Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 708
Filtrar
1.
Hear Res ; 447: 109008, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38636186

RESUMO

The auditory cortex is the source of descending connections providing contextual feedback for auditory signal processing at almost all levels of the lemniscal auditory pathway. Such feedback is essential for cognitive processing. It is likely that corticofugal pathways are degraded with aging, becoming important players in age-related hearing loss and, by extension, in cognitive decline. We are testing the hypothesis that surface, epidural stimulation of the auditory cortex during aging may regulate the activity of corticofugal pathways, resulting in modulation of central and peripheral traits of auditory aging. Increased auditory thresholds during ongoing age-related hearing loss in the rat are attenuated after two weeks of epidural stimulation with direct current applied to the surface of the auditory cortex for two weeks in alternate days (Fernández del Campo et al., 2024). Here we report that the same cortical electrical stimulation protocol induces structural and cytochemical changes in the aging cochlea and auditory brainstem, which may underlie recovery of age-degraded auditory sensitivity. Specifically, we found that in 18 month-old rats after two weeks of cortical electrical stimulation there is, relative to age-matched non-stimulated rats: a) a larger number of choline acetyltransferase immunoreactive neuronal cell body profiles in the ventral nucleus of the trapezoid body, originating the medial olivocochlear system.; b) a reduction of age-related dystrophic changes in the stria vascularis; c) diminished immunoreactivity for the pro-inflammatory cytokine TNFα in the stria vascularis and spiral ligament. d) diminished immunoreactivity for Iba1 and changes in the morphology of Iba1 immunoreactive cells in the lateral wall, suggesting reduced activation of macrophage/microglia; d) Increased immunoreactivity levels for calretinin in spiral ganglion neurons, suggesting excitability modulation by corticofugal stimulation. Altogether, these findings support that non-invasive neuromodulation of the auditory cortex during aging preserves the cochlear efferent system and ameliorates cochlear aging traits, including stria vascularis dystrophy, dysregulated inflammation and altered excitability in primary auditory neurons.


Assuntos
Envelhecimento , Córtex Auditivo , Vias Auditivas , Cóclea , Estimulação Elétrica , Presbiacusia , Animais , Córtex Auditivo/metabolismo , Córtex Auditivo/fisiopatologia , Cóclea/inervação , Cóclea/metabolismo , Cóclea/fisiopatologia , Cóclea/patologia , Presbiacusia/fisiopatologia , Presbiacusia/metabolismo , Presbiacusia/patologia , Vias Auditivas/fisiopatologia , Vias Auditivas/metabolismo , Masculino , Envelhecimento/patologia , Envelhecimento/metabolismo , Modelos Animais de Doenças , Fatores Etários , Neurônios Eferentes/metabolismo , Microglia/metabolismo , Microglia/patologia , Limiar Auditivo , Colina O-Acetiltransferase/metabolismo , Núcleo Olivar/metabolismo , Potenciais Evocados Auditivos do Tronco Encefálico , Audição , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/metabolismo , Proteínas de Ligação ao Cálcio , Proteínas dos Microfilamentos
2.
Neurosci Bull ; 40(2): 255-267, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37391607

RESUMO

Macrophages are essential components of the innate immune system and constitute a non-specific first line of host defense against pathogens and inflammation. Mitochondria regulate macrophage activation and innate immune responses in various inflammatory diseases, including cochlear inflammation. The distribution, number, and morphological characteristics of cochlear macrophages change significantly across different inner ear regions under various pathological conditions, including noise exposure, ototoxicity, and age-related degeneration. However, the exact mechanism underlying the role of mitochondria in macrophages in auditory function remains unclear. Here, we summarize the major factors and mitochondrial signaling pathways (e.g., metabolism, mitochondrial reactive oxygen species, mitochondrial DNA, and the inflammasome) that influence macrophage activation in the innate immune response. In particular, we focus on the properties of cochlear macrophages, activated signaling pathways, and the secretion of inflammatory cytokines after acoustic injury. We hope this review will provide new perspectives and a basis for future research on cochlear inflammation.


Assuntos
Imunidade Inata , Macrófagos , Humanos , Cóclea/metabolismo , Cóclea/patologia , Inflamação/metabolismo , Mitocôndrias
3.
Hear Res ; 441: 108916, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38103445

RESUMO

Flat epithelium (FE) is a condition characterized by the loss of both hair cells (HCs) and supporting cells and the transformation of the organ of Corti into a simple flat or cuboidal epithelium, which can occur after severe cochlear insults. The transcription factors Gfi1, Atoh1, Pou4f3, and Six1 (GAPS) play key roles in HC differentiation and survival in normal ears. Previous work using a single transcription factor, Atoh1, to induce HC regeneration in mature ears in vivo usually produced very few cells and failed to produce HCs in severely damaged organs of Corti, especially those with FE. Studies in vitro suggested combinations of transcription factors may be more effective than any single factor, thus the current study aims to examine the effect of co-overexpressing GAPS genes in deafened mature guinea pig cochleae with FE. Deafening was achieved through the infusion of neomycin into the perilymph, leading to the formation of FE and substantial degeneration of nerve fibers. Seven days post neomycin treatment, adenovirus vectors carrying GAPS were injected into the scala media and successfully expressed in the FE. One or two months following GAPS inoculation, cells expressing Myosin VIIa were observed in regions under the FE (located at the scala tympani side of the basilar membrane), rather than within the FE. The number of cells, which we define as induced HCs (iHCs), was not significantly different between one and two months, but the larger N at two months made it more apparent that there were significantly more iHCs in GAPS treated animals than in controls. Additionally, qualitative observations indicated that ears with GAPS gene expression in the FE had more nerve fibers than FE without the treatment. In summary, our results showed that co-overexpression of GAPS enhances the potential for HC regeneration in a severe lesion model of FE.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos , Fatores de Transcrição , Animais , Cobaias , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Células Ciliadas Auditivas/patologia , Epitélio/metabolismo , Cóclea/metabolismo , Neomicina
4.
Gene Ther ; 31(3-4): 154-164, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38097651

RESUMO

The adeno-associated virus (AAV) gene therapy has been widely applied to mouse models for deafness. But, AAVs could transduce non-targeted organs after inner ear delivery due to their low cell-type specificity. This study compares transgene expression and biodistribution of AAV1, AAV2, Anc80L65, AAV9, AAV-PHP.B, and AAV-PHP.eB after round window membrane (RWM) injection in neonatal mice. The highest virus concentration was detected in the injected cochlea. AAV2, Anc80L65, AAV9, AAV-PHP.B, and AAV-PHP.eB transduced both inner hair cells (IHCs) and outer hair cells (OHCs) with high efficiency, while AAV1 transduced IHCs with high efficiency but OHCs with low efficiency. All AAV subtypes finitely transduced contralateral inner ear, brain, heart, and liver compared with the injected cochlea. In most brain regions, the enhanced green fluorescent protein (eGFP) expression of AAV1 and AAV2 was lower than that of other four subtypes. We suggested the cochlear aqueduct might be one of routes for vectors instantaneously infiltrating into the brain from the cochlea through a dye tracking test. In summary, our results provide available data for further investigating the biodistribution of vectors through local inner ear injection and afford a reference for selecting AAV serotypes for gene therapy toward deafness.


Assuntos
Surdez , Vetores Genéticos , Animais , Camundongos , Distribuição Tecidual , Vetores Genéticos/genética , Cóclea/metabolismo , Terapia Genética/métodos , Surdez/metabolismo , Dependovirus/genética , Dependovirus/metabolismo , Transdução Genética
5.
J Neurosci ; 43(27): 5057-5075, 2023 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-37268417

RESUMO

Age-related hearing loss, or presbyacusis, is a common degenerative disorder affecting communication and quality of life for millions of older adults. Multiple pathophysiologic manifestations, along with many cellular and molecular alterations, have been linked to presbyacusis; however, the initial events and causal factors have not been clearly established. Comparisons of the transcriptome in the lateral wall (LW) with other cochlear regions in a mouse model (of both sexes) of "normal" age-related hearing loss revealed that early pathophysiological alterations in the stria vascularis (SV) are associated with increased macrophage activation and a molecular signature indicative of inflammaging, a common form of immune dysfunction. Structure-function correlation analyses in mice across the lifespan showed that the age-dependent increase in macrophage activation in the stria vascularis is associated with a decline in auditory sensitivity. High-resolution imaging analysis of macrophage activation in middle-aged and aged mouse and human cochleas, along with transcriptomic analysis of age-dependent changes in mouse cochlear macrophage gene expression, support the hypothesis that aberrant macrophage activity is an important contributor to age-dependent strial dysfunction, cochlear pathology, and hearing loss. Thus, this study highlights the SV as a primary site of age-related cochlear degeneration and aberrant macrophage activity and dysregulation of the immune system as early indicators of age-related cochlear pathology and hearing loss. Importantly, novel new imaging methods described here now provide a means to analyze human temporal bones in a way that had not previously been feasible and thereby represent a significant new tool for otopathological evaluation.SIGNIFICANCE STATEMENT Age-related hearing loss is a common neurodegenerative disorder affecting communication and quality of life. Current interventions (primarily hearing aids and cochlear implants) offer imperfect and often unsuccessful therapeutic outcomes. Identification of early pathology and causal factors is crucial for the development of new treatments and early diagnostic tests. Here, we find that the SV, a nonsensory component of the cochlea, is an early site of structural and functional pathology in mice and humans that is characterized by aberrant immune cell activity. We also establish a new technique for evaluating cochleas from human temporal bones, an important but understudied area of research because of a lack of well-preserved human specimens and difficult tissue preparation and processing approaches.


Assuntos
Surdez , Presbiacusia , Masculino , Pessoa de Meia-Idade , Feminino , Humanos , Animais , Camundongos , Idoso , Estria Vascular/patologia , Qualidade de Vida , Cóclea/metabolismo , Presbiacusia/patologia , Surdez/patologia , Macrófagos , Inflamação/metabolismo
6.
J Cancer Res Ther ; 19(Supplement): S0, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37147955

RESUMO

Introduction: Chemotherapeutic agents can have both serious side effects and ototoxicity, which can be caused by direct toxic effects or by metabolic derangement by the agents. Cabazitaxel (CBZ) is a next-generation semi-synthetic taxane derivative that is effective in both preclinical models of human tumors that are sensitive or resistant to chemotherapy and in patients suffering from progressive prostate cancer despite docetaxel treatment. The primary aim of this study is to investigate the ototoxicity of CBZ in a rat model. Materials and Methods: : A total of 24 adult male Wistar-Albino rats were equally and randomly divided into four groups. CBZ (Jevtana, Sanofi-Aventis USA) was intraperitoneally administered to Groups 2, 3, and 4 at doses of 0.5, 1.0, and 1.5 mg/kg/week, respectively, for 4 consecutive weeks; Group 1 received only i.p. saline at the same time. At the end of the study, the animals were sacrificed and their cochlea removed for histopathological examination. Results: : Intraperitoneal administration of CBZ exerted an ototoxic effect on rats, and the histopathological results became worse in a dose-dependent manner (P < 0.05). Conclusion: : Our findings suggest that CBZ may be an ototoxic agent and can damage the cochlea. More clinical studies should be conducted to understand its ototoxicity.


Assuntos
Antineoplásicos , Ototoxicidade , Neoplasias da Próstata , Humanos , Animais , Ratos , Masculino , Antineoplásicos/toxicidade , Antineoplásicos/metabolismo , Ototoxicidade/metabolismo , Ototoxicidade/patologia , Ratos Wistar , Cóclea/metabolismo , Cóclea/patologia , Neoplasias da Próstata/patologia
7.
Cell Signal ; 108: 110732, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37245680

RESUMO

In the cochlea, extracellular ATP (eATP) plays an important role in both physiological and pathological processes, but its role in the hypoxic cochlea remains unclear. The present study aims to investigate the relationship between eATP and hypoxic marginal cells (MCs) in the stria vascularis in cochlea. Combining various methodologies, we found that eATP accelerates cell death and decreases tight junction protein zonula occludens-1 (ZO-1) in hypoxic MCs. Flow cytometry and western blot analyses revealed an increase in apoptosis levels and suppression of autophagy, indicating that eATP causes additional cell death by increasing the apoptosis of hypoxic MCs. Given that autophagy inhibits apoptosis to protect MCs under hypoxia, apoptosis is probably enchanced by suppressing autophagy. Interleukin-33(IL-33)/suppression of tumorigenicity-2(ST-2)/matrix metalloprotein 9(MMP9) pathway activation was also observed during the process. Further experiments involving the use of additional IL-33 protein and an MMP9 inhibitor indicated that this pathway is responsible for the damage to the ZO-1 protein in hypoxic MCs. Our study revealed an adverse effect of eATP on the survival and ZO-1 protein expression of hypoxic MCs, as well as the underlying mechanism.


Assuntos
Interleucina-33 , Metaloproteinase 9 da Matriz , Ratos , Animais , Animais Recém-Nascidos , Metaloproteinase 9 da Matriz/metabolismo , Interleucina-33/metabolismo , Junções Íntimas/metabolismo , Proteína da Zônula de Oclusão-1/metabolismo , Cóclea/metabolismo , Morte Celular , Hipóxia/metabolismo , Trifosfato de Adenosina/metabolismo
8.
Cell Mol Biol Lett ; 28(1): 23, 2023 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-36959542

RESUMO

BACKGROUND: Phosphatidylserine is translocated to the inner leaflet of the phospholipid bilayer membrane by the flippase function of type IV P-tape ATPase (P4-ATPase), which is critical to maintain cellular stability and homeostasis. Transmembrane protein 30A (TMEM30A) is the ß-subunit of P4-ATPase. Loss of P4-ATPase function causes sensorineural hearing loss and visual dysfunction in human. However, the function of TMEM30A in the auditory system is unclear. METHODS: P4-ATPase subtype expression in the cochlea was detected by immunofluorescence staining and quantitative real-time polymerase chain reaction (qRT-PCR) at different developmental stages. Hair cell specific TMEM30A knockout mice and wild-type littermates were used for the following functional and morphological analysis. Auditory function was evaluated by auditory brainstem response. We investigated hair cell and stereocilia morphological changes by immunofluorescence staining. Scanning electron microscopy was applied to observe the stereocilia ultrastructure. Differentially expressed transcriptomes were analyzed based on RNA-sequencing data from knockout and wild-type mouse cochleae. Differentially expressed genes were verified by qRT-PCR. RESULTS: TMEM30A and subtypes of P4-ATPase are expressed in the mouse cochlea in a temporal-dependent pattern. Deletion of TMEM30A in hair cells impaired hearing onset due to progressive hair cell loss. The disrupted kinocilia placement and irregular distribution of spectrin-α in cuticular plate indicated the hair cell planar polarity disruption in TMEM30A deletion hair cells. Hair cell degeneration begins at P7 and finishes around P14. Transcriptional analysis indicates that the focal adhesion pathway and stereocilium tip-related genes changed dramatically. Without the TMEM30A chaperone, excessive ATP8A2 accumulated in the cytoplasm, leading to overwhelming endoplasmic reticulum stress, which eventually contributed to hair cell death. CONCLUSIONS: Deletion of TMEM30A led to disrupted planar polarity and stereocilia bundles, and finally led to hair cell loss and auditory dysfunction. TMEM30A is essential for hair cell polarity maintenance and membrane homeostasis. Our study highlights a pivotal role of TMEM30A in the postnatal development of hair cells and reveals the possible mechanisms underlying P4-ATPase-related genetic hearing loss.


Assuntos
Polaridade Celular , Cóclea , Camundongos , Animais , Humanos , Camundongos Knockout , Cóclea/metabolismo , Adenosina Trifosfatases/metabolismo , Proteínas de Membrana/metabolismo
9.
J Neurosci ; 43(12): 2075-2089, 2023 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-36810227

RESUMO

Resident cochlear macrophages rapidly migrate into the inner hair cell synaptic region and directly contact the damaged synaptic connections after noise-induced synaptopathy. Eventually, such damaged synapses are spontaneously repaired, but the precise role of macrophages in synaptic degeneration and repair remains unknown. To address this, cochlear macrophages were eliminated using colony stimulating factor 1 receptor (CSF1R) inhibitor, PLX5622. Sustained treatment with PLX5622 in CX3CR1 GFP/+ mice of both sexes led to robust elimination of resident macrophages (∼94%) without significant adverse effects on peripheral leukocytes, cochlear function, and structure. At 1 day (d) post noise exposure of 93 or 90 dB SPL for 2 hours, the degree of hearing loss and synapse loss were comparable in the presence and absence of macrophages. At 30 d after exposure, damaged synapses appeared repaired in the presence of macrophages. However, in the absence of macrophages, such synaptic repair was significantly reduced. Remarkably, on cessation of PLX5622 treatment, macrophages repopulated the cochlea, leading to enhanced synaptic repair. Elevated auditory brainstem response thresholds and reduced auditory brainstem response Peak 1 amplitudes showed limited recovery in the absence of macrophages but recovered similarly with resident and repopulated macrophages. Cochlear neuron loss was augmented in the absence of macrophages but showed preservation with resident and repopulated macrophages after noise exposure. While the central auditory effects of PLX5622 treatment and microglia depletion remain to be investigated, these data demonstrate that macrophages do not affect synaptic degeneration but are necessary and sufficient to restore cochlear synapses and function after noise-induced synaptopathy.SIGNIFICANCE STATEMENT The synaptic connections between cochlear inner hair cells and spiral ganglion neurons can be lost because of noise over exposure or biological aging. This loss may represent the most common causes of sensorineural hearing loss also known as hidden hearing loss. Synaptic loss results in degradation of auditory information, leading to difficulty in listening in noisy environments and other auditory perceptual disorders. We demonstrate that resident macrophages of the cochlea are necessary and sufficient to restore synapses and function following synaptopathic noise exposure. Our work reveals a novel role for innate-immune cells, such as macrophages in synaptic repair, that could be harnessed to regenerate lost ribbon synapses in noise- or age-linked cochlear synaptopathy, hidden hearing loss, and associated perceptual anomalies.


Assuntos
Células Ciliadas Auditivas Internas , Perda Auditiva Provocada por Ruído , Masculino , Feminino , Animais , Camundongos , Células Ciliadas Auditivas Internas/fisiologia , Estimulação Acústica/efeitos adversos , Limiar Auditivo/fisiologia , Cóclea/metabolismo , Sinapses/fisiologia , Potenciais Evocados Auditivos do Tronco Encefálico/fisiologia , Macrófagos/metabolismo
10.
Biochem Pharmacol ; 209: 115440, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36720354

RESUMO

Cisplatin is commonly used to treat cancers and is associated with a significant risk of irreversible sensorineural hearing loss. However, no effective preventive strategies are available for cisplatin-induced HL. Therefore, significant efforts have been made to discover new drugs protecting cochlear hair cells from cisplatin-induced damage. We found that a new phytochemical, aucubin, attenuated cisplatin-induced apoptosis, the production of reactive oxygen species, and mitochondrial dysfunction in House Ear Institute Organ of Corti 1 cells and cochlear hair cells. Moreover, aucubin attenuated cisplatin-induced sensorineural hearing loss and hair cells loss in vivo. Furthermore, RNA sequencing analysis revealed that the otoprotective effects of aucubin were mainly mediated by increased STAT3 phosphorylation via the PI3K/AKT pathway. Inhibition of the STAT3 signaling pathway with the inhibitor S3I-201 or siRNA disrupted the protective effects of aucubin on cisplatin-induced apoptosis. In conclusion, we identified an otoprotective effect of aucubin. Therefore, aucubin could be used to prevent cisplatin-induced ototoxicity.


Assuntos
Antineoplásicos , Perda Auditiva Neurossensorial , Perda Auditiva , Ototoxicidade , Camundongos , Animais , Cisplatino/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Perda Auditiva/induzido quimicamente , Perda Auditiva/tratamento farmacológico , Perda Auditiva/prevenção & controle , Ototoxicidade/metabolismo , Cóclea/metabolismo , Células Ciliadas Auditivas , Apoptose , Espécies Reativas de Oxigênio/metabolismo , Perda Auditiva Neurossensorial/tratamento farmacológico , Perda Auditiva Neurossensorial/metabolismo , Antineoplásicos/farmacologia
11.
Cell Tissue Res ; 391(1): 43-54, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36287265

RESUMO

Damage-associated molecular pattern molecules (DAMPs) play a critical role in mediating cochlear cell death, which leads to noise-induced hearing loss (NIHL). High-mobility group box 1 (HMGB1), a prototypical DAMP released from cells, has been extensively studied in the context of various diseases. However, whether extracellular HMGB1 contributes to cochlear pathogenesis in NIHL and the potential signals initiating HMGB1 release from cochlear cells are not well understood. Here, through the transfection of the adeno-associated virus with HMGB1-HA-tag, we first investigated early cytoplasmic accumulation of HMGB1 in cochlear hair cells after noise exposure. We found that the cochlear administration of HMGB1-neutralizing antibody immediately after noise exposure significantly alleviated hearing loss and outer hair cells (OHCs) death induced by noise exposure. In addition, activation of signal transducer and activators of transcription 1 (STAT1) and cellular hyperacetylation were verified as potential canonical initiators of HMGB1 cytoplasmic accumulation. These findings reveal the adverse effects of extracellular HMGB1 on the cochlea and the potential signaling events mediating HMGB1 release in hair cells, indicating multiple potential pharmacotherapeutic targets for NIHL.


Assuntos
Cóclea , Proteína HMGB1 , Perda Auditiva Provocada por Ruído , Ruído , Animais , Camundongos , Cóclea/metabolismo , Cóclea/patologia , Citoplasma/metabolismo , Células Ciliadas Auditivas Externas/metabolismo , Perda Auditiva Provocada por Ruído/etiologia , Perda Auditiva Provocada por Ruído/metabolismo , Perda Auditiva Provocada por Ruído/patologia , Proteína HMGB1/metabolismo , Ruído/efeitos adversos
12.
Int J Mol Sci ; 23(23)2022 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-36499200

RESUMO

The mammalian cochlea is the sensory organ of hearing with a delicate, highly organised structure that supports unique operating mechanisms. ATP release from the secretory tissues of the cochlear lateral wall (stria vascularis) triggers numerous physiological responses by activating P2 receptors in sensory, supporting and neural tissues. Two families of P2 receptors, ATP-gated ion channels (P2X receptors) and G protein-coupled P2Y receptors, activate intracellular signalling pathways that regulate cochlear development, homeostasis, sensory transduction, auditory neurotransmission and response to stress. Of particular interest is a purinergic hearing adaptation, which reflects the critical role of the P2X2 receptor in adaptive cochlear response to elevated sound levels. Other P2 receptors are involved in the maturation of neural processes and frequency selectivity refinement in the developing cochlea. Extracellular ATP signalling is regulated by a family of surface-located enzymes collectively known as "ectonucleotidases" that hydrolyse ATP to adenosine. Adenosine is a constitutive cell metabolite with an established role in tissue protection and regeneration. The differential activation of A1 and A2A adenosine receptors defines the cochlear response to injury caused by oxidative stress, inflammation, and activation of apoptotic pathways. A1 receptor agonism, A2A receptor antagonism, and increasing adenosine levels in cochlear fluids all represent promising therapeutic tools for cochlear rescue from injury and prevention of hearing loss.


Assuntos
Trifosfato de Adenosina , Cóclea , Animais , Trifosfato de Adenosina/metabolismo , Cóclea/metabolismo , Audição/fisiologia , Transdução de Sinais/fisiologia , Adenosina/metabolismo , Mamíferos/metabolismo
13.
Zhongguo Ying Yong Sheng Li Xue Za Zhi ; 38(4): 348-355, 2022 Jul.
Artigo em Chinês | MEDLINE | ID: mdl-36414560

RESUMO

Objective: To investigate the role of Cav1.2 and its possible mechanism in the apoptosis of cochlear spiral ganglion neurons(SGNs) induced by cisplatin (CDDP) in C57BL/6J mice. Methods: Animal experiment: 8-week-old male C57BL/6J mice were randomly divided into the following two groups (10 mice/group) : normal saline group (Control group) and Cisplatin group (Cisplatin group). The Control group received daily intraperitoneal injections of normal saline, Cisplatin group was injected with cisplatin intraperitoneally at a dose of 3 mg/kg at the first 4 days of each cycle, and normal saline was injected daily at the last 10 days,repeat for 3 cycles. After administration, auditory threshold was detected by auditory brainstem response (ABR). Blood samples were collected from inner canthus of mice, and cochlea was cut off from neck. SOD and MDA kits were used to detect SOD activity and MDA content in serum and cochlea tissues. The expressions of apoptosis proteins in cochlear tissues were detected by Western blot. Morphological changes of spiral ganglion in mouse cochlea were observed by hematoxylin-eosin (HE) staining. TUNEL staining was used to observe the apoptosis of SGNs in cochlea of mice. The distribution and expression of Cav1.2 in SGNs of cochlea were observed by immunofluorescence. Cell experiment: Primary cultured SGNs were randomly divided into: control group (Control), solvent group (DMSO), Cav1.2 blocker group (N), cisplatin group, cisplatin and Cav1.2 blocker co-incubation group (Cisplatin+N). 5 µmol/L cisplatin was selected to treat SGNs based on the results of CCK8. Western blot was used to detect the protein expressions of Cav1.2.and apoptotic proteins. Hoechst33342 staining was used to observe the apoptosis of each group. Flow cytometry was used to detect the apoptosis rate of each group. Mitochondrial superoxide indicator (MitoSOXTM-Red) was used to detect the ROS release of mitochondria. Results: Animal experiments: Compared to the Control group, the hearing threshold was increased in Cisplatin group (P<0.01), the content of MDA in serum and cochlea tissues, apoptosis protein Cleaved caspase-3, Bax protein level, TUNEL positive rate, Cav1.2 protein expression level were increased significantly (P<0.05, P<0.01); the activity of SOD in serum and cochlear tissue, anti-apoptotic protein bcl-2 protein level and SGCs density in cochlear tissue were decreased significantly (P<0.05, P<0.01). Cell tests: Compared with the Control group, the expression of Cav1.2, apoptosis rate, Cleaved caspase-3, Bax protein level, intracellular calcium ion concentration, and ROS release were increased significantly only in Cisplatin group (P<0.05, P<0.01). The levels of bcl-2 protein and mitochondrial membrane potential were decreased significantly (P<0.01). Cav1.2 blockers could partially reverse the above changes (P<0.05). Conclusion: Cisplatin may increase intracellular Ca2+ concentration through up-regulation of Cav1.2, and then damage mitochondria, causing oxidative stress injury of SGNs and inducing neuronal apoptosis.


Assuntos
Cisplatino , Gânglio Espiral da Cóclea , Masculino , Camundongos , Animais , Gânglio Espiral da Cóclea/metabolismo , Cisplatino/farmacologia , Cisplatino/metabolismo , Proteína X Associada a bcl-2/metabolismo , Caspase 3/metabolismo , Camundongos Endogâmicos C57BL , Solução Salina , Espécies Reativas de Oxigênio/metabolismo , Cóclea/metabolismo , Apoptose , Neurônios , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Superóxido Dismutase/metabolismo
14.
JCI Insight ; 7(20)2022 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-36278489

RESUMO

Recessive PJVK mutations that cause a deficiency of pejvakin, a protein expressed in both sensory hair cells and first-order neurons of the inner ear, are an important cause of hereditary hearing impairment. Patients with PJVK mutations garner limited benefits from cochlear implantation; thus, alternative biological therapies may be required to address this clinical difficulty. The synthetic adeno-associated viral vector Anc80L65, with its wide tropism and high transduction efficiency in various inner ear cells, may provide a solution. We delivered the PJVK transgene to the inner ear of Pjvk mutant mice using the synthetic Anc80L65 vector. We observed robust exogenous pejvakin expression in the hair cells and neurons of the cochlea and vestibular organs. Subsequent morphologic and audiologic studies demonstrated significant restoration of spiral ganglion neuron density and hair cells in the cochlea, along with partial recovery of sensorineural hearing impairment. In addition, we observed a recovery of vestibular ganglion neurons and balance function to WT levels. Our study demonstrates the utility of Anc80L65-mediated gene delivery in Pjvk mutant mice and provides insights into the potential of gene therapy for PJVK-related inner ear deficits.


Assuntos
Terapia Genética , Perda Auditiva Neurossensorial , Camundongos , Animais , Células Ciliadas Auditivas/metabolismo , Perda Auditiva Neurossensorial/genética , Perda Auditiva Neurossensorial/terapia , Perda Auditiva Neurossensorial/metabolismo , Cóclea/metabolismo , Fenótipo , Proteínas/genética
15.
ACS Chem Neurosci ; 13(16): 2464-2472, 2022 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-35939349

RESUMO

Both exosomes derived from neural progenitor cells (NPCs) can suppress inflammation. Whether exosomes derived from miR-21-transfected NPCs (miR-21-Exo) could be utilized to alleviate hearing loss is investigated. NPCs were transfected with lentiviral vectors overexpressing miR-21, and miR-21-Exo was purified. Morphology and exosome membrane markers were examined with nanoparticle tracking analysis, transmission electron microscopy, and Western blot. After incubation with different concentrations of miR-21-Exo, the viability of RAW 264.7 cells and the relative expressions of miR-21 and IL-10 were determined. The ischemia and reperfusion (I/R) model of C57BL/6 J mice was constructed, and the treatment benefit of miR-21-Exo was revealed by the auditory brainstem response (ABR) test. Immunofluorescence staining of caspase-3 and parvalbumin was used to detect apoptosis hair cells in the cochlea, and Western blot was utilized to detect the relative expressions of P53 and inflammatory cytokines in the cochlea. Isolated exosomes were confirmed by the size of 96 ± 25 nm, single membrane, and positive expression of CD9 and Tsg101. Upregulated miR-21 expression was detected in miR-21-transfected NPCs and miR-21-Exo. miR-21-Exo incubation demonstrated no cytotoxicity but upregulated miR-21 and IL-10 expressions in RAW 264.7 cells. The administration of miR-21-Exo inhibited the increased ABR threshold under 8, 16, and 32 kHz frequencies in cochlea-I/R injury mice and diminished the mean fluorescent intensity of caspase-3/parvalbumin. Moreover, miR-21-Exo treatment increased the IL-10 expression and prevented the increased TNF-α and IL-1ß expressions in the cochlea of I/R mice both in mRNA and protein levels. Inner ear administration of miR-21-Exo effectively improved hearing damage caused by I/R.


Assuntos
Exossomos , Perda Auditiva , Células-Tronco Mesenquimais , MicroRNAs , Traumatismo por Reperfusão , Animais , Caspase 3/metabolismo , Cóclea/metabolismo , Exossomos/metabolismo , Perda Auditiva/etiologia , Perda Auditiva/prevenção & controle , Interleucina-10/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Parvalbuminas/metabolismo , Traumatismo por Reperfusão/complicações
16.
Hum Gene Ther ; 33(13-14): 729-739, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35726398

RESUMO

Adeno-associated virus (AAV)-based gene therapy has been demonstrated to be extremely effective for treating genetic hearing loss over the past several years. However, successful gene therapies for hereditary deafness have not been well-studied in adult mice. To explore the possibility of gene therapy after peripheral auditory maturity, we used AAV8 to express vesicular glutamate transporter 3 (Vglut3) in the cochleae of 5w, 8w, and 20w Vglut3KO mice. Results indicated that AAV8-Vglut3 could mediate the exogenous expression of Vglut3 in all inner hair cells (IHCs). Auditory function was successfully restored, and the hearing threshold remained stable for at least 12 weeks after rescue. Moreover, the results revealed that the number of synaptic ribbons, as well as their morphology, was significantly recovered after gene therapy, potentially indicating the glutamate-dependent plasticity of IHCs. Taken together, our data introduce the possibility of gene therapy in adult mice and advance our knowledge of the role of Vglut3 in presynaptic plasticity.


Assuntos
Células Ciliadas Auditivas Internas , Audição , Animais , Cóclea/metabolismo , Terapia Genética/métodos , Células Ciliadas Auditivas Internas/metabolismo , Camundongos , Camundongos Knockout
17.
PLoS Genet ; 18(6): e1010232, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35727824

RESUMO

Dync1li1, a subunit of cytoplasmic dynein 1, is reported to play important roles in intracellular retrograde transport in many tissues. However, the roles of Dync1li1 in the mammalian cochlea remain uninvestigated. Here we first studied the expression pattern of Dync1li1 in the mouse cochlea and found that Dync1li1 is highly expressed in hair cells (HCs) in both neonatal and adult mice cochlea. Next, we used Dync1li1 knockout (KO) mice to investigate its effects on hearing and found that deletion of Dync1li1 leads to early onset of progressive HC loss via apoptosis and to subsequent hearing loss. Further studies revealed that loss of Dync1li1 destabilizes dynein and alters the normal function of dynein. In addition, Dync1li1 KO results in a thinner Golgi apparatus and the accumulation of LC3+ autophagic vacuoles, which triggers HC apoptosis. We also knocked down Dync1li1 in the OC1 cells and found that the number of autophagosomes were significantly increased while the number of autolysosomes were decreased, which suggested that Dync1li1 knockdown leads to impaired transportation of autophagosomes to lysosomes and therefore the accumulation of autophagosomes results in HC apoptosis. Our findings demonstrate that Dync1li1 plays important roles in HC survival through the regulation of autophagosome transportation.


Assuntos
Autofagossomos , Dineínas do Citoplasma , Células Ciliadas Auditivas , Animais , Apoptose/fisiologia , Autofagossomos/metabolismo , Cóclea/citologia , Cóclea/metabolismo , Dineínas do Citoplasma/metabolismo , Dineínas/metabolismo , Células Ciliadas Auditivas/citologia , Células Ciliadas Auditivas/metabolismo , Camundongos
18.
Protein Pept Lett ; 29(7): 567-573, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35546749

RESUMO

Protein tyrosine phosphatase receptor-type Q (PTPRQ), a member of the type III tyrosine phosphatase receptor (R3 PTPR) family, is composed of three domains, including 18 extracellular fibronectin type III (FN3) repeats, a transmembrane helix, and a cytoplasmic phosphotyrosine phosphatase (PTP) domain. PTPRQ was initially identified as a transcript upregulated in glomerular mesangial cells in a rat model of glomerulonephritis. Subsequently, studies found that PTPRQ has phosphotyrosine phosphatase and phosphatidylinositol phosphatase activities and can regulate cell proliferation, apoptosis, differentiation, and survival. Further in vivo studies showed that PTPRQ is necessary for the maturation of cochlear hair bundles and is considered a potential gene for deafness. In the recent two decades, 21 mutations in PTPRQ have been linked to autosomal recessive hearing loss (DFNB84) and autosomal dominant hearing loss (DFNA73). Recent mutations, deletions, and amplifications of PTPRQ have been observed in many types of cancers, which indicate that PTPRQ might play an essential role in the development of many cancers. In this review, we briefly describe PTPRQ structure and enzyme activity and focus on the correlation between PTPRQ and human disease. A profound understanding of PTPRQ could be helpful in the identification of new therapeutic targets to treat associated diseases.


Assuntos
Cóclea/metabolismo , Perda Auditiva , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/metabolismo , Animais , Cóclea/crescimento & desenvolvimento , Fibronectinas , Perda Auditiva/genética , Humanos , Fosfatidilinositóis , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/metabolismo , Ratos , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/química
19.
Hear Res ; 415: 108427, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-34999290

RESUMO

Spiral ligament fibrocytes of the cochlea play homoeostatic roles in hearing and their degeneration contributes to hearing loss. Culturing fibrocytes in vitro provides a way to evaluate their functional characteristics and study possible therapies for hearing loss. We investigated whether in vivo characteristics of fibrocytes could be recapitulated in vitro by modifying the culture substrates and carried out proof of concept studies for potential transplantation of culture cells into the inner ear. Fibrocytes cultured from 4 to 5-week old CD/1 mice were grown on 2D substrates coated with collagen I, II, V or IX and, after harvesting, onto or into 3D substrates (hydrogels) of collagen I alone or mixed collagen I and II at a 1:1 ratio. We also assessed magnetic nanoparticle (MNP) uptake. Cell counts, immunohistochemical and ultrastructural studies showed that fibrocytes grown on 2D substrates proliferated, formed both small spindle-shaped and large flat cells that avidly took up MNPs. Of the different collagen coatings, only collagen II had an effect, causing a reduced size of the larger cells. On hydrogels, the cells were plump/rounded with extended processes, resembling native cells. They formed networks over the surface and became incorporated into the gel. In all culture formats, the majority co-expressed caldesmon, aquaporin 1, S-100 and sodium potassium ATPase, indicating a mixed or uncharacterised phenotype. Time-course experiments showed a decrease to ∼50% of the starting population by 4d after seeding on collagen I hydrogels, but better survival (∼60%) was found on collagen I + II gels, whilst TEM revealed the presence of apoptotic cells. Cells grown within gels additionally showed necrosis. These results demonstrate that fibrocytes grown in 3D recapitulate in vivo morphology of native fibrocytes, but have poorer survival, compared with 2D. Therefore hydrogel cultures could be used to study fibrocyte function and might also offer avenues for cell-replacement therapies, but need more optimization for therapeutic use. Fibrocyte function could be modified using MNPs in combination, for example, with gene transfection.


Assuntos
Cóclea , Orelha Interna , Animais , Técnicas de Cultura de Células , Cóclea/metabolismo , Orelha Interna/metabolismo , Audição , Camundongos , ATPase Trocadora de Sódio-Potássio/metabolismo
20.
Int J Mol Sci ; 24(1)2022 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-36613734

RESUMO

As noise-induced hearing loss (NIHL) is a leading cause of occupational diseases, there is an urgent need for the development of preventive and therapeutic interventions. To avoid user-compliance-based problems occurring with conventional protection devices, the pharmacological prevention is currently in the focus of hearing research. Noise exposure leads to an increase in reactive oxygen species (ROS) in the cochlea. This way antioxidant agents are a promising option for pharmacological interventions. Previous animal studies reported preventive as well as therapeutic effects of Insulin-like growth factor 1 (IGF-1) in the context of NIHL. Unfortunately, in patients the time point of the noise trauma cannot always be predicted, and additive effects may occur. Therefore, continuous prevention seems to be beneficial. The present study aimed to investigate the preventive potential of continuous administration of low concentrations of IGF-1 to the inner ear in an animal model of NIHL. Guinea pigs were unilaterally implanted with an osmotic minipump. One week after surgery they received noise trauma, inducing a temporary threshold shift. Continuous IGF-1 delivery lasted for seven more days. It did not lead to significantly improved hearing thresholds compared to control animals. Quite the contrary, there is a hint for a higher noise susceptibility. Nevertheless, changes in the perilymph proteome indicate a reduced damage and better repair mechanisms through the IGF-1 treatment. Thus, future studies should investigate delivery methods enabling continuous prevention but reducing the risk of an overdosage.


Assuntos
Perda Auditiva Provocada por Ruído , Fator de Crescimento Insulin-Like I , Animais , Cobaias , Limiar Auditivo , Cóclea/metabolismo , Audição , Perda Auditiva Provocada por Ruído/prevenção & controle , Perda Auditiva Provocada por Ruído/terapia , Fator de Crescimento Insulin-Like I/uso terapêutico , Perilinfa , Sinapses
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA