Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
OTO Open ; 8(3): e162, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38974181

RESUMO

Objective: Evaluate ergonomic differences of various modalities for performing middle ear surgery. Study Design: Observational study. Setting: Two academic tertiary care centers. Methods: Attending physicians and residents performing middle ear surgery were photographed intraoperatively. Intraoperative photographs were analyzed using the validated Rapid Upper Limb Assessment (RULA) tool to measure musculoskeletal disease (MSD) risk. Descriptive statistics and significance testing were used to characterize and compare ergonomic differences between surgical modalities. Multivariable ordinal regression was performed to assess factors associated with increased MSD risk, as determined by the final RULA score. Results: Most of our 110 intraoperative photos featured attendings (82.7%) performing combined middle ear surgery and mastoidectomy (60.0%). Body angles and the final RULA score varied significantly among modalities. On subset analysis, microscopic surgery exhibited significantly worse wrist, trunk, and neck angles compared to endoscopic and exoscopic surgery. Exoscopic surgery had significantly lower final RULA scores than both endoscopic and microscopic surgery, indicating significantly lower MSD risk. Microscopic and endoscopic surgery final scores did not vary significantly. In a multivariable ordinal regression of factors associated with increased RULA score, exoscopic surgery had statistically significantly less ergonomic risk relative to microscopic surgery (odds ratio = 0.12, 95% confidence interval = [0.03-0.43]). Conclusion: Exoscopic, endoscopic, and microscopic surgery all featured low ergonomic risk, although exoscopic middle ear surgery demonstrated the lowest risk profile among studied surgical modalities. This demonstrates the importance of using each modality in combination with other ergonomic interventions to provide meaningful musculoskeletal benefits.

2.
J Neurosurg ; 138(4): 962-971, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36087315

RESUMO

OBJECTIVE: Vestibular schwannomas (VSs) are benign nerve sheath tumors that result from mutation in the tumor suppressor gene NF2, with functional loss of the protein merlin. The authors have previously shown that c-Jun N-terminal kinase (JNK) is constitutively active in human VS cells and plays a central role in their survival by suppressing accumulation of mitochondrial superoxides, implicating JNK inhibitors as a potential systemic treatment for VS. Thus, the authors hypothesized that the adenosine 5'-triphosphate-competitive JNK inhibitor AS602801 would demonstrate antitumor activity in multiple VS models. METHODS: Treatment with AS602801 was tested in primary human VS cultures, human VS xenografts, and a genetic mouse model of schwannoma (Postn-Cre;Nf2flox/flox). Primary human VS cell cultures were established from freshly obtained surgical tumor specimens; treatment group media was enriched with AS602801. VS xenograft tumors were established in male athymic nude mice from freshly collected human tumor. Four weeks postimplantation, a pretreatment MRI scan was obtained, followed by 65 days of AS602801 (n = 18) or vehicle control (n = 19) treatment. Posttreatment MRI scans were used to measure final tumor volume. Tumors were then harvested. Finally, Postn-Cre;Nf2flox/flox mice were treated with AS602801 (n = 10) or a vehicle (n = 13) for 65 days. Posttreatment auditory brainstem responses were obtained. Dorsal root ganglia from Postn-Cre;Nf2flox/flox mice were then harvested. In all models, schwannoma identity was confirmed with anti-S100 staining, cell proliferation was measured with the EdU assay, and cell death was measured with terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling staining. All protocols were approved by the local institutional review board and Institutional Animal Care and Use Committees. RESULTS: Treatment with AS602801 decreased cell proliferation and increased apoptosis in primary human VS cultures. The systemic administration of AS602801 in mice with human VS xenografts reduced tumor volume and cell proliferation. Last, the AS602801-treated Postn-Cre;Nf2flox/flox mice demonstrated decreased cell proliferation in glial cells in the dorsal root ganglia. However, AS602801 did not significantly delay hearing loss in Postn-Cre;Nf2flox/flox mice up to 3 months posttreatment. CONCLUSIONS: The data suggest that JNK inhibition with AS602801 suppresses growth of sporadic and neurofibromatosis type 2-associated VSs. As such, AS602801 is a potential systemic therapy for VS and warrants further investigation.


Assuntos
Neurofibromatose 2 , Neuroma Acústico , Humanos , Masculino , Camundongos , Animais , Neurofibromatose 2/complicações , Neurofibromatose 2/tratamento farmacológico , Neuroma Acústico/tratamento farmacológico , Neuroma Acústico/patologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos Nus
3.
Otolaryngol Clin North Am ; 55(3): 595-606, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35680205

RESUMO

Though there have been considerable strides in the diagnosis and care of orofacial pain disorders, facial neuralgias, and myofascial pain dysfunction syndrome remain incredibly cumbersome for patients and difficult to manage for providers. Cranial neuralgias, myofascial pain syndromes, temporomandibular dysfunction (TMD), dental pain, tumors, neurovascular pain, and psychiatric diseases can all present with similar symptoms. As a result, a patient's quest for the treatment of their orofacial pain often begins on the wrong foot, with a misdiagnosis or unnecessary procedure, which makes it all the more frustrating for them. Understanding the natural history, clinical presentation, and management of facial neuralgias and myofascial pain dysfunction syndrome can help clinicians better recognize and treat these conditions. In this article, we review updated knowledge on the pathophysiology, incidence, clinical features, diagnostic criteria, and medical management of TN, GPN, GN, and MPDS.


Assuntos
Doenças dos Nervos Cranianos , Neuralgia Facial , Neuralgia , Neuralgia do Trigêmeo , Doenças dos Nervos Cranianos/diagnóstico , Dor Facial/diagnóstico , Dor Facial/etiologia , Dor Facial/terapia , Cefaleia , Humanos , Neuralgia/diagnóstico , Neuralgia do Trigêmeo/diagnóstico
4.
Laryngoscope Investig Otolaryngol ; 6(3): 458-468, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34195368

RESUMO

OBJECTIVE: Radiation therapy is a mainstay in the treatment of numerous neoplasms. Numerous publications have reported good clinical outcomes for primary radiation therapy for Vestibular Schwannomas (VS). However, there are relatively few pathologic specimens of VSs available to evaluate post-radiation, which has led to a relative dearth in research on the cellular mechanisms underlying the effects of radiation therapy on VSs. METHODS: Here we review the latest literature on the complex biological effects of radiation therapy on these benign tumors-including resistance to oxidative stress, mechanisms of DNA damage repair, alterations in normal growth factor pathways, changes in surrounding vasculature, and alterations in immune responses following radiation. RESULTS: Although VSs are highly radioresistant, radiotherapy is often successful in arresting their growth. CONCLUSION: By better understanding the mechanisms underlying these effects, we could potentially harness such mechanisms in the future to potentiate the clinical effects of radiotherapy on VSs. LEVEL OF EVIDENCE: N/A.

5.
Laryngoscope ; 131(1): E259-E270, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32438526

RESUMO

OBJECTIVES: The objective was to explore the effect of the proneuronal transcription factor neurogenic differentiation 1 (Neurod1, ND1) on Schwann cells (SC) and schwannoma cell proliferation. METHODS: Using a variety of transgenic mouse lines, we investigated how expression of Neurod1 effects medulloblastoma (MB) growth, schwannoma tumor progression, vestibular function, and SC cell proliferation. Primary human vestibular schwannoma (VS) cell cultures were transduced with adenoviral vectors expressing Neurod1. Cell proliferation was assessed by 5-ethynyl-2'-deoxyuridine (EdU) uptake. STUDY DESIGN: Basic science investigation. RESULTS: Expression of Neurod1 reduced the growth of slow-growing but not fast-growing MB models. Gene transfer of Neurod1 in human schwannoma cultures significantly reduced cell proliferation in dose-dependent way. Deletion of the neurofibromatosis type 2 (Nf2) tumor-suppressor gene via Cre expression in SCs led to increased intraganglionic SC proliferation and mildly reduced vestibular sensory-evoked potentials (VsEP) responses compared to age-matched wild-type littermates. The effect of Neurod1-induced expression on intraganglionic SC proliferation in animals lacking Nf2 was mild and highly variable. Sciatic nerve axotomy significantly increased SC proliferation in wild-type and Nf2-null animals, and expression of Neurod1 reduced the proliferative capacity of both wild-type and Nf2-null SCs following nerve injury. CONCLUSION: Expression of Neurod1 reduces slow-growing MB progression and reduces human SC proliferation in primary VS cultures. In a genetic mouse model of schwannomas, we find some effects of Neurod1 expression; however, the high variability indicates that more tightly regulated Neurod1 expression levels that mimic our in vitro data are needed to fully validate Neurod1 effects on schwannoma progression. LEVEL OF EVIDENCE: NA Laryngoscope, 131:E259-E270, 2021.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Neuroma Acústico/metabolismo , Células de Schwann/metabolismo , Animais , Proliferação de Células , Humanos , Meduloblastoma/patologia , Camundongos , Camundongos Transgênicos , Neuroma Acústico/patologia , Células de Schwann/citologia , Células Tumorais Cultivadas
6.
Ann Otol Rhinol Laryngol ; 130(3): 314-318, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32772562

RESUMO

BACKGROUND: Petrous apicitis (PA) is a serious infection involving the apical portion of the petrous temporal bone. The classic triad of purulent otorrhea, ipsilateral abducens nerve palsy and retroorbital pain is rarely seen due to early detection and widespread use of antibiotics. Medical management is the primary treatment modality with surgery reserved for cases of recalcitrant petrous apex abscess. METHODS AND RESULTS: We presented a case of PA with previously untreated otitis media. After multidisciplinary evaluation, the patient was initially treated with intravenous antibiotics followed by drainage of the abscess using a combined transmastoid and middle cranial fossa (MCF) approach. The patient recovered well with no recurrence of the infection based on imaging and symptoms. DISCUSSION: While a variety of different surgical approaches can be used in treatment of PA, we recommend the MCF approach in cases where access to the anterior petrous apex may be challenging via transcanal or transmastoid approach.


Assuntos
Abscesso/terapia , Antibacterianos/uso terapêutico , Ventriculite Cerebral/terapia , Drenagem/métodos , Mastoidectomia/métodos , Mastoidite/terapia , Meningite/terapia , Otite Média Supurativa/terapia , Petrosite/terapia , Abscesso/diagnóstico por imagem , Ventriculite Cerebral/diagnóstico , Ventriculite Cerebral/etiologia , Confusão/etiologia , Fossa Craniana Média , Dor de Orelha , Humanos , Masculino , Mastoidite/diagnóstico por imagem , Meningite/diagnóstico , Meningite/etiologia , Pessoa de Meia-Idade , Otite Média Supurativa/diagnóstico por imagem , Petrosite/diagnóstico por imagem , Fotofobia/etiologia , Streptococcus pneumoniae , Tomografia Computadorizada por Raios X
7.
Mol Ther Methods Clin Dev ; 17: 1167-1177, 2020 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-32518805

RESUMO

Gene delivery is a key component for the treatment of genetic hearing loss. To date, a myriad of adeno-associated virus (AAV) serotypes and surgical approaches have been employed to deliver transgenes to cochlear hair cells, but the efficacy of dual transduction remains unclear. Herein, we investigated cellular tropism of single injections of AAV serotype 1 (AAV1), AAV2, AAV8, AAV9, and Anc80L65, and quantitated dual-vector co-transduction rates following co-injection of AAV2 and AAV9 vectors in adult murine cochlea. We used the combined round window membrane and canal fenestration (RWM+CF) injection technique for vector delivery. Single AAV2 injections were most robust and transduced 96.7% ± 1.1% of inner hair cells (IHCs) and 83.9% ± 2.0% of outer hair cells (OHCs) throughout the cochlea without causing hearing impairment or hair cell loss. Dual AAV2 injection co-transduced 96.9% ± 1.7% of IHCs and 65.6% ± 8.95% of OHCs. Together, RWM+CF-injected single or dual AAV2 provides the highest auditory hair cell transduction efficiency of the AAV serotypes we studied. These findings broaden the application of cochlear gene therapy targeting hair cells.

9.
Mol Ther ; 27(3): 681-690, 2019 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-30686588

RESUMO

Hearing loss is the most common human sensory deficit. Its correction has been the goal of several gene-therapy based studies exploring a variety of interventions. Although these studies report varying degrees of success, all treatments have targeted developing inner ears in neonatal mice, a time point in the structural maturation of the cochlea prior to 26 weeks gestational age in humans. It is unclear whether cochlear gene therapy can salvage hearing in the mature organ of Corti. Herein, we report the first study to test gene therapy in an adult murine model of human deafness. Using a single intracochlear injection of an artificial microRNA carried in an AAV vector, we show that RNAi-mediated gene silencing can slow progression of hearing loss, improve inner hair cell survival, and prevent stereocilia bundle degeneration in the mature Beethoven mouse, a model of human TMC1 deafness. The ability to study gene therapy in mature murine ears constitutes a significant step toward its translation to human subjects.


Assuntos
Surdez/genética , Vetores Genéticos/genética , Proteínas de Membrana/metabolismo , Animais , Cóclea/metabolismo , Cóclea/fisiopatologia , Cóclea/ultraestrutura , Surdez/fisiopatologia , Dependovirus/genética , Modelos Animais de Doenças , Orelha Interna/metabolismo , Orelha Interna/fisiopatologia , Orelha Interna/ultraestrutura , Terapia Genética , Perda Auditiva/genética , Perda Auditiva/fisiopatologia , Humanos , Imuno-Histoquímica , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C3H , Microscopia Eletrônica de Varredura , Interferência de RNA , Vestíbulo do Labirinto/metabolismo , Vestíbulo do Labirinto/fisiopatologia , Vestíbulo do Labirinto/ultraestrutura
10.
Sci Rep ; 8(1): 2980, 2018 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-29445157

RESUMO

Cochlear gene therapy holds promise for the treatment of genetic deafness. Assessing its impact in adult murine models of hearing loss, however, has been hampered by technical challenges that have made it difficult to establish a robust method to deliver transgenes to the mature murine inner ear. Here in we demonstrate the feasibility of a combined round window membrane injection and semi-circular canal fenestration technique in the adult cochlea. Injection of both AAV2/9 and AAV2/Anc80L65 via this approach in P15-16 and P56-60 mice permits robust eGFP transduction of virtually all inner hair cells throughout the cochlea with variable transduction of vestibular hair cells. Auditory thresholds are not compromised. Transduction rate and cell tropism is primarily influenced by viral titer and AAV serotype but not age at injection. This approach is safe, versatile and efficient. Its use will facilitate studies using cochlear gene therapy in murine models of hearing loss over a wide range of time points.


Assuntos
Cóclea/fisiologia , Surdez/terapia , Fenestração do Labirinto/métodos , Técnicas de Transferência de Genes , Terapia Genética/métodos , Células Ciliadas Auditivas Internas/fisiologia , Perda Auditiva/terapia , Adenoviridae/genética , Animais , Cóclea/cirurgia , Implante Coclear , Surdez/genética , Modelos Animais de Doenças , Meato Acústico Externo/cirurgia , Feminino , Vetores Genéticos , Perda Auditiva/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C3H , Janela da Cóclea/cirurgia , Membrana Timpânica/cirurgia
11.
Sci Rep ; 7(1): 9609, 2017 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-28852025

RESUMO

Gene therapy for genetic deafness is a promising approach by which to prevent hearing loss or to restore hearing after loss has occurred. Although a variety of direct approaches to introduce viral particles into the inner ear have been described, presumed physiological barriers have heretofore precluded investigation of systemic gene delivery to the cochlea. In this study, we sought to characterize systemic delivery of a rAAV2/9 vector as a non-invasive means of cochlear transduction. In wild-type neonatal mice (postnatal day 0-1), we show that intravenous injection of rAAV2/9 carrying an eGFP-reporter gene results in binaural transduction of inner hair cells, spiral ganglion neurons and vestibular hair cells. Transduction efficiency increases in a dose-dependent manner. Inner hair cells are transduced in an apex-to-base gradient, with transduction reaching 96% in the apical turn. Hearing acuity in treated animals is unaltered at postnatal day 30. Transduction is influenced by viral serotype and age at injection, with less efficient cochlear transduction observed with systemic delivery of rAAV2/1 and in juvenile mice with rAAV2/9. Collectively, these data validate intravenous delivery of rAAV2/9 as a novel and atraumatic technique for inner ear transgene delivery in early postnatal mice.


Assuntos
Cóclea/metabolismo , Dependovirus/genética , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Administração Intravenosa , Animais , Animais Recém-Nascidos , Dependovirus/classificação , Expressão Gênica , Genes Reporter , Vetores Genéticos/administração & dosagem , Imuno-Histoquímica , Camundongos , Gânglio Espiral da Cóclea/metabolismo , Transdução Genética , Transgenes
12.
Am J Hum Genet ; 98(6): 1101-1113, 2016 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-27236922

RESUMO

Hearing impairment is the most common sensory deficit. It is frequently caused by the expression of an allele carrying a single dominant missense mutation. Herein, we show that a single intracochlear injection of an artificial microRNA carried in a viral vector can slow progression of hearing loss for up to 35 weeks in the Beethoven mouse, a murine model of non-syndromic human deafness caused by a dominant gain-of-function mutation in Tmc1 (transmembrane channel-like 1). This outcome is noteworthy because it demonstrates the feasibility of RNA-interference-mediated suppression of an endogenous deafness-causing allele to slow progression of hearing loss. Given that most autosomal-dominant non-syndromic hearing loss in humans is caused by this mechanism of action, microRNA-based therapeutics might be broadly applicable as a therapy for this type of deafness.


Assuntos
Vias Auditivas , Perda Auditiva/prevenção & controle , Proteínas de Membrana/fisiologia , MicroRNAs/genética , Mutação de Sentido Incorreto/genética , Animais , Dependovirus/genética , Perda Auditiva/etiologia , Perda Auditiva/patologia , Humanos , Mecanotransdução Celular , Proteínas de Membrana/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , MicroRNAs/administração & dosagem , Interferência de RNA
13.
PLoS Genet ; 11(3): e1005137, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25816005

RESUMO

Hereditary hearing loss is a clinically and genetically heterogeneous disorder. More than 80 genes have been implicated to date, and with the advent of targeted genomic enrichment and massively parallel sequencing (TGE+MPS) the rate of novel deafness-gene identification has accelerated. Here we report a family segregating post-lingual progressive autosomal dominant non-syndromic hearing loss (ADNSHL). After first excluding plausible variants in known deafness-causing genes using TGE+MPS, we completed whole exome sequencing in three hearing-impaired family members. Only a single variant, p.Arg185Pro in HOMER2, segregated with the hearing-loss phenotype in the extended family. This amino acid change alters a highly conserved residue in the coiled-coil domain of HOMER2 that is essential for protein multimerization and the HOMER2-CDC42 interaction. As a scaffolding protein, HOMER2 is involved in intracellular calcium homeostasis and cytoskeletal organization. Consistent with this function, we found robust expression in stereocilia of hair cells in the murine inner ear and observed that over-expression of mutant p.Pro185 HOMER2 mRNA causes anatomical changes of the inner ear and neuromasts in zebrafish embryos. Furthermore, mouse mutants homozygous for the targeted deletion of Homer2 present with early-onset rapidly progressive hearing loss. These data provide compelling evidence that HOMER2 is required for normal hearing and that its sequence alteration in humans leads to ADNSHL through a dominant-negative mode of action.


Assuntos
Proteínas de Transporte/genética , Orelha Interna/metabolismo , Exoma/genética , Perda Auditiva Neurossensorial/genética , Animais , Proteínas de Transporte/biossíntese , Cóclea/metabolismo , Cóclea/patologia , Orelha Interna/patologia , Regulação da Expressão Gênica , Perda Auditiva Neurossensorial/patologia , Sequenciamento de Nucleotídeos em Larga Escala , Proteínas de Arcabouço Homer , Humanos , Camundongos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Estereocílios/genética , Estereocílios/patologia , Peixe-Zebra , Proteína cdc42 de Ligação ao GTP/genética , Proteína cdc42 de Ligação ao GTP/metabolismo
14.
Sci Rep ; 5: 8619, 2015 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-25726967

RESUMO

Cochlear hair cell loss results in secondary regression of peripheral auditory fibers (PAFs) and loss of spiral ganglion neurons (SGNs). The performance of cochlear implants (CI) in rehabilitating hearing depends on survival of SGNs. Here we compare the effects of adeno-associated virus vectors with neurotrophin gene inserts, AAV.BDNF and AAV.Ntf3, on guinea pig ears deafened systemically (kanamycin and furosemide) or locally (neomycin). AAV.BDNF or AAV.Ntf3 was delivered to the guinea pig cochlea one week following deafening and ears were assessed morphologically 3 months later. At that time, neurotrophins levels were not significantly elevated in the cochlear fluids, even though in vitro and shorter term in vivo experiments demonstrate robust elevation of neurotrophins with these viral vectors. Nevertheless, animals receiving these vectors exhibited considerable re-growth of PAFs in the basilar membrane area. In systemically deafened animals there was a negative correlation between the presence of differentiated supporting cells and PAFs, suggesting that supporting cells influence the outcome of neurotrophin over-expression aimed at enhancing the cochlear neural substrate. Counts of SGN in Rosenthal's canal indicate that BDNF was more effective than NT-3 in preserving SGNs. The results demonstrate that a transient elevation in neurotrophin levels can sustain the cochlear neural substrate in the long term.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/uso terapêutico , Surdez/terapia , Dependovirus/metabolismo , Orelha/patologia , Neurotrofina 3/uso terapêutico , Envelhecimento , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Sobrevivência Celular , Meios de Cultura , Surdez/patologia , Epitélio/metabolismo , Epitélio/patologia , Feminino , Vetores Genéticos , Cobaias , Células Labirínticas de Suporte/metabolismo , Células Labirínticas de Suporte/patologia , Masculino , Neomicina , Neurotrofina 3/genética , Perilinfa/metabolismo , Gânglio Espiral da Cóclea/metabolismo , Gânglio Espiral da Cóclea/patologia , Resultado do Tratamento
15.
Hum Gene Ther ; 23(3): 302-10, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22074321

RESUMO

Cochlear gene therapy can be a new avenue for the treatment of severe hearing loss by inducing regeneration or phenotypic rescue. One necessary step to establish this therapy is the development of a safe and feasible inoculation surgery, ideally without drilling the bony cochlear wall. The round window membrane (RWM) is accessible in the middle-ear space, but viral vectors placed on this membrane do not readily cross the membrane to the cochlear tissues. In an attempt to enhance permeability of the RWM, we applied hyaluronic acid (HA), a nontoxic and biodegradable reagent, onto the RWM of guinea pigs, prior to delivering an adenovirus carrying enhanced green fluorescent protein (eGFP) reporter gene (Ad-eGFP) at the same site. We examined distribution of eGFP in the cochlea 1 week after treatment, comparing delivery of the vector via the RWM, with or without HA, to delivery by a cochleostomy into the perilymph. We found that cochlear tissue treated with HA-assisted delivery of Ad-eGFP demonstrated wider expression of transgenes in cochlear cells than did tissue treated by cochleostomy injection. HA-assisted vector delivery facilitated expression in cells lining the scala media, which are less accessible and not transduced after perilymphatic injection. We assessed auditory function by measuring auditory brainstem responses and determined that thresholds were significantly better in the ears treated with HA-assisted Ad-eGFP placement on the RWM as compared with cochleostomy. Together, these data demonstrate that HA-assisted delivery of viral vectors provides an atraumatic and clinically feasible method to introduce transgenes into cochlear cells, thereby enhancing both research methods and future clinical application.


Assuntos
Cóclea/metabolismo , Perda Auditiva/terapia , Ácido Hialurônico/farmacologia , Animais , Cóclea/cirurgia , Potenciais Evocados Auditivos do Tronco Encefálico , Feminino , Técnicas de Transferência de Genes , Terapia Genética , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Cobaias , Perda Auditiva/genética , Perda Auditiva/fisiopatologia , Ácido Hialurônico/metabolismo , Masculino , Janela da Cóclea/metabolismo , Transgenes
16.
Exp Neurol ; 223(2): 464-72, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20109446

RESUMO

Sensory organs typically use receptor cells and afferent neurons to transduce environmental signals and transmit them to the CNS. When sensory cells are lost, nerves often regress from the sensory area. Therapeutic and regenerative approaches would benefit from the presence of nerve fibers in the tissue. In the hearing system, retraction of afferent innervation may accompany the degeneration of auditory hair cells that is associated with permanent hearing loss. The only therapy currently available for cases with severe or complete loss of hair cells is the cochlear implant auditory prosthesis. To enhance the therapeutic benefits of a cochlear implant, it is necessary to attract nerve fibers back into the cochlear epithelium. Here we show that forced expression of the neurotrophin gene BDNF in epithelial or mesothelial cells that remain in the deaf ear induces robust regrowth of nerve fibers towards the cells that secrete the neurotrophin, and results in re-innervation of the sensory area. The process of neurotrophin-induced neuronal regeneration is accompanied by significant preservation of the spiral ganglion cells. The ability to regrow nerve fibers into the basilar membrane area and protect the auditory nerve will enhance performance of cochlear implants and augment future cell replacement therapies such as stem cell implantation or induced transdifferentiation. This model also provides a general experimental stage for drawing nerve fibers into a tissue devoid of neurons, and studying the interaction between the nerve fibers and the tissue.


Assuntos
Membrana Basilar/citologia , Fator Neurotrófico Derivado do Encéfalo/genética , Surdez/terapia , Terapia Genética/métodos , Regeneração Nervosa/fisiologia , Gânglio Espiral da Cóclea/citologia , Adenoviridae/genética , Animais , Animais Geneticamente Modificados , Membrana Basilar/fisiologia , Surdez/fisiopatologia , Células Epiteliais/fisiologia , Epitélio/fisiologia , Proteínas de Fluorescência Verde/genética , Cobaias , Células Ciliadas Auditivas/fisiologia , Células Ciliadas Auditivas/ultraestrutura , Masculino , Fibras Nervosas/fisiologia , Gânglio Espiral da Cóclea/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA