Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Microvasc Res ; 143: 104403, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35753505

RESUMO

Blood-labyrinth barrier (BLB) disruption plays a crucial role in the development of otitis media. The aims of our study was to explore the role and action mechanism of adenosine in LPS-induced endothelial cells (ECs) damage, which are one of the major principal cell type for blood-labyrinth barrier (BLB), and so as to assess the potential of adenosine to be used in the treatment of BLB disruption in animal experiment. In our study, ECs were treated with LPS to mimic BLB damage in vitro. Our data showed that adenosine at dosage of 1, 10, and 20 µM had no influence on the cell viability of ECs. LPS treatment obviously suppressed the expression of Occludin and Zonula occludens-1 (ZO-1) in ECs, which was partly recused by adenosine treatment. Meantime, LPS-induced increasing in reactive oxygen species (ROS) production and ECs permeability also was rescued by adenosine treatment. However, inhibition the A2A receptor (A2AR) could attenuate the influence of adenosine on LPS-treated ECs, indicating that adenosine alleviated LPS-induced BLB damage by activating A2AR. Moreover, the inhibition of adenosine to LPS-induced inactivation of AMPK/AKT signaling pathway was partly recused by A2AR suppression. In addition, Compound C (an AMPK inhibitor) decreased the expression of Occludin and ZO-1 in ECs following LPS combined with adenosine treatment. In conclusion, adenosine alleviates LPS-induced BLB damage via AMPK/AKT pathway through activation of A2AR. This work suggests that adenosine may be a candidate drug for the treatment of BLB dysfunction-related diseases.


Assuntos
Células Endoteliais , Lipopolissacarídeos , Proteínas Quinases Ativadas por AMP/metabolismo , Adenosina/metabolismo , Adenosina/farmacologia , Animais , Células Endoteliais/metabolismo , Lipopolissacarídeos/farmacologia , Ocludina/metabolismo , Permeabilidade , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo
2.
Am J Pathol ; 190(9): 1943-1959, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32562655

RESUMO

Acoustic trauma disrupts cochlear blood flow and damages sensory hair cells. Damage and regression of capillaries after acoustic trauma have long been observed, but the underlying mechanism of pathology has not been understood. We show herein that loud sound causes change of phenotype from neural/glial antigen 2 positive/α-smooth muscle actin negative to neural/glial antigen 2 positive/α-smooth muscle actin positive in some pericytes (PCs) on strial capillaries that is strongly associated with up-regulation of transforming growth factor-ß1. The acoustic trauma also reduced capillary density and increased deposition of matrix proteins, particularly in the vicinity of transformed PCs. In a newly established in vitro three-dimensional endothelial cell (EC) and PC co-culture model, transformed PCs induced thicker capillary-like branches in ECs and increased collagen IV and laminin expression. Transplantation of exogenous PCs derived from neonatal day 10 mouse cochleae to acoustic traumatized cochleae, however, significantly attenuated the decreased vascular density in the stria. Transplantation of PCs pretransfected with adeno-associated virus 1-vascular endothelial growth factor-A165 under control of a hypoxia-response element markedly promotes vascular volume and blood flow, increased proliferation of PCs and ECs, and attenuated loud sound-caused loss in endocochlear potential and hearing. Our results indicate that loud sound-triggered PC transformation contributes to capillary wall thickening and regression, and young PC transplantation effectively rehabilitates the vascular regression and improves hearing.


Assuntos
Capilares/patologia , Cóclea/patologia , Perda Auditiva Provocada por Ruído/patologia , Pericitos/patologia , Pericitos/transplante , Animais , Atrofia/patologia , Transdiferenciação Celular , Cóclea/irrigação sanguínea , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miofibroblastos/patologia
3.
Hum Genet ; 135(5): 513-524, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27023905

RESUMO

The high prevalence/incidence of hearing loss (HL) in humans makes it the most common sensory defect. The majority of the cases are of genetic origin. Non-syndromic hereditary HL is extremely heterogeneous. Genetic approaches have been instrumental in deciphering genes that are crucial for auditory function. In this study, we first used NADf chip to exclude the implication of known North-African mutations in HL in a large consanguineous Tunisian family (FT13) affected by autosomal recessive non-syndromic HL (ARNSHL). We then performed genome-wide linkage analysis and assigned the deafness gene locus to ch:5q23.2-31.1, corresponding to the DFNB60 ARNSHL locus. Moreover, we performed whole exome sequencing on FT13 patient DNA and uncovered amino acid substitution p.Cys113Tyr in SLC22A4, a transporter of organic cations, cosegregating with HL in FT13 and therefore the cause of ARNSHL DFNB60. We also screened a cohort of small Tunisian HL families and uncovered an additional deaf proband of consanguineous parents that is homozygous for p.Cys113Tyr carried by the same microsatellite marker haplotype as in FT13, indicating that this mutation is ancestral. Using immunofluorescence, we found that Slc22a4 is expressed in stria vascularis (SV) endothelial cells of rodent cochlea and targets their apical plasma membrane. We also found Slc22a4 transcripts in our RNA-seq library from purified primary culture of mouse SV endothelial cells. Interestingly, p.Cys113Tyr mutation affects the trafficking of the transporter and severely alters ergothioneine uptake. We conclude that SLC22A4 is an organic cation transporter of the SV endothelium that is essential for hearing, and its mutation causes DFNB60 form of HL.


Assuntos
Cóclea/patologia , Consanguinidade , Endotélio/patologia , Genes Recessivos/genética , Perda Auditiva/genética , Mutação/genética , Proteínas de Transporte de Cátions Orgânicos/genética , Sequência de Aminoácidos , Animais , Células Cultivadas , Cóclea/metabolismo , Endotélio/metabolismo , Exoma/genética , Feminino , Células HEK293 , Perda Auditiva/patologia , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Linhagem , Reação em Cadeia da Polimerase , Polimorfismo de Fragmento de Restrição , Ratos , Ratos Sprague-Dawley , Homologia de Sequência de Aminoácidos , Simportadores
4.
Cell Tissue Res ; 361(3): 685-96, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25740201

RESUMO

Tight control over cochlear blood flow (CoBF) and the blood-labyrinth barrier (BLB) in the striavascularis is critical for maintaining the ionic, fluid and energy balance necessary for hearing function. Inefficient CoBF and disruption of BLB integrity have long been considered major etiologic factors in a variety of hearing disorders. In this study, we investigate structural changes in the BLB of the striavascularis in age-graded C57BL/6 mice (1 to 21 months) with a focus on changes in two blood barrier accessory cells, namely pericytes (PCs) and perivascular-resident macrophage-like melanocytes (PVM/Ms). Decreased capillary density was detectable at 6 months, with significant capillary degeneration seen in 9- to 21-month-old mice. Reduced capillary density was highly correlated with lower numbers of PCs and PVM/Ms. "Drop-out" of PCs and "activation" of PVM/Ms were seen at 6 months, with drastic changes being observed by 21 months. With newly established in vitro three-dimensional cell-based co-culture models, we demonstrate that PCs and PVM/Ms are essential for maintaining cochlear vascular architecture and stability.


Assuntos
Envelhecimento/fisiologia , Permeabilidade Capilar/fisiologia , Cóclea/irrigação sanguínea , Orelha Interna/metabolismo , Macrófagos/citologia , Melanócitos/metabolismo , Animais , Técnicas de Cocultura , Camundongos Endogâmicos C57BL , Pericitos/citologia
5.
Proc Natl Acad Sci U S A ; 109(26): 10388-93, 2012 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-22689949

RESUMO

The microenvironment of the cochlea is maintained by the barrier between the systemic circulation and the fluids inside the stria vascularis. However, the mechanisms that control the permeability of the intrastrial fluid-blood barrier remain largely unknown. The barrier comprises endothelial cells connected to each other by tight junctions and an underlying basement membrane. In a recent study, we found that the intrastrial fluid-blood barrier also includes a large number of perivascular cells with both macrophage and melanocyte characteristics. The perivascular-resident macrophage-like melanocytes (PVM/Ms) are in close contact with vessels through cytoplasmic processes. Here we demonstrate that PVM/Ms have an important role in maintaining the integrity of the intrastrial fluid-blood barrier and hearing function. Using a cell culture-based in vitro model and a genetically induced PVM/M-depleted animal model, we show that absence of PVM/Ms increases the permeability of the intrastrial fluid-blood barrier to both low- and high-molecular-weight tracers. The increased permeability is caused by decreased expression of pigment epithelial-derived factor, which regulates expression of several tight junction-associated proteins instrumental to barrier integrity. When tested for endocochlear potential and auditory brainstem response, PVM/M-depleted animals show substantial drop in endocochlear potential with accompanying hearing loss. Our results demonstrate a critical role for PVM/Ms in regulating the permeability of the intrastrial fluid-blood barrier for establishing a normal endocochlear potential hearing threshold.


Assuntos
Orelha Interna/patologia , Macrófagos/fisiologia , Melanócitos/fisiologia , Animais , Humanos , Camundongos , Camundongos Transgênicos , Estria Vascular/fisiologia , Junções Íntimas/fisiologia
6.
FASEB J ; 27(9): 3730-40, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23729595

RESUMO

Tissue perivascular resident macrophages (PVM/Ms), a hybrid cell type with characteristics of both macrophages and melanocytes, are critical for establishing and maintaining the endocochlear potential (EP) required for hearing. The PVM/Ms modulate expression of tight- and adherens-junction proteins in the endothelial barrier of the stria vascularis (intrastrial fluid-blood barrier) through secretion of a signaling molecule, pigment epithelium growth factor (PEDF). Here, we identify a significant link between abnormalities in PVM/Ms and endothelial barrier breakdown from acoustic trauma to the mouse ear. We find that acoustic trauma causes activation of PVM/Ms and physical detachment from capillary walls. Concurrent with the detachment, we find loosened tight junctions between endothelial cells and decreased production of tight- and adherens-junction protein, resulting in leakage of serum proteins from the damaged barrier. A key factor in the intrastrial fluid-blood barrier hyperpermeability exhibited in the mice is down-regulation of PVM/M modulated PEDF production. We demonstrate that delivery of PEDF to the damaged ear ameliorates hearing loss by restoring intrastrial fluid-blood barrier integrity. PEDF up-regulates expression of tight junction-associated proteins (ZO-1 and VE-cadherin) and PVM/M stabilizing neural cell adhesion molecule (NCAM-120). These studies point to the critical role PVM/Ms play in regulating intrastrial fluid-blood barrier integrity in healthy and noise-damaged ears.


Assuntos
Perda Auditiva Provocada por Ruído/metabolismo , Perda Auditiva Provocada por Ruído/patologia , Perda Auditiva/metabolismo , Perda Auditiva/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Melanócitos/metabolismo , Melanócitos/patologia , Junções Aderentes/metabolismo , Junções Aderentes/patologia , Animais , Células Cultivadas , Orelha/lesões , Orelha/patologia , Ensaio de Imunoadsorção Enzimática , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Junções Íntimas/metabolismo , Junções Íntimas/patologia
7.
Elife ; 122023 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-36719173

RESUMO

The inner ear has a rich population of pericytes, a multi-functional mural cell essential for sensory hair cell heath and normal hearing. However, the mechanics of how pericytes contribute to the homeostasis of the auditory vascular-neuronal complex in the spiral ganglion are not yet known. In this study, using an inducible and conditional pericyte depletion mouse (PDGFRB-CreERT2; ROSA26iDTR) model, we demonstrate, for the first time, that pericyte depletion causes loss of vascular volume and spiral ganglion neurons (SGNs) and adversely affects hearing sensitivity. Using an in vitro trans-well co-culture system, we show pericytes markedly promote neurite and vascular branch growth in neonatal SGN explants and adult SGNs. The pericyte-controlled neural growth is strongly mediated by pericyte-released exosomes containing vascular endothelial growth factor-A (VEGF-A). Treatment of neonatal SGN explants or adult SGNs with pericyte-derived exosomes significantly enhances angiogenesis, SGN survival, and neurite growth, all of which were inhibited by a selective blocker of VEGF receptor 2 (Flk1). Our study demonstrates that pericytes in the adult ear are critical for vascular stability and SGN health. Cross-talk between pericytes and SGNs via exosomes is essential for neuronal and vascular health and normal hearing.


Assuntos
Pericitos , Gânglio Espiral da Cóclea , Animais , Camundongos , Fator A de Crescimento do Endotélio Vascular , Neurônios/fisiologia , Neuritos/fisiologia
8.
J Vis Exp ; (175)2021 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-34633361

RESUMO

Transduction of sound is metabolically demanding, and the normal function of the microvasculature in the lateral wall is critical for maintaining endocochlear potential, ion transport, and fluid balance. Different forms of hearing disorders are reported to involve abnormal microcirculation in the cochlea. Investigation of how cochlear blood flow (CoBF) pathology affects hearing function is challenging due to the lack of feasible interrogation methods and the difficulty in accessing the inner ear. An open vessel-window in the lateral cochlear wall, combined with fluorescence intravital microscopy, has been used for studying CoBF changes in vivo, but mostly in guinea pig and only recently in the mouse. This paper and the associated video describe the open vessel-window method for visualizing blood flow in the mouse cochlea. Details include 1) preparation of the fluorescent-labeled blood cell suspension from mice; 2) construction of an open vessel-window for intravital microscopy in an anesthetized mouse, and 3) measurement of blood flow velocity and volume using an offline recording of the imaging. The method is presented in video format to show how to use the open window approach in mouse to investigate structural and functional changes in the cochlear microcirculation under normal and pathological conditions.


Assuntos
Hemodinâmica , Microscopia Intravital , Animais , Cóclea , Cobaias , Camundongos , Microcirculação , Microscopia de Fluorescência
9.
Hum Cell ; 34(4): 1093-1102, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33768511

RESUMO

Perivascular-resident macrophage-like melanocytes (PVM/Ms) can upregulate the expression of tight junction-related proteins in endothelial cells (ECs) by secreting pigment epithelial-derived factor (PEDF), and thereby regulate the permeability of the intrastrial fluid-blood barrier critical for maintaining inner ear homeostasis. This study aimed to investigate the effects of long non-coding RNA (lncRNA) Rian on cell growth of PVM/Ms and PVM/Ms regulation of intrastrial fluid-blood barrier integrity mediated by PEDF. Rian was downregulated in the aged cochlea from 12-month-old C57BL/6 mice. Rian overexpression inhibited cell apoptosis and promoted cell viability of hypoxia-injured PVM/Ms as well as increased the concentration and expression of PEDF secreted by PVM/Ms. In contrast, Rian silencing exerted the opposite effects. Furthermore, in a cell co-culture model of ECs and PVM/Ms, Rian overexpression in PVM/Ms increased the expression of the junction-associated proteins in co-cultured ECs, and this effect was abrogated by blockade of PEDF by anti-PEDF in PVM/Ms. Further mechanistical investigation revealed that Rian promoted STAT3 nuclear translocation and activation by binding to FUS, and thereby promoted the secretion of PEDF. Collectively, Rian attenuates PVM/Ms injury and strengthens the ability of PVM/Ms to maintain the integrity of the endothelial barrier by promoting PEDF expression.


Assuntos
Células Endoteliais/metabolismo , Proteínas do Olho/metabolismo , Expressão Gênica/genética , Melanócitos/fisiologia , Fatores de Crescimento Neural/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , RNA Longo não Codificante/fisiologia , Serpinas/metabolismo , Proteínas de Junções Íntimas/genética , Proteínas de Junções Íntimas/metabolismo , Animais , Apoptose/genética , Sobrevivência Celular/genética , Células Cultivadas , Cóclea/metabolismo , Proteínas do Olho/genética , Camundongos Endogâmicos C57BL , Fatores de Crescimento Neural/genética , Proteínas Nucleares/metabolismo , Ligação Proteica/genética , Proteína FUS de Ligação a RNA/metabolismo , Fator de Transcrição STAT3/metabolismo , Serpinas/genética
10.
JCI Insight ; 6(8)2021 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-33690221

RESUMO

Millions of people are affected by hearing loss. Hearing loss is frequently caused by noise or aging and often associated with loss of pericytes. Pericytes populate the small vessels in the adult cochlea. However, their role in different types of hearing loss is largely unknown. Using an inducible and conditional pericyte depletion mouse model and noise-exposed mouse model, we show that loss of pericytes leads to marked changes in vascular structure, in turn leading to vascular degeneration and hearing loss. In vitro, using advanced tissue explants from pericyte fluorescence reporter models combined with exogenous donor pericytes, we show that pericytes, signaled by VEGF isoform A165 (VEGFA165), vigorously drive new vessel growth in both adult and neonatal mouse inner ear tissue. In vivo, the delivery of an adeno-associated virus serotype 1-mediated (AAV1-mediated) VEGFA165 viral vector to pericyte-depleted or noise-exposed animals prevented and regenerated lost pericytes, improved blood supply, and attenuated hearing loss. These studies provide the first clear-cut evidence that pericytes are critical for vascular regeneration, vascular stability, and hearing in adults. The restoration of vascular function in the damaged cochlea, including in noise-exposed animals, suggests that VEGFA165 gene therapy could be a new strategy for ameliorating vascular associated hearing disorders.


Assuntos
Cóclea/irrigação sanguínea , Perda Auditiva Provocada por Ruído/fisiopatologia , Neovascularização Fisiológica/genética , Pericitos/patologia , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Orelha Interna/irrigação sanguínea , Terapia Genética , Perda Auditiva Provocada por Ruído/terapia , Técnicas In Vitro , Camundongos , Camundongos Transgênicos
11.
Biomed Res Int ; 2021: 6624744, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34258273

RESUMO

OBJECTIVE: Genetic variants in the WFS1 gene can cause Wolfram syndrome (WS) or autosomal dominant nonsyndromic low-frequency hearing loss (HL). This study is aimed at investigating the molecular basis of HL in an affected Chinese family and the genotype-phenotype correlation of WFS1 variants. METHODS: The clinical phenotype of the five-generation Chinese family was characterized using audiological examinations and pedigree analysis. Target exome sequencing of 129 known deafness genes and bioinformatics analysis were performed among six patients and four normal subjects to screen suspected pathogenic variants. We built a complete WFS1 protein model to assess the potential effects of the variant on protein structure. RESULTS: A novel heterozygous pathogenic variant NM_006005.3 c.2020G>T (p.Gly674Trp) was identified in the WFS1 gene, located in the C-terminal domain of the wolframin protein. We further showed that HL-related WFS1 missense variants were mainly concentrated in the endoplasmic reticulum (ER) domain. In contrast, WS-related missense variants are randomly distributed throughout the protein. CONCLUSIONS: In this family, we identified a novel variant p.Gly674Trp of WFS1 as the primary pathogenic variant causing the low-frequency sensorineural HL, enriching the mutational spectrum of the WFS1 gene.


Assuntos
Retículo Endoplasmático/metabolismo , Genes Dominantes , Perda Auditiva/genética , Proteínas de Membrana/genética , Mutação de Sentido Incorreto/genética , Adulto , Idoso de 80 Anos ou mais , Sequência de Bases , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Linhagem , Fenótipo
12.
Front Physiol ; 11: 974, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32922309

RESUMO

Objective: Connexin 43 (Cx43) is a protein constituent of gap junctions (GJs) in various barrier cells, especially astrocytes and microglia of the blood-brain-barrier (BBB), where it plays an important role in intercellular communication and regulation of the barrier. Despite the importance of Cx43 in other blood barriers, not much attention has been paid to expression and function of Cx43 in the blood-labyrinth-barrier (BLB) of the stria vascularis in the cochlea. Methods: We used multiple research approaches, including immunocytochemical staining, patch-clamp dye loading technique, real-time quantitative reverse transcription (RT)-PCR, western blot, measurement of endocochlear potential (EP) with an electrode through the scala media, and auditory brainstem response to test hearing function. Results: We found Cx43 expressed in vascular endothelial cells (ECs) and perivascular resident macrophages (PVMs) in the stria vascularis of adult C57BL/6 mouse cochleae. In particular, we found Cx43 expressed in foot processes of PVMs at points of contact with the endothelium. Consistent with Cx43 expression in vivo, we also found Cx43 expressed in EC-EC and EC-PVM interfaces in a co-cultured cell line model. Using a patch-clamp dye loading technique, we demonstrated that Alexa Fluor® 568 dye injected into PVMs diffuses to connected neighboring ECs. The functional coupling between the ECs and PVMs is blocked by 18α-Glycyrrhetinic acid (18α-GA), a GJ blocker. Suppression of Cx43 with small interfering RNA (siRNA) in vivo significantly elevated hearing threshold and caused the EP to drop and the blood barrier to become more permeable. In further study, using in vitro primary EC cell line models, we demonstrated that suppression of Cx43 disrupts intercellular tight junctions (TJs) in the EC monolayer and increases endothelial monolayer permeability. Conculsion: Taken together, these findings underscore the importance of Cx43 expression in the normal ear for maintaining BLB integrity, normal EP, and hearing function.

13.
Mol Med Rep ; 16(6): 8643-8648, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28990052

RESUMO

Previous studies have suggested that macrophage migration inhibitory factor (MIF) serves an important role in hearing function; however, the underlying mechanism remains unclear. In the present study, perivascular­resident macrophage­like melanocytes (PVM/Ms) from the stria vascularis of the lateral cochlear wall in young and aged mice were isolated. The mRNA and protein expression levels of MIF were determined using reverse transcription­quantitative polymerase chain reaction analysis, and western blotting, respectively. MIF expression was knocked down in vitro and in vivo using small interfering RNA. Cell viability was determined using an MTT assay and cell apoptosis was determined using flow cytometry analysis. The hearing ability was assessed through the auditory brain stem response in vivo. The results of the current study demonstrated that the expression of MIF was significantly downregulated in aged mice compared with in young mice. Furthermore, the viability of PVM/Ms in aged mice was significantly decreased and the number of apoptotic PVM/Ms was significantly increased compared with that in young mice. Further studies demonstrated that the MIF knockdown accentuated hearing loss in young mice as compared with the scramble control group. In addition, the MIF knockdown in PVM/Ms significantly inhibited cell viability and lead to a significant increase in the apoptotic cell number as compared with the control group. In summary, these results revealed that the MIF knockdown significantly accentuates hearing loss in young mice in vivo, and significantly inhibits the viability and induces the apoptosis of PVM/Ms in vitro. Thus, the results of the present study may provide a novel potential therapeutic approach and prevention method for presbycusis.


Assuntos
Apoptose , Técnicas de Silenciamento de Genes , Fatores Inibidores da Migração de Macrófagos/genética , Macrófagos/citologia , Melanócitos/citologia , Envelhecimento/metabolismo , Animais , Contagem de Células , Sobrevivência Celular , Cóclea/metabolismo , Regulação para Baixo , Perda Auditiva/metabolismo , Perda Auditiva/patologia , Fatores Inibidores da Migração de Macrófagos/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Melanócitos/metabolismo , Camundongos Endogâmicos C57BL
14.
Neuroscience ; 360: 220-229, 2017 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-28694172

RESUMO

Macrophage migration inhibitory factor (MIF) plays an important role in hearing function; however, the underlying mechanism remains indistinct. PVM/Ms from the stria vascularis of lateral wall of cochlea in young and aged mice were isolated, and the mRNA and protein expression levels were detected. MIF was knocked down or overexpresssed in vitro, and transfection was performed in vivo. Cell viability and apoptosis were determined by MTT assay and flow cytometry analysis, respectively. The hearing ability was tested by the auditory brain stem response. The results showed that MIF expression was significantly downregulated in aged mice. In aged mice, the viability of PVM/Ms significantly decreased, but the apoptotic number markedly increased. MIF knockdown in PVM/Ms in vitro significantly inhibited cell viability and induced cell apoptosis, but MIF overexpression showed contrasting results. Further studies showed that MIF knockdown in young mice resulted in serious hearing loss, but MIF overexpression in aged mice restored the hearing. Si-MIF inhibited the viability and induced apoptosis of PVM/Ms from young mice, whereas Ad-MIF induced the viability and inhibited apoptosis of PVM/Ms from aged mice. Moreover, MIF effectively altered the expression levels of CDK1, BRAF, p-ERK1/2, p-PI3K, and p-Akt. Furthermore, ERK inhibitor PD98059 or PI3K inhibitor LY294002 significantly reversed the effects of Si-MIF on PVM/Ms from young mice, whereas ERK activator EGF or PI3K activator IGF significantly reversed the effects of Ad-MIF on PVM/Ms from aged mice. Taken together, MIF mediates the viability and apoptosis of PVM/Ms, at least partially, through MAPK and/or PI3K/Akt pathway.


Assuntos
Apoptose/fisiologia , Perda Auditiva/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Sobrevivência Celular/fisiologia , Fatores Inibidores da Migração de Macrófagos/genética , Camundongos Endogâmicos C57BL , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Transdução de Sinais
15.
Hear Res ; 313: 38-46, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24780131

RESUMO

Normal microvessel structure and function in the cochlea is essential for maintaining the ionic and metabolic homeostasis required for hearing function. Abnormal cochlear microcirculation has long been considered an etiologic factor in hearing disorders. A better understanding of cochlear blood flow (CoBF) will enable more effective amelioration of hearing disorders that result from aberrant blood flow. However, establishing the direct relationship between CoBF and other cellular events in the lateral wall and response to physio-pathological stress remains a challenge due to the lack of feasible interrogation methods and difficulty in accessing the inner ear. Here we report on new methods for studying the CoBF in a mouse model using a thin or open vessel-window in combination with fluorescence intra-vital microscopy (IVM). An open vessel-window enables investigation of vascular cell biology and blood flow permeability, including pericyte (PC) contractility, bone marrow cell migration, and endothelial barrier leakage, in wild type and fluorescent protein-labeled transgenic mouse models with high spatial and temporal resolution. Alternatively, the thin vessel-window method minimizes disruption of the homeostatic balance in the lateral wall and enables study CoBF under relatively intact physiological conditions. A thin vessel-window method can also be used for time-based studies of physiological and pathological processes. Although the small size of the mouse cochlea makes surgery difficult, the methods are sufficiently developed for studying the structural and functional changes in CoBF under normal and pathological conditions.


Assuntos
Cóclea/irrigação sanguínea , Microscopia de Fluorescência/métodos , Microvasos/fisiologia , Estimulação Acústica , Animais , Velocidade do Fluxo Sanguíneo , Transplante de Medula Óssea , Permeabilidade Capilar , Rastreamento de Células , Dextranos/administração & dosagem , Fluoresceína-5-Isotiocianato/administração & dosagem , Fluoresceína-5-Isotiocianato/análogos & derivados , Corantes Fluorescentes/administração & dosagem , Infusões Intravenosas , Proteínas Luminescentes/biossíntese , Proteínas Luminescentes/genética , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Microcirculação , Microvasos/metabolismo , Microvasos/cirurgia , Modelos Animais , Fluxo Sanguíneo Regional , Fatores de Tempo
16.
J Assoc Res Otolaryngol ; 14(2): 175-85, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23247886

RESUMO

The integrity of the fluid-blood barrier in the stria vascularis is critical for maintaining inner ear homeostasis, especially for sustaining the endocochlear potential, an essential driving force for hearing function. However, the mechanisms that control intrastrial fluid-blood barrier permeability remain largely unknown. At the cellular level, the intrastrial fluid-blood barrier comprises cochlear microvascular endothelial cells connected to each other by tight junctions (TJs), an underlying basement membrane, and a second line of support consisting of cochlear pericytes and perivascular resident macrophage-type melanocytes. In this study, we use a newly established primary cell culture-based in vitro model to show that endothelial cells, pericytes, and perivascular resident macrophage-type melanocytes interact to control intrastrial fluid-blood barrier permeability. When the endothelial cell monolayer was treated with pericyte--or perivascular resident macrophage-type melanocyte--conditioned media, the permeability of the endothelial cell monolayer was significantly reduced relative to an untreated endothelial cell monolayer. Further study has shown the pericytes and perivascular resident macrophage-type melanocytes to regulate TJ expression in the endothelial cell monolayer. The new cell culture-based in vitro model offers a unique opportunity to obtain information on the organ-specific characteristics of the cochlear blood/tissue barrier. Our finding demonstrates the importance of signaling among pericytes, endothelial cells, and perivascular resident macrophage-type melanocytes to the integrity of the intrastrial fluid-blood barrier.


Assuntos
Comunicação Celular/fisiologia , Permeabilidade da Membrana Celular/fisiologia , Cóclea/irrigação sanguínea , Endotélio Vascular/citologia , Macrófagos/citologia , Melanócitos/citologia , Pericitos/citologia , Animais , Antígenos CD/fisiologia , Caderinas/fisiologia , Linhagem Celular , Células Cultivadas , Cóclea/citologia , Cóclea/fisiologia , Endotélio Vascular/fisiologia , Audição/fisiologia , Técnicas In Vitro , Macrófagos/fisiologia , Melanócitos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Pericitos/fisiologia , Transdução de Sinais/fisiologia , Proteínas de Junções Íntimas/fisiologia , Proteína da Zônula de Oclusão-1/fisiologia
17.
J Assoc Res Otolaryngol ; 14(5): 635-43, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23821347

RESUMO

A large number of perivascular cells expressing both macrophage and melanocyte characteristics (named perivascular-resident macrophage-like melanocytes, PVM/Ms), previously found in the intra-strial fluid-blood barrier, are also found in the blood-labyrinth barrier area of the vestibular system in normal adult cochlea, including in the three ampullae of the semicircular canals (posterior, superior, and horizontal), utricle, and saccule. The cells were identified as PVM/Ms, positive for the macrophage and melanocyte marker proteins F4/80 and GSTα4. Similar to PVM/Ms present in the stria vascularis, the PVM/Ms in the vestibular system are closely associated with microvessels and structurally intertwined with endothelial cells and pericytes, with a density in normal (unstimulated) utricle of 225 ± 43/mm(2); saccule 191 ± 25/mm(2); horizontal ampullae 212 ± 36/mm(2); anterior ampullae 238 ± 36/mm(2); and posterior ampullae 223 ± 64/mm(2). Injection of bacterial lipopolysaccharide into the middle ear through the tympanic membrane causes the PVM/Ms to activate and arrange in an irregular pattern along capillary walls in all regions within a 48-h period. The inflammatory response significantly increases vascular permeability and leakage. The results underscore the morphological complexity of the blood barrier in the vestibular system, with its surrounding basal lamina, pericytes, as well as second line of defense in PVM/Ms. PVM/Ms may be important to maintain blood barrier integrity and initiating local inflammatory response in the vestibular system.


Assuntos
Macrófagos/imunologia , Melanócitos/imunologia , Otite/imunologia , Vestíbulo do Labirinto/citologia , Vestíbulo do Labirinto/imunologia , Animais , Capilares/citologia , Capilares/imunologia , Permeabilidade Capilar/imunologia , Contagem de Células , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Otite/induzido quimicamente , Vestíbulo do Labirinto/irrigação sanguínea
18.
Nat Protoc ; 8(4): 709-20, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23493068

RESUMO

This protocol describes a growth medium-based approach for obtaining cochlear endothelial cells (ECs), pericytes (PCs) and perivascular resident macrophage-like melanocytes (PVM/Ms) from the stria vascularis of mice aged between P10 and P15 (P, postnatal day). The procedure does not involve mechanical or enzymatic digestion of the sample tissue. Explants of stria vascularis, 'mini-chips', are selectively cultured in growth medium, and primary cell lines are obtained in 7-10 d. The method is simple and reliable, and it provides high-quality ECs, PVM/Ms and PCs with a purity >90% after two passages. This protocol is suitable for producing primary culture cells from organs and tissues of small volume and high anatomical complexity, such as the inner ear capillaries. The highly purified primary cell lines enable cell culture-based in vitro modeling of cell-cell interactions, barrier control function and drug action.


Assuntos
Técnicas de Cultura de Células , Células Endoteliais/citologia , Macrófagos/citologia , Melanócitos/citologia , Pericitos/citologia , Estria Vascular/citologia , Animais , Linhagem Celular , Separação Celular/métodos , Criopreservação/métodos , Meios de Cultura , Orelha , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL
19.
Lin Chuang Er Bi Yan Hou Tou Jing Wai Ke Za Zhi ; 22(22): 1026-7, 1031, 2008 Nov.
Artigo em Zh | MEDLINE | ID: mdl-19266817

RESUMO

OBJECTIVE: To survey the etiology of sensorineural hearing loss with unknown reason and the incidence of the mutation of SLC26A4 2168A > G in Henan province. METHOD: The evaluation of hearing loss, etiologic survey, the molecular genetic analysis and temporal bone CT examination for genes common to hereditary hearing disorders were performed in 95 hearing-impaired patients in Henan province. RESULT: In the deafness group, the incidence of large vestibular aqueduct syndrome (LVAS) which correlates with SLC26A4 2168A > G is 6.32%. The incidence of the gene diagnosis conformed to the clinical one is 83.3%. CONCLUSION: There is a high incidence of SLC26A4 2168 A > G mutation in sensorineural hearing loss with unknown reason. Molecular genetic screening for these mutations and genetic counseling are effective methods to prevent the occurrence of hereditary hearing loss.


Assuntos
Perda Auditiva Neurossensorial/genética , Proteínas de Membrana Transportadoras/genética , Mutação , Sequência de Bases , China/epidemiologia , Testes Genéticos , Perda Auditiva Neurossensorial/epidemiologia , Humanos , Transportadores de Sulfato , Aqueduto Vestibular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA