisplatin significantly increased the proliferation rate. No studies on cell regeneration in ear tissue evaluated the effect of the PDGF/Dex combination. The aim of this study was to investigate the effects of cisplatin on cochlear pericytes and propose new otoprotective agents aimed at preventing the reduction of their vitality and thus maintaining the BLB structure.


Assuntos
Pericitos , Estria Vascular , Animais , Bovinos , Estria Vascular/metabolismo , Cisplatino/toxicidade , Cisplatino/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Becaplermina/metabolismo , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo
8.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35197290

RESUMO

Aminoglycosides (AGs) are commonly used antibiotics that cause deafness through the irreversible loss of cochlear sensory hair cells (HCs). How AGs enter the cochlea and then target HCs remains unresolved. Here, we performed time-lapse multicellular imaging of cochlea in live adult hearing mice via a chemo-mechanical cochleostomy. The in vivo tracking revealed that systemically administered Texas Red-labeled gentamicin (GTTR) enters the cochlea via the stria vascularis and then HCs selectively. GTTR uptake into HCs was completely abolished in transmembrane channel-like protein 1 (TMC1) knockout mice, indicating mechanotransducer channel-dependent AG uptake. Blockage of megalin, the candidate AG transporter in the stria vascularis, by binding competitor cilastatin prevented GTTR accumulation in HCs. Furthermore, cilastatin treatment markedly reduced AG-induced HC degeneration and hearing loss in vivo. Together, our in vivo real-time tracking of megalin-dependent AG transport across the blood-labyrinth barrier identifies new therapeutic targets for preventing AG-induced ototoxicity.


Assuntos
Antibacterianos/metabolismo , Gentamicinas/metabolismo , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Animais , Antibacterianos/toxicidade , Transporte Biológico , Cilastatina/farmacologia , Endolinfa/metabolismo , Gentamicinas/toxicidade , Células Ciliadas Auditivas/efeitos dos fármacos , Células Ciliadas Auditivas/metabolismo , Audição/efeitos dos fármacos , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/antagonistas & inibidores , Camundongos , Estria Vascular/metabolismo
9.
Neural Plast ; 2021: 5585394, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33959158

RESUMO

Purpose: Slc26a4-/- mice exhibit severer defects in the development of the cochlea and develop deafness, while the underlying mechanisms responsible for these effects remain unclear. Our study was to investigate the potential mechanism linking SLC26A4 deficiency to hearing loss. Materials and Methods: RNA sequencing was applied to analyze the differential gene expression of the stria vascularis (SV) from wildtype and Slc26a4-/- mice. GO and KEGG pathway analysis were performed. Quantitative RT-PCR was applied to validate the expression of candidate genes affected by Slc26a4. ELISA and immunofluorescence technique were used to detect the homocysteine (Hcy) level in serum, brain, and SV, respectively. Results: 183 upregulated genes and 63 downregulated genes were identified in the SV associated with Slc26a4 depletion. Transcriptomic profiling revealed that Slc26a4 deficiency significantly affected the expression of genes associated with cell adhesion, transmembrane transport, and the biogenesis of multicellular organisms. The SV from Slc26a4-/- mice exhibited a higher expression of Bhmt mRNAs, as well as altered homocysteine (Hcy) metabolism. Conclusions: The altered expression of Bhmt results in a dramatic change in multiple biochemical reactions and a disruption of nutrient homeostasis in the endolymph which may contribute to hearing loss of Slc26a4 knockout mouse.


Assuntos
Bócio Nodular/genética , Perda Auditiva Neurossensorial/genética , Homocisteína/metabolismo , Estria Vascular/metabolismo , Animais , Betaína-Homocisteína S-Metiltransferase/genética , Adesão Celular , Modelos Animais de Doenças , Regulação da Expressão Gênica , Bócio Nodular/patologia , Perda Auditiva Neurossensorial/patologia , Camundongos , Camundongos Knockout , RNA/genética , Transdução de Sinais/genética , Estria Vascular/patologia , Transportadores de Sulfato/genética , Transcriptoma
10.
Mol Brain ; 14(1): 82, 2021 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-34001214

RESUMO

Mitochondrial dysfunction is associated with aging and age-related hearing loss (AHL). However, the precise mechanisms underlying the pathophysiology of hearing loss remain unclear. Cdk5 regulatory subunit-associated protein 1 (CDK5RAP1) enables efficient intramitochondrial translation by catalyzing the deposition of 2-methylthio modifications on mitochondrial tRNAs. Here we investigated the effect of defective mitochondrial protein translation on hearing and AHL in a Cdk5rap1 deficiency C57BL/6 mouse model. Compared to control C57BL/6 mice, Cdk5rap1-knockout female mice displayed hearing loss phenotypically similar to AHL from an early age. The premature hearing loss in Cdk5rap1-knockout mice was associated with the degeneration of the spiral ligament and reduction of endocochlear potentials following the loss of auditory sensory cells. Furthermore, cultured primary mouse embryonic fibroblasts displayed early onset of cellular senescence associated with high oxidative stress and cell death. These results indicate that the CDK5RAP1 deficiency-induced defective mitochondrial translation might cause early hearing loss through the induction of cellular senescence and cochlear dysfunction in the inner ear. Our results suggest that the accumulation of dysfunctional mitochondria might promote AHL progression. Furthermore, our findings suggest that mitochondrial dysfunction and dysregulated mitochondrial tRNA modifications mechanistically cause AHL. Understanding the mechanisms underlying AHL will guide future clinical investigations and interventions in the attempt to mitigate the consequences of AHL.


Assuntos
Envelhecimento/patologia , Proteínas de Ciclo Celular/deficiência , Proteínas Mitocondriais/genética , Presbiacusia/genética , Transferases de Grupos de Enxofre/genética , Potenciais de Ação , Animais , Apoptose , Proteínas de Ciclo Celular/metabolismo , Feminino , Fibroblastos/metabolismo , Células Ciliadas Auditivas/metabolismo , Metaboloma , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Fenótipo , Espécies Reativas de Oxigênio/metabolismo , Gânglio Espiral da Cóclea/metabolismo , Ligamento Espiral da Cóclea/metabolismo , Estresse Fisiológico , Estria Vascular/metabolismo
11.
Pigment Cell Melanoma Res ; 34(3): 585-597, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33484097

RESUMO

In the inner ear, the neural crest gives rise to the glia of the VIII ganglion and two types of melanocytic cells: The pigmented cells of the vestibular system and intermediate cells of the stria vascularis. We analyzed the transcriptome of neonatal intermediate cells in an effort to better understand the development of the stria vascularis. We found that the expression of endothelin receptor B, which is essential for melanocyte development, persists in intermediate cells long after birth. In contrast, skin melanocytes rapidly downregulate the expression of EdnrB. Our findings suggest that endothelins might have co-opted new functions in the inner ear during evolution of the auditory organ.


Assuntos
Cóclea/metabolismo , Orelha Interna/metabolismo , Melanócitos/metabolismo , Receptor de Endotelina B/metabolismo , Pele/metabolismo , Transcriptoma , Animais , Cóclea/citologia , Orelha Interna/citologia , Regulação da Expressão Gênica no Desenvolvimento , Melanócitos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Receptor de Endotelina B/genética , Pele/citologia , Estria Vascular/citologia , Estria Vascular/metabolismo , Sistema Vestibular/citologia , Sistema Vestibular/metabolismo
12.
Hear Res ; 400: 108113, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33221698

RESUMO

Na+, K+-ATPase (Na,K-ATPase) is an ubiquitous enzyme in the inner ear and a key factor in the maintenance of the osmotic gradient of the endolymph. This study uses Na,K-ATPase α1 subunit immunoreactivity (IR) to identify cellular structures in the normal and disease human cochlea. Formalin-fixed celloidin-embedded (FFCE) human temporal bone sections were immunoreacted with mouse monoclonal antibodies against Na,K-ATPase α1 subunit. Na,K-ATPase α1 IR was examined in the cochlea of 30 patients: four with normal hearing, 5 with Meniere's disease, and 21 with other inner ear diseases: 11 male, 19 female; ages 42 to 96 years-old (yo), average age of 77 yo. Na,K-ATPase α1 IR area was quantified using the ImageJ software program. Na,K-ATPase α1 IR was located in the stria vascularis, and in type I, II and IV fibrocytes of the spiral ligament in the cochlea from patients with normal hearing. Na,K-ATPase α1 IR was seen in Deiters's cells and inner phalangeal cells of the organ of Corti. Na,K-ATPase α1 IR was present in satellite cells that surround the neurons of the spiral ganglia. In the inner ear of pathological specimens, Na,K-ATPase IR area was decreased (compared to the normal) in the stria vascularis, supporting cells in the organ of Corti and satellite cells of the spiral ganglia. These results show that Na,K-ATPase α1 IR is a good marker to identify cellular structures of the human inner ear and may be used to study cellular changes in the cochlea associated with aging and disease. The ubiquitous localization of Na,K-ATPase α1 in the human cochlea is consistent with the Na,K-ATPase role in ionic homeostasis and osmolarity, similar to that seen in animal models.


Assuntos
Orelha Interna , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Cóclea/metabolismo , Orelha Interna/metabolismo , Endolinfa/metabolismo , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , ATPase Trocadora de Sódio-Potássio/metabolismo , Estria Vascular/metabolismo
13.
Sci Rep ; 10(1): 18100, 2020 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-33093630

RESUMO

The stria vascularis (SV) in the cochlea generates and maintains the endocochlear potential, thereby playing a pivotal role in normal hearing. Knowing transcriptional profiles and gene regulatory networks of SV cell types establishes a basis for studying the mechanism underlying SV-related hearing loss. While we have previously characterized the expression profiles of major SV cell types in the adult mouse, transcriptional profiles of rare SV cell types remained elusive due to the limitation of cell capture in single-cell RNA-Seq. The role of these rare cell types in the homeostatic function of the adult SV remain largely undefined. In this study, we performed single-nucleus RNA-Seq on the adult mouse SV in conjunction with sample preservation treatments during the isolation steps. We distinguish rare SV cell types, including spindle cells and root cells, from other cell types, and characterize their transcriptional profiles. Furthermore, we also identify and validate novel specific markers for these rare SV cell types. Finally, we identify homeostatic gene regulatory networks within spindle and root cells, establishing a basis for understanding the functional roles of these cells in hearing. These novel findings will provide new insights for future work in SV-related hearing loss and hearing fluctuation.


Assuntos
Biomarcadores/análise , Cóclea/metabolismo , Regulação da Expressão Gênica , RNA-Seq/métodos , Estria Vascular/metabolismo , Transcriptoma , Animais , Feminino , Masculino , Camundongos
14.
PLoS One ; 15(8): e0237907, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32822386

RESUMO

Previous work demonstrates that the hearing loss in Alport mice is caused by defects in the stria vascularis. As the animals age, progressive thickening of strial capillary basement membranes (SCBMs) occurs associated with elevated levels of extracellular matrix expression and hypoxia-related gene and protein expression. These conditions render the animals susceptible to noise-induced hearing loss. In an effort to develop a more comprehensive understanding of how the underlying mutation in the COL4A3 gene influences homeostasis in the stria vascularis, we performed vascular permeability studies combined with RNA-seq analysis using isolated stria vascularis from 7-week old wild-type and Alport mice on the 129 Sv background. Alport SCBMs were found to be less permeable than wild-type littermates. RNA-seq and bioinformatics analysis revealed 68 genes were induced and 61 genes suppressed in the stria from Alport mice relative to wild-type using a cut-off of 2-fold. These included pathways involving transcription factors associated with the regulation of pro-inflammatory responses as well as cytokines, chemokines, and chemokine receptors that are up- or down-regulated. Canonical pathways included modulation of genes associated with glucose and glucose-1-PO4 degradation, NAD biosynthesis, histidine degradation, calcium signaling, and glutamate receptor signaling (among others). In all, the data point to the Alport stria being in an inflammatory state with disruption in numerous metabolic pathways indicative of metabolic stress, a likely cause for the susceptibility of Alport mice to noise-induced hearing loss under conditions that do not cause permanent hearing loss in age/strain-matched wild-type mice. The work lays the foundation for studies aimed at understanding the nature of strial pathology in Alport mice. The modulation of these genes under conditions of therapeutic intervention may provide important pre-clinical data to justify trials in humans afflicted with the disease.


Assuntos
Regulação da Expressão Gênica/genética , Perda Auditiva Provocada por Ruído/metabolismo , Nefrite Hereditária/metabolismo , Estria Vascular/metabolismo , Animais , Autoantígenos/genética , Autoantígenos/metabolismo , Membrana Basal/metabolismo , Quimiocinas/genética , Quimiocinas/metabolismo , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Regulação para Baixo , Matriz Extracelular/metabolismo , Feminino , Glucose/genética , Glucose/metabolismo , Perda Auditiva Provocada por Ruído/genética , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos , Nefrite Hereditária/genética , Nefrite Hereditária/patologia , RNA-Seq , Transdução de Sinais/genética , Estria Vascular/patologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcriptoma/genética , Regulação para Cima
15.
Neural Plast ; 2020: 2908182, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32802035

RESUMO

Background: Waardenburg syndrome is an uncommon genetic condition characterized by at least some degree of congenital hearing loss and pigmentation deficiencies. However, the genetic pathway affecting the development of stria vascularis is not fully illustrated. Methods: The transcript profile of stria vascularis of Waardenburg syndrome was studied using Mitf-M mutant pig and mice models. Therefore, GO analysis was performed to identify the differential gene expression caused by Mitf-M mutation. Results: There were 113 genes in tyrosine metabolism, melanin formation, and ion transportations showed significant changes in pig models and 191 genes in mice models. In addition, there were some spice's specific gene changes in the stria vascularis in the mouse and porcine models. The expression of tight junction-associated genes, including Cadm1, Cldn11, Pcdh1, Pcdh19, and Cdh24 genes, were significantly higher in porcine models compared to mouse models. Vascular-related and ion channel-related genes in the stria vascularis were also shown significantly difference between the two species. The expression of Col2a1, Col3a1, Col11a1, and Col11a2 genes were higher, and the expression of Col8a2, Cd34, and Ncam genes were lower in the porcine models compared to mouse models. Conclusions: Our data suggests that there is a significant difference on the gene expression and function between these two models.


Assuntos
Estria Vascular/metabolismo , Transcriptoma , Síndrome de Waardenburg/genética , Síndrome de Waardenburg/metabolismo , Animais , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Camundongos , Fator de Transcrição Associado à Microftalmia/genética , Mutação , Sus scrofa
16.
Hear Res ; 390: 107935, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32234583

RESUMO

In 129 Sv autosomal Alport mice, the strial capillary basement membranes (SCBMs) progressively thicken between 5 and 9 weeks of age resulting in a hypoxic microenvironment with metabolic stress and induction of pro-inflammatory cytokines and chemokines. These events occur concomitant with a drop in endocochlear potential and a susceptibility to noise-induced hearing loss under conditions that do not permanently affect age/strain-matched littermates. Here we aimed to gain an understanding of events that occur before the onset of SCBM thickening. Alport stria has normal thickness and shows levels of extracellular matrix (ECM) molecules in the SCBMs commensurate with wild-type mice. Hearing thresholds in the 3-week Alport mice do not differ from those of wild-type mice. We performed RNAseq analysis using RNA from stria vascularis isolated from 3-week Alport mice and wild type littermates. Data was processed using Ingenuity Pathway Analysis software and further distilled using manual procedures. RNAseq analysis revealed significant dysregulation of genes involved in cell adhesion, cell migration, formation of protrusions, and both actin and tubulin cytoskeletal dynamics. Overall, the data suggested changes in the cellular architecture of the stria might be apparent. To test this notion, we performed dual immunofluorescence analysis on whole mounts of the stria vascularis from these same animals stained with anti-isolectin gs-ib4 (endothelial cell marker) and anti-desmin (pericyte marker) antibodies. The results showed evidence of pericyte detachment and migration as well as the formation of membrane ruffling on pericytes in z-stacked confocal images from Alport mice compared to wild type littermates. This was confirmed by TEM analysis. Earlier work from our lab showed that endothelin A receptor blockade prevents SCBM thickening and ECM accumulation in the SCBMs. Treating cultured pericytes with endothelin-1 induced actin cytoskeletal rearrangement, increasing the ratio of filamentous to globular actin. Collectively, these findings suggest that the change in type IV collagen composition in the Alport SCBMs results in cellular insult to the pericyte compartment, activating detachment and altered cytoskeletal dynamics. These events precede SCBM thickening and hearing loss in Alport mice, and thus constitute the earliest event so far recognized in Alport strial pathology.


Assuntos
Citoesqueleto de Actina/ultraestrutura , Membrana Basal/ultraestrutura , Nefrite Hereditária/patologia , Pericitos/ultraestrutura , Estria Vascular/ultraestrutura , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Animais , Membrana Basal/efeitos dos fármacos , Membrana Basal/metabolismo , Adesão Celular , Movimento Celular , Células Cultivadas , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Modelos Animais de Doenças , Endotelina-1/farmacologia , Feminino , Imunofluorescência , Perfilação da Expressão Gênica , Masculino , Camundongos da Linhagem 129 , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Nefrite Hereditária/genética , Nefrite Hereditária/metabolismo , Pericitos/efeitos dos fármacos , Pericitos/metabolismo , RNA-Seq , Receptor de Endotelina A/agonistas , Receptor de Endotelina A/genética , Receptor de Endotelina A/metabolismo , Transdução de Sinais , Estria Vascular/efeitos dos fármacos , Estria Vascular/metabolismo
17.
Theranostics ; 9(24): 7184-7199, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31695761

RESUMO

Rationale: Mutations of SLC26A4 that abrogate pendrin, expressed in endolymphatic sac, cochlea and vestibule, are known to cause autosomal recessive sensorineural hearing loss with enlargement of the membranous labyrinth. This is the first study to demonstrate the feasibility of gene therapy for pendrin-related hearing loss. Methods: We used a recombinant viral vector to transfect Slc26a4 cDNA into embryonic day 12.5 otocysts of pendrin-deficient knock-out (Slc26a4∆/∆ ) and pendrin-deficient knock-in (Slc26a4tm1Dontuh/tm1Dontuh ) mice. Results: Local gene-delivery resulted in spatially and temporally limited pendrin expression, prevented enlargement, failed to restore vestibular function, but succeeded in the restoration of hearing. Restored hearing phenotypes included normal hearing as well as sudden, fluctuating, and progressive hearing loss. Conclusion: Our study illustrates the feasibility of gene therapy for pendrin-related hearing loss, suggests differences in the requirement of pendrin between the cochlea and the vestibular labyrinth, and documents that insufficient pendrin expression during late embryonal and early postnatal development of the inner ear can cause sudden, fluctuating and progressive hearing loss without obligatory enlargement of the membranous labyrinth.


Assuntos
Terapia Genética , Perda Auditiva Neurossensorial/genética , Perda Auditiva Neurossensorial/terapia , Audição/genética , Mutação/genética , Transportadores de Sulfato/genética , Animais , Cóclea/metabolismo , Dependovirus , Orelha Interna/metabolismo , Saco Endolinfático/embriologia , Saco Endolinfático/metabolismo , Células Epiteliais/metabolismo , Células Ciliadas Auditivas/metabolismo , Concentração de Íons de Hidrogênio , Camundongos Endogâmicos C57BL , Camundongos Knockout , Membrana dos Otólitos/patologia , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Estria Vascular/metabolismo , Transportadores de Sulfato/metabolismo , Transcrição Gênica
18.
Drug Des Devel Ther ; 13: 3199-3206, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31686782

RESUMO

BACKGROUND: p66Shc, a Src homologue and collagen homologue (Shc) adaptor protein, mediates oxidative stress signaling. The p66Shc-null mice have increased lifespan and enhanced resistance to oxidative stress. Studies have also indicated its potential role in inner ear aging, which can lead to deafness. OBJECTIVE: The aim of this study was to determine the effects of p66Shc down-regulation on the marginal cells (MCs) of the inner ear stria vascularis. METHODS: Primary MCs were isolated from neonatal rats and treated with glucose oxidase to induce oxidative stress. The cells were transduced with adenovirus expressing siRNA, and the knockdown was verified by Western blotting. The reactive oxygen species (ROS) levels and apoptosis were analyzed using the DCFH-DA probe and Annexin-V/7-AAD staining respectively. The ultrastructure of the differentially-treated cells was examined by transmission electron microscopy (TEM).Results: The in vitro oxidative stress model was established successfully in rat MCs. Knockdown of p66Shc alleviated the high ROS levels and apoptosis in the glucose oxidase-treated cells. In addition, glucose oxidase significantly increased the number of peroxisomes in the MCs, which was decreased by p66Shc inhibition. CONCLUSION: Oxidative stress increases p66Shc levels in the marginal cells of the inner ear, which aggravates ROS production and cellular injury. Blocking p66Shc expression can effectively reduce oxidative stress and protect the MCs.


Assuntos
Apoptose , Modelos Animais de Doenças , Regulação para Baixo , Estresse Oxidativo , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src/deficiência , Estria Vascular/metabolismo , Animais , Células Cultivadas , Células Epiteliais/metabolismo , Ratos , Ratos Sprague-Dawley , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src/metabolismo
19.
Int J Pediatr Otorhinolaryngol ; 127: 109656, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31470202

RESUMO

OBJECTIVE: To determine the distribution of perivascularresident macrophages (PVMs) in BLB and their relationship with capillaries, and to explore the possible mechanisms responsible for lipopolysaccharide (LPS)-induced activation of PVMs and the breakdown of BLB. METHODS: Adult Balb/c mice were either trans-tympanically injected with LPS, or mock-treated. Auditory brainstem response was tested before and 48 h after treatments. Distribution of pericytes, PVMs and capillaries was analyzed by immunohistochemical staining, and BLB permeability was estimated by FITC-dextran leakage assay. Ultrastructure of stria vascularis was examined by transmission electron microscope. Protein and mRNA level of matrix metallopeptidase 9 (MMP-9), zona occludens-1 (ZO-1), interleukin-33 (IL-33) and its receptor suppression of tumorigenicity 2 (ST2) was measured by IHC and qRT-PCR. RESULTS: Unlike pericytes that surround one capillary, PVMs branched to connect with more than one capillary. LPS caused hearing loss in mice. Following LPS challenge, cochleae showed vascular leakage in stria vascularis, and PVMs presented morphological changes including reduced contact with capillaries. TEM revealed a reduced number of tight junction contact points between endothelial cells and a wider space between PVMs, pericytes and endothelial cells. The mRNA and protein levels of MMP-9 and ST2 in stria vascularis were up-regulated, while ZO-1 were down-regulated after exposure to LPS. CONCLUSIONS: Our results suggest that PVMs may play a more significant role than pericytes in maintaining the integrity of BLB. Our findings also reveal a possible mechanism contributing to LPS-induced activation of PVMs, breakdown of BLB and hearing loss.


Assuntos
Perda Auditiva/metabolismo , Macrófagos/patologia , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Estria Vascular/ultraestrutura , Vestíbulo do Labirinto , Animais , Capilares/patologia , Cóclea/irrigação sanguínea , Modelos Animais de Doenças , Regulação para Baixo , Células Endoteliais/ultraestrutura , Potenciais Evocados Auditivos do Tronco Encefálico/efeitos dos fármacos , Perda Auditiva/induzido quimicamente , Proteína 1 Semelhante a Receptor de Interleucina-1/genética , Proteína 1 Semelhante a Receptor de Interleucina-1/metabolismo , Lipopolissacarídeos , Macrófagos/ultraestrutura , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica de Transmissão , Pericitos/ultraestrutura , Estria Vascular/metabolismo , Estria Vascular/patologia , Junções Íntimas/ultraestrutura , Regulação para Cima , Proteína da Zônula de Oclusão-1/genética , Proteína da Zônula de Oclusão-1/metabolismo
20.
ORL J Otorhinolaryngol Relat Spec ; 81(4): 202-214, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31340214

RESUMO

Cisplatin is an anti-cancer drug that causes oxotoxic side effects such as impairment of inner ear function and hearing loss. We aimed to investigate the effects of allicin against cisplatin-induced stria vascularis damage in mice, and to clarify the mechanism underlying the protective effects of allicin against ototoxicity. Stria vascularis injury was induced in mice by intraperitoneal injection of cisplatin, which was significantly prevented by pretreatment with allicin. Allicin not only reduced cisplatin-activated expression of cleaved caspase-3 in marginal cells, PVM/Ms (perivascular resident macrophage-like melanocytes), and basal cells of the stria vascularis, but also decreased the expression of poly(ADP-ribose) polymerase-1 (PARP-1) and apoptosis-inducing factor (AIF) nuclear translocation in the stria vascularis cells. Our results demonstrate that allicin plays an effective role in protecting stria vascularis injury induced by cisplatin by inhibiting caspase-dependent, as well as caspase-independent PARP-1-AIF-mediated apoptotic pathways. Therefore, allicin may be useful in preventing cisplatin-induced ototoxicity.


Assuntos
Apoptose , Caspase 3/efeitos dos fármacos , Perda Auditiva/prevenção & controle , Poli(ADP-Ribose) Polimerase-1/antagonistas & inibidores , Estria Vascular/efeitos dos fármacos , Ácidos Sulfínicos/farmacologia , Animais , Antioxidantes/farmacologia , Caspase 3/metabolismo , Cisplatino/toxicidade , Modelos Animais de Doenças , Dissulfetos , Feminino , Perda Auditiva/induzido quimicamente , Perda Auditiva/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Poli(ADP-Ribose) Polimerase-1/metabolismo , Estria Vascular/metabolismo , Estria Vascular/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...