Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Int J Mol Sci ; 24(8)2023 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-37108158

RESUMO

Sensorineural hearing loss is the most prevalent sensory deficit in humans. Most cases of hearing loss are due to the degeneration of key structures of the sensory pathway in the cochlea, such as the sensory hair cells, the primary auditory neurons, and their synaptic connection to the hair cells. Different cell-based strategies to replace damaged inner ear neurosensory tissue aiming at the restoration of regeneration or functional recovery are currently the subject of intensive research. Most of these cell-based treatment approaches require experimental in vitro models that rely on a fine understanding of the earliest morphogenetic steps that underlie the in vivo development of the inner ear since its initial induction from a common otic-epibranchial territory. This knowledge will be applied to various proposed experimental cell replacement strategies to either address the feasibility or identify novel therapeutic options for sensorineural hearing loss. In this review, we describe how ear and epibranchial placode development can be recapitulated by focusing on the cellular transformations that occur as the inner ear is converted from a thickening of the surface ectoderm next to the hindbrain known as the otic placode to an otocyst embedded in the head mesenchyme. Finally, we will highlight otic and epibranchial placode development and morphogenetic events towards progenitors of the inner ear and their neurosensory cell derivatives.


Assuntos
Orelha Interna , Perda Auditiva Neurossensorial , Humanos , Orelha Interna/fisiologia , Células Ciliadas Auditivas/metabolismo , Audição , Perda Auditiva Neurossensorial/terapia , Perda Auditiva Neurossensorial/metabolismo , Células Receptoras Sensoriais
2.
Stem Cells ; 39(6): 697-706, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33522002

RESUMO

The sense of hearing depends on a specialized sensory organ in the inner ear, called the cochlea, which contains the auditory hair cells (HCs). Noise trauma, infections, genetic factors, side effects of ototoxic drugs (ie, some antibiotics and chemotherapeutics), or simply aging lead to the loss of HCs and their associated primary neurons. This results in irreversible sensorineural hearing loss (SNHL) as in mammals, including humans; the inner ear lacks the capacity to regenerate HCs and spiral ganglion neurons. SNHL is a major global health problem affecting millions of people worldwide and provides a growing concern in the aging population. To date, treatment options are limited to hearing aids and cochlear implants. A major bottleneck for development of new therapies for SNHL is associated to the lack of human otic cell bioassays. Human induced pluripotent stem cells (hiPSCs) can be induced in two-dimensional and three-dimensional otic cells in vitro models that can generate inner ear progenitors and sensory HCs and could be a promising preclinical platform from which to work toward restoring SNHL. We review the potential applications of hiPSCs in the various biological approaches, including disease modeling, bioengineering, drug testing, and autologous stem cell based-cell therapy, that offer opportunities to understand the pathogenic mechanisms of SNHL and identify novel therapeutic strategies.


Assuntos
Orelha Interna/patologia , Perda Auditiva Neurossensorial/terapia , Células-Tronco Pluripotentes Induzidas/citologia , Gânglio Espiral da Cóclea/citologia , Animais , Diferenciação Celular/fisiologia , Perda Auditiva Neurossensorial/patologia , Humanos , Regeneração/fisiologia
3.
Int J Mol Sci ; 22(19)2021 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-34639189

RESUMO

We analyzed transcriptomic data from otic sensory cells differentiated from human induced pluripotent stem cells (hiPSCs) by a previously described method to gain new insights into the early human otic neurosensory lineage. We identified genes and biological networks not previously described to occur in the human otic sensory developmental cell lineage. These analyses identified and ranked genes known to be part of the otic sensory lineage program (SIX1, EYA1, GATA3, etc.), in addition to a number of novel genes encoding extracellular matrix (ECM) (COL3A1, COL5A2, DCN, etc.) and integrin (ITG) receptors (ITGAV, ITGA4, ITGA) for ECM molecules. The results were confirmed by quantitative PCR analysis of a comprehensive panel of genes differentially expressed during the time course of hiPSC differentiation in vitro. Immunocytochemistry validated results for select otic and ECM/ITG gene markers in the in vivo human fetal inner ear. Our screen shows ECM and ITG gene expression changes coincident with hiPSC differentiation towards human otic neurosensory cells. Our findings suggest a critical role of ECM-ITG interactions with otic neurosensory lineage genes in early neurosensory development and cell fate determination in the human fetal inner ear.


Assuntos
Diferenciação Celular , Orelha Interna/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Neurais/citologia , Células Receptoras Sensoriais/citologia , Células Receptoras Sensoriais/metabolismo , Transcriptoma , Linhagem da Célula , Orelha Interna/metabolismo , Matriz Extracelular/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Integrinas/genética , Integrinas/metabolismo , Células-Tronco Neurais/metabolismo , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo
4.
Mol Ther ; 27(6): 1101-1113, 2019 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-31005598

RESUMO

Most cases of sensorineural deafness are caused by degeneration of hair cells. Although stem/progenitor cell therapy is becoming a promising treatment strategy in a variety of organ systems, cell engraftment in the adult mammalian cochlea has not yet been demonstrated. In this study, we generated human otic progenitor cells (hOPCs) from induced pluripotent stem cells (iPSCs) in vitro and identified these cells by the expression of known otic markers. We showed successful cell transplantation of iPSC-derived-hOPCs in an in vivo adult guinea pig model of ototoxicity. The delivered hOPCs migrated throughout the cochlea, engrafted in non-sensory regions, and survived up to 4 weeks post-transplantation. Some of the engrafted hOPCs responded to environmental cues within the cochlear sensory epithelium and displayed molecular features of early sensory differentiation. We confirmed these results with hair cell progenitors derived from Atoh1-GFP mice as donor cells. These mouse otic progenitors transplanted using the same in vivo delivery system migrated into damaged cochlear sensory epithelium and adopted a partial sensory cell fate. This is the first report of the survival and differentiation of hOPCs in ototoxic-injured mature cochlear epithelium, and it should stimulate further research into cell-based therapies for treatment of deafness.


Assuntos
Crescimento Celular , Células Ciliadas Auditivas/efeitos dos fármacos , Perda Auditiva/cirurgia , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/transplante , Ototoxicidade/cirurgia , Transplante de Células-Tronco/métodos , Amicacina/efeitos adversos , Amicacina/farmacologia , Animais , Limiar Auditivo/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Ciclosporina/farmacologia , Modelos Animais de Doenças , Fator 10 de Crescimento de Fibroblastos/farmacologia , Fator 3 de Crescimento de Fibroblastos/farmacologia , Cobaias , Células Ciliadas Auditivas/imunologia , Células Ciliadas Auditivas/metabolismo , Perda Auditiva/induzido quimicamente , Humanos , Imunossupressores/farmacologia , Células-Tronco Pluripotentes Induzidas/imunologia , Doadores Vivos
5.
Front Aging Neurosci ; 14: 814528, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35250542

RESUMO

Age-related hearing loss (ARHL) is a common, increasing problem for older adults, affecting about 1 billion people by 2050. We aim to correlate the different reductions of hearing from cochlear hair cells (HCs), spiral ganglion neurons (SGNs), cochlear nuclei (CN), and superior olivary complex (SOC) with the analysis of various reasons for each one on the sensory deficit profiles. Outer HCs show a progressive loss in a basal-to-apical gradient, and inner HCs show a loss in a apex-to-base progression that results in ARHL at high frequencies after 70 years of age. In early neonates, SGNs innervation of cochlear HCs is maintained. Loss of SGNs results in a considerable decrease (~50% or more) of cochlear nuclei in neonates, though the loss is milder in older mice and humans. The dorsal cochlear nuclei (fusiform neurons) project directly to the inferior colliculi while most anterior cochlear nuclei reach the SOC. Reducing the number of neurons in the medial nucleus of the trapezoid body (MNTB) affects the interactions with the lateral superior olive to fine-tune ipsi- and contralateral projections that may remain normal in mice, possibly humans. The inferior colliculi receive direct cochlear fibers and second-order fibers from the superior olivary complex. Loss of the second-order fibers leads to hearing loss in mice and humans. Although ARHL may arise from many complex causes, HC degeneration remains the more significant problem of hearing restoration that would replace the cochlear implant. The review presents recent findings of older humans and mice with hearing loss.

6.
Stem Cells ; 27(11): 2722-33, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19785035

RESUMO

In humans and rodents the adult spinal cord harbors neural stem cells located around the central canal. Their identity, precise location, and specific signaling are still ill-defined and controversial. We report here on a detailed analysis of this niche. Using microdissection and glial fibrillary acidic protein (GFAP)-green fluorescent protein (GFP) transgenic mice, we demonstrate that neural stem cells are mostly dorsally located GFAP(+) cells lying ependymally and subependymally that extend radial processes toward the pial surface. The niche also harbors doublecortin protein (Dcx)(+) Nkx6.1(+) neurons sending processes into the lumen. Cervical and lumbar spinal cord neural stem cells maintain expression of specific rostro-caudal Hox gene combinations and the niche shows high levels of signaling proteins (CD15, Jagged1, Hes1, differential screening-selected gene aberrative in neuroblastoma [DAN]). More surprisingly, the niche displays mesenchymal traits such as expression of epithelial-mesenchymal-transition zinc finger E-box-binding protein 1 (ZEB1) transcription factor and smooth muscle actin. We found ZEB1 to be essential for neural stem cell survival in vitro. Proliferation within the niche progressively ceases around 13 weeks when the spinal cord reaches its final size, suggesting an active role in postnatal development. In addition to hippocampus and subventricular zone niches, adult spinal cord constitutes a third central nervous system stem cell niche with specific signaling, cellular, and structural characteristics that could possibly be manipulated to alleviate spinal cord traumatic and degenerative diseases.


Assuntos
Proteína Glial Fibrilar Ácida/metabolismo , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Medula Espinal/citologia , Medula Espinal/metabolismo , Nicho de Células-Tronco/citologia , Nicho de Células-Tronco/metabolismo , Células-Tronco/citologia , Actinas/metabolismo , Animais , Proliferação de Células , Proteína Duplacortina , Regulação da Expressão Gênica no Desenvolvimento , Proteína Glial Fibrilar Ácida/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Transgênicos , Neurônios/citologia , Neurônios/metabolismo , Células-Tronco/metabolismo , Homeobox 1 de Ligação a E-box em Dedo de Zinco
7.
PLoS One ; 15(10): e0240731, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33048968

RESUMO

[This corrects the article DOI: 10.1371/journal.pone.0042987.].

8.
PLoS One ; 15(10): e0240732, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33048993

RESUMO

[This corrects the article DOI: 10.1371/journal.pone.0088757.].

9.
Anat Rec (Hoboken) ; 303(3): 451-460, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31943808

RESUMO

The mammalian inner ear mediates hearing and balance and during development generates both cochleo-vestibular ganglion neurons and sensory epithelial receptor cells, that is, hair cells and support cells. Cell marking experiments have shown that both hair cells and support cells can originate from a common progenitor. Here, we demonstrate the lineage potential of individual otic epithelial cell clones using three cell lines established by a combination of limiting dilution and gene-marking techniques from an embryonic day 12 (E12) rat otocyst. Cell-type specific marker analyses of these clonal lines under proliferation and differentiation culture conditions demonstrate that during differentiation immature cell markers (Nanog and Nestin) were downregulated and hair cell (Myosin VIIa and Math1), support cell (p27Kip1 and cytokeratin) and neuronal cell (NF-H and NeuroD) markers were upregulated. Our results suggest that the otic epithelium of the E12 mammalian inner ear possess multipotent progenitor cells able to generate cell types of both sensory epithelial and neural cell lineages when cultured under a differentiation culture condition. Understanding the molecular mechanisms of proliferation and differentiation of multipotent otic progenitor cells may provide insights that could contribute to the development of a novel cell therapy with a potential to initiate or stimulate the sensorineural repair of damaged inner ear sensory receptors. Anat Rec, 303:451-460, 2020. © 2019 American Association for Anatomy.


Assuntos
Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Orelha Interna/citologia , Células Ciliadas Auditivas/citologia , Neurônios/citologia , Células-Tronco/citologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Orelha Interna/embriologia , Orelha Interna/metabolismo , Células Ciliadas Auditivas/metabolismo , Miosina VIIa/metabolismo , Proteína Homeobox Nanog/metabolismo , Nestina/metabolismo , Neurônios/metabolismo , Ratos , Ratos Wistar , Células-Tronco/metabolismo
11.
Front Mol Neurosci ; 11: 452, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30618604

RESUMO

Age-related neurosensory deficit of the inner ear is mostly due to a loss of hair cells (HCs). Development of stem cell-based therapy requires a better understanding of factors and signals that drive stem cells into otic sensory progenitor cells (OSPCs) to replace lost HCs. Human induced pluripotent stem cells (hiPSCs) theoretically represent an unlimited supply for the generation of human OSPCs in vitro. In this study, we developed a monolayer-based differentiation system to generate an enriched population of OSPCs via a stepwise differentiation of hiPSCs. Gene and protein expression analyses revealed the efficient induction of a comprehensive panel of otic/placodal and late otic markers over the course of the differentiation. Furthermore, whole transcriptome analysis confirmed a developmental path of OSPC differentiation from hiPSCs. We found that modulation of WNT and transforming growth factor-ß (TGF-ß) signaling combined with fibroblast growth factor 3 (FGF3) and FGF10 treatment over a 6-day period drives the expression of early otic/placodal markers followed by late otic sensory markers within 13 days, indicative of a differentiation into embryonic-like HCs. In summary, we report a rapid and efficient strategy to generate an enriched population of OSPCs from hiPSCs, thereby establishing the value of this approach for disease modeling and cell-based therapies of the inner ear.

12.
PLoS One ; 13(6): e0198954, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29902227

RESUMO

The inner ear represents a promising system to develop cell-based therapies from human induced pluripotent stem cells (hiPSCs). In the developing ear, Notch signaling plays multiple roles in otic region specification and for cell fate determination. Optimizing hiPSC induction for the generation of appropriate numbers of otic progenitors and derivatives, such as hair cells, may provide an unlimited supply of cells for research and cell-based therapy. In this study, we used monolayer cultures, otic-inducing agents, Notch modulation, and marker expression to track early and otic sensory lineages during hiPSC differentiation. Otic/placodal progenitors were derived from hiPSC cultures in medium supplemented with FGF3/FGF10 for 13 days. These progenitor cells were then treated for 7 days with retinoic acid (RA) and epidermal growth factor (EGF) or a Notch inhibitor. The differentiated cultures were analyzed in parallel by qPCR and immunocytochemistry. After the 13 day induction, hiPSC-derived cells displayed an upregulated expression of a panel of otic/placodal markers. Strikingly, a subset of these induced progenitor cells displayed key-otic sensory markers, the percentage of which was increased in cultures under Notch inhibition as compared to RA/EGF-treated cultures. Our results show that modulating Notch pathway during in vitro differentiation of hiPSC-derived otic/placodal progenitors is a valuable strategy to promote the expression of human otic sensory lineage genes.


Assuntos
Orelha Interna/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Diferenciação Celular , Linhagem da Célula , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Receptores Notch/metabolismo
13.
Hear Res ; 226(1-2): 168-77, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17098385

RESUMO

Neomycin ototoxicity and electrode insertion trauma both involve activation of the mitogen activated protein kinase (MAPK)/c-Jun-N-terminal kinase (JNK) cell death signal cascade. This article discusses mechanisms of cell death on a cell biology level (e.g. necrosis and apoptosis) and proposes the blocking of JNK signaling as a therapeutic approach for preventing the development of a permanent hearing loss that can be initiated by either neomycin ototoxicity or electrode insertion trauma. Blocking of JNK molecules incorporates the use of a peptide inhibitor (i.e. D-JNKI-1), which is specific for all three isoforms of JNK and has been demonstrated to prevent loss of hearing following either electrode insertion trauma or loss of both hearing and hair cells following exposure to an ototoxic level of neomycin. We present previously unpublished results that control for the effect of perfusate washout of aminoglycoside antibiotic by perfusion of the scala tympani with an inactive form of D-JNKI-1 peptide, i.e. JNKI-1(mut) peptide, which was not presented in the original J. Neurosci. article that tested locally delivered D-JNKI-1 peptide against both noise- and neomycin-induced hearing loss (i.e. Wang, J., Van De Water, T.R., Bonny, C., de Ribaupierre, F., Puel, J.L., Zine, A. 2003a. A peptide inhibitor of c-Jun N-terminal kinase protects against both aminoglycoside and acoustic trauma-induced auditory hair cell death and hearing loss. J. Neurosci. 23, 8596-8607). D-JNKI-1 is a cell permeable peptide that blocks JNK signaling at the level of the three JNK molecular isoforms, which when blocked prevents the increases in hearing thresholds and the loss of auditory hair cells. This unique therapeutic approach may have clinical application for preventing: (1) hearing loss caused by neomycin ototoxicity; and (2) the progressive component of electrode insertion trauma-induced hearing loss.


Assuntos
Perda Auditiva/prevenção & controle , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Animais , Antibacterianos/toxicidade , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Eletrodos/efeitos adversos , Radicais Livres/metabolismo , Cobaias , Perda Auditiva/induzido quimicamente , Perda Auditiva/enzimologia , Perda Auditiva/etiologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Neomicina/toxicidade , Peptídeos/farmacologia , Transdução de Sinais/efeitos dos fármacos
14.
J Tissue Eng Regen Med ; 11(9): 2629-2642, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-27099197

RESUMO

The generation of replacement inner ear hair cells (HCs) remains a challenge and stem cell therapy holds the potential for developing therapeutic solutions to hearing and balance disorders. Recent developments have made significant strides in producing mouse otic progenitors using cell culture techniques to initiate HC differentiation. However, no consensus has been reached as to efficiency and therefore current methods remain unsatisfactory. In order to address these issues, we compare the generation of otic and HC progenitors from embryonic stem (ES) cells in two cell culture systems: suspension vs. adherent conditions. In the present study, an ES cell line derived from an Atoh1-green fluorescent protein (GFP) transgenic mouse was used to track the generation of otic progenitors, initial HCs and to compare these two differentiation systems. We used a two-step short-term differentiation method involving an induction period of 5 days during which ES cells were cultured in the presence of Wnt/transforming growth factor TGF-ß inhibitors and insulin-like growth factor IGF-1 to suppress mesoderm and reinforce presumptive ectoderm and otic lineages. The generated embryoid bodies were then differentiated in medium containing basic fibroblast growth factor (bFGF) for an additional 5 days using either suspension or adherent culture methods. Upon completion of differentiation, quantitative polymerase chain reaction analysis and immunostaining monitored the expression of otic/HC progenitor lineage markers. The results indicate that cells differentiated in suspension cultures produced cells expressing otic progenitor/HC markers at a higher efficiency compared with the production of these cell types within adherent cultures. Furthermore, we demonstrated that a fraction of these cells can incorporate into ototoxin-injured mouse postnatal cochlea explants and express MYO7A after transplantation. Copyright © 2016 John Wiley & Sons, Ltd.


Assuntos
Antígenos de Diferenciação/metabolismo , Diferenciação Celular , Células Ciliadas Auditivas Internas , Células-Tronco Embrionárias Murinas/metabolismo , Transplante de Células-Tronco , Animais , Técnicas de Cultura de Células , Células Ciliadas Auditivas Internas/citologia , Células Ciliadas Auditivas Internas/metabolismo , Células Ciliadas Auditivas Internas/transplante , Camundongos , Camundongos Transgênicos , Células-Tronco Embrionárias Murinas/citologia
15.
Otol Neurotol ; 27(4): 504-11, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16791042

RESUMO

HYPOTHESES: 1) Hearing loss caused by electrode insertion trauma has both acute and delayed components; and 2) the delayed component of trauma-initiated hearing loss can be prevented by a direct delivery of a peptide inhibitor of the c-Jun N-terminal kinase cell death signal cascade, that is, D-JNKI-1, immediately after the electrode insertion within the cochlea. BACKGROUND: Acute trauma to the macroscopic elements of the cochlea from electrode insertion is well known. The impact of trauma-induced oxidative stress within injured cochlear tissues and the efficacy of drugs (e.g., D-JNKI-1) to prevent apoptosis of damaged hair cells is not well defined. METHODS: Hearing function was tested by pure-tone evoked auditory brainstem responses (ABRs) and distortion products of otoacoustic emissions (DPOAEs). D-JNKI-1 in artificial perilymph (AP) or AP alone was delivered into the scala tympani immediately after electrode trauma and for 7 days. Controls were nontreated contralateral and D-JNKI-1-treated ears without electrode insertion trauma. RESULTS: There was no increase in the hearing thresholds of either the contralateral control ears or in the D-JNKI-1 without trauma animals. There was a progressive increase in ABR thresholds and decrease in DPOAE amplitudes after electrode insertion trauma in untreated and in AP-treated cochleae. Treatment with D-JNKI-1 prevented the progressive increase in ABR thresholds and decrease in DPOAE amplitudes that occur after electrode insertion trauma. CONCLUSION: Hearing loss caused by cochlear implant electrode insertion trauma in guinea pigs has both acute and delayed components. The delayed component can be prevented by treating the cochlea with D-JNKI-1.


Assuntos
Cóclea/lesões , Implante Coclear/efeitos adversos , Perda Auditiva/prevenção & controle , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Peptídeos/uso terapêutico , Análise de Variância , Animais , Audiometria de Tons Puros , Limiar Auditivo/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Eletrodos/efeitos adversos , Potenciais Evocados Auditivos do Tronco Encefálico , Cobaias , Perda Auditiva/etiologia , Perda Auditiva/terapia , Órgão Espiral/efeitos dos fármacos , Emissões Otoacústicas Espontâneas , Peptídeos/farmacologia , Distribuição Aleatória , Fatores de Tempo , Resultado do Tratamento
16.
Rev Neurosci ; 16(1): 57-67, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15810654

RESUMO

The c-Jun N-terminal kinases (JNKs) are members of the family of mitogen activated protein kinases (MAPKs). While the functions of the JNKs under physiological conditions are diverse and not completely understood, there is increasing evidence that JNKs are potent effectors of apoptosis in both the brain and the mammalian inner ear following a variety of injuries. The activation of the inducible transcription factor c-Jun by N-terminal phosphorylation is a central event in JNK-mediated neural and inner ear hair cell death. A cell permeable peptide designed specifically to inhibit JNK signaling has proven successful in in vivo models of both neuronal degeneration following cerebral ischemia and auditory hair cell degeneration following exposure to either acoustic trauma or a toxic level of an aminoglycoside antibiotic. Here we discuss the evidence supporting the application of JNK inhibitors to prevent cellular degeneration in several central nervous system (CNS) and peripheral nervous system (PNS) diseases with an emphasis on traumatic ischemic damage to the CNS and acquired deafness in the PNS receptors.


Assuntos
Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Degeneração Neural/tratamento farmacológico , Degeneração Neural/enzimologia , Doenças do Sistema Nervoso/tratamento farmacológico , Doenças do Sistema Nervoso/enzimologia , Fármacos Neuroprotetores/farmacologia , Animais , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/enzimologia , Isquemia Encefálica/fisiopatologia , Surdez/tratamento farmacológico , Surdez/enzimologia , Surdez/fisiopatologia , Células Ciliadas Auditivas/efeitos dos fármacos , Células Ciliadas Auditivas/enzimologia , Células Ciliadas Auditivas/fisiopatologia , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/efeitos dos fármacos , Degeneração Neural/prevenção & controle , Doenças do Sistema Nervoso/fisiopatologia , Fármacos Neuroprotetores/química , Síndromes Neurotóxicas/tratamento farmacológico , Síndromes Neurotóxicas/enzimologia , Síndromes Neurotóxicas/fisiopatologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
17.
Front Cell Neurosci ; 9: 96, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25873860

RESUMO

Loss of auditory sensory hair cells (HCs) is the most common cause of hearing loss. This review addresses the signaling pathways that are involved in the programmed and necrotic cell death of auditory HCs that occur in response to ototoxic and traumatic stressor events. The roles of inflammatory processes, oxidative stress, mitochondrial damage, cell death receptors, members of the mitogen-activated protein kinase (MAPK) signal pathway and pro- and anti-cell death members of the Bcl-2 family are explored. The molecular interaction of these signal pathways that initiates the loss of auditory HCs following acoustic trauma is covered and possible therapeutic interventions that may protect these sensory HCs from loss via apoptotic or non-apoptotic cell death are explored.

18.
Mol Neurobiol ; 27(2): 223-38, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12777689

RESUMO

Mechanosensory hair cells of the vertebrate cochlea offer an excellent developmental system to study cell-fate specification, and to gain insight into the many human neurological deficits which result in a hearing loss, by affecting primarily the hair cells. Therefore, there is great interest in studying the molecular mechanisms that regulate their specification and differentiation. Recent studies, based mostly on loss-of-function experiments that target the role of Notch signaling and basic helix-loop-helix genes in inner-ear development have indicated that they can regulate mechanosensory hair cell-fate specification and their initial differentiation.


Assuntos
Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Células Ciliadas Auditivas/embriologia , Células Ciliadas Auditivas/metabolismo , Proteínas de Membrana/metabolismo , Receptores de Superfície Celular , Células-Tronco/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Células Ciliadas Auditivas/ultraestrutura , Humanos , Estrutura Terciária de Proteína/fisiologia , Receptor Notch1 , Células-Tronco/ultraestrutura , Fatores de Transcrição/metabolismo
19.
Hear Res ; 170(1-2): 22-31, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12208538

RESUMO

The sensory epithelium of the mammalian cochlea (the organ of Corti) represents an excellent developmental system. The organ of Corti contains two main cell types: the sensory hair cells and the supporting cells which are organized in a defined mosaic pattern. Previous results have demonstrated the participation of Notch signaling in the regulation of the pattern of hair cell differentiation within this sensory mosaic. It has also been shown that the basic helix-loop-helix (bHLH) transcription factor Math1 is a positive regulator of hair cell differentiation. We demonstrated that Hes1 and Hes5, two members of the inhibitory bHLH transcription factors, act as negative regulators of hair cell differentiation. Loss-of-function studies implicating the neurogenic genes Notch1, Jag2, Hes1 and Hes5 generated a significant increase in the number of hair cells. However, their functional interplay within the organ of Corti has not been determined. To clarify the mechanisms that regulate hair cell differentiation, we examined the expression of Notch/Notch ligand system and Math1 in the developing organ of Corti of Hes1- and Hes5-deficient mice. Our study suggests complex specific relationships between Notch signaling, Math1 and Hes1/Hes5 in the control of hair cell differentiation in the developing organ of Corti.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Homeodomínio/genética , Proteínas de Membrana/metabolismo , Mutação/fisiologia , Órgão Espiral/metabolismo , Receptores de Superfície Celular , Proteínas Repressoras/genética , Fatores de Transcrição/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Proteínas de Ligação ao Cálcio , Proteínas de Transporte/metabolismo , Cóclea/anormalidades , Cóclea/metabolismo , Anormalidades Congênitas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Homeodomínio/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Proteína Jagged-2 , Ligantes , Camundongos , Camundongos Mutantes , Proteínas/metabolismo , Receptor Notch1 , Receptores Notch , Valores de Referência , Proteínas Repressoras/metabolismo , Proteínas Serrate-Jagged , Fatores de Transcrição HES-1
20.
PLoS One ; 9(2): e88757, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24551154

RESUMO

Hmga2 protein belongs to the non-histone chromosomal high-mobility group (HMG) protein family. HMG proteins have been shown to function as architectural transcription regulators, facilitating enhanceosome formation on a variety of mammalian promoters. Hmga2 are expressed at high levels in embryonic and transformed cells. Terminally differentiated cells, however, have been reported to express only minimal, if any, Hmga2. Our previous affymetrix array data showed that Hmga2 is expressed in the developing and adult mammalian cochleas. However, the spatio-temporal expression pattern of Hmga2 in the murine cochlea remained unknown. In this study, we report the expression of Hmga2 in developing and adult cochleas using immunohistochemistry and quantitative real time PCR analysis. Immunolabeling of Hmga2 in the embryonic, postnatal, and mature cochleas showed broad Hmga2 expression in embryonic cochlea (E14.5) at the level of the developing organ of Corti in differentiating hair cells, supporting cells, in addition to immature cells in the GER and LER areas. By postnatal stage (P0-P3), Hmga2 is predominantly expressed in the hair and supporting cells, in addition to cells in the LER area. By P12, Hmga2 immunolabeling is confined to the hair cells and supporting cells. In the adult ear, Hmga2 expression is maintained in the hair and supporting cell subtypes (i.e. Deiters' cells, Hensen cells, pillar cells, inner phalangeal and border cells) in the cochlear epithelium. Using quantitative real time PCR, we found a decrease in transcript level for Hmga2 comparable to other known inner ear developmental genes (Sox2, Atoh1, Jagged1 and Hes5) in the cochlear epithelium of the adult relative to postnatal ears. These data provide for the first time the tissue-specific expression and transcription level of Hmga2 during inner ear development and suggest its potential dual role in early differentiation and maintenance of both hair and supporting cell phenotypes.


Assuntos
Cóclea/embriologia , Cóclea/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteína HMGA2/metabolismo , Fatores de Transcrição/metabolismo , Animais , Animais Recém-Nascidos , Cóclea/crescimento & desenvolvimento , Regulação para Baixo/genética , Feminino , Proteína HMGA2/genética , Células Ciliadas Auditivas/metabolismo , Masculino , Camundongos , Órgão Espiral/embriologia , Órgão Espiral/crescimento & desenvolvimento , Órgão Espiral/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Células Receptoras Sensoriais/citologia , Células Receptoras Sensoriais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA