Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Mol Psychiatry ; 26(9): 4616-4632, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-32612250

RESUMO

In mammals, most adult neural stem cells (NSCs) are located in the ventricular-subventricular zone (V-SVZ) along the wall of the lateral ventricles and they are the source of olfactory bulb interneurons. Adult NSCs exhibit an apico-basal polarity; they harbor a short apical process and a long basal process, reminiscent of radial glia morphology. In the adult mouse brain, we detected extremely long radial glia-like fibers that originate from the anterior-ventral V-SVZ and that are directed to the ventral striatum. Interestingly, a fraction of adult V-SVZ-derived neuroblasts dispersed in close association with the radial glia-like fibers in the nucleus accumbens (NAc). Using several in vivo mouse models, we show that newborn neurons integrate into preexisting circuits in the NAc where they mature as medium spiny neurons (MSNs), i.e., a type of projection neurons formerly believed to be generated only during embryonic development. Moreover, we found that the number of newborn neurons in the NAc is dynamically regulated by persistent pain, suggesting that adult neurogenesis of MSNs is an experience-modulated process.


Assuntos
Neurogênese , Núcleo Accumbens , Animais , Ventrículos Laterais , Camundongos , Neurônios , Bulbo Olfatório , Dor
3.
Proc Natl Acad Sci U S A ; 111(29): 10726-31, 2014 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-25002477

RESUMO

Postnatal neurogenesis in mammals is confined to restricted brain regions, including the subventricular zone (SVZ). In rodents, the SVZ is a lifelong source of new neurons fated to migrate to the olfactory bulb (OB), where the majority become GABAergic interneurons. The plastic capacity of neonatal and adult SVZ stem/progenitor cells is still largely unknown. By overexpressing the transcription factor Fezf2, a powerful master gene specifying the phenotype of glutamatergic subcerebral projecting neurons, we investigated whether the fate of postnatally generated SVZ neurons can be altered. Following lentiviral delivery of Fezf2 in the neonatal and adult SVZ niche, we showed that ectopic Fezf2 expression is sufficient to redirect the fate of SVZ stem cells. Thus, based on in vivo and in vitro experiments, we provide evidence that numerous Fezf2-positive OB neurons expressed glutamatergic pyramidal cell molecular markers instead of developing a GABAergic identity. Overexpression of Fezf2 had no effect on transit-amplifying progenitors or neuroblasts but was restricted to neural stem cells. Fezf2-respecified neurons bore features of pyramidal cells, exhibiting a larger cell body and a more elaborate dendritic tree, compared with OB granule cells. Patch-clamp recordings further indicated that Fezf2-respecified neurons had synaptic properties and a firing pattern reminiscent of a pyramidal cell-like phenotype. Together, the results demonstrate that neonatal and adult SVZ stem cells retain neuronal fate plasticity.


Assuntos
Diferenciação Celular , Córtex Cerebral/citologia , Ventrículos Cerebrais/citologia , Proteínas de Ligação a DNA/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Potenciais de Ação , Envelhecimento/metabolismo , Animais , Linhagem da Célula , Dendritos/metabolismo , Neurônios GABAérgicos/citologia , Neurônios GABAérgicos/metabolismo , Glutamatos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Fenótipo , Células Piramidais/citologia , Células Piramidais/metabolismo , Sinapses/metabolismo
4.
J Neurosci ; 35(40): 13659-72, 2015 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-26446219

RESUMO

Neuroblast migration is a highly orchestrated process that ensures the proper integration of newborn neurons into complex neuronal circuits. In the postnatal rodent brain, neuroblasts migrate long distances from the subependymal zone of the lateral ventricles to the olfactory bulb (OB) within the rostral migratory stream (RMS). They first migrate tangentially in close contact to each other and later radially as single cells until they reach their final destination in the OB. Sphingosine 1-phosphate (S1P) is a bioactive lipid that interacts with cell-surface receptors to exert different cellular responses. Although well studied in other systems and a target for the treatment of multiple sclerosis, little is known about S1P in the postnatal brain. Here, we report that the S1P receptor 1 (S1P1) is expressed in neuroblasts migrating in the RMS. Using in vivo and in vitro gain- and loss-of-function approaches in both wild-type and transgenic mice, we found that the activation of S1P1 by its natural ligand S1P, acting as a paracrine signal, contributes to maintain neuroblasts attached to each other while they migrate in chains within the RMS. Once in the OB, neuroblasts cease to express S1P1, which results in cell detachment and initiation of radial migration, likely via downregulation of NCAM1 and ß1 integrin. Our results reveal a novel physiological function for S1P1 in the postnatal brain, directing the path followed by newborn neurons in the neurogenic niche. SIGNIFICANCE STATEMENT: The function of each neuron is highly determined by the position it occupies within a neuronal circuit. Frequently, newborn neurons must travel long distances from their birthplace to their predetermined final location and, to do so, they use different modes of migration. In this study, we identify the sphingosine 1-phosphate (S1P) receptor 1 (S1P1) as one of the key players that govern the switch from tangential to radial migration of postnatally generated neuroblasts in the olfactory bulb. Of interest is the evidence that the ligand, S1P, is provided by nearby astrocytes. Finally, we also propose adhesion molecules that act downstream of S1P1 and initiate the transition from tangential chain migration to individual radial migration outside of the stream.


Assuntos
Movimento Celular/genética , Regulação para Baixo/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/fisiologia , Bulbo Olfatório/citologia , Receptores de Lisoesfingolipídeo/metabolismo , Animais , Animais Recém-Nascidos , Antígeno CD56/genética , Antígeno CD56/metabolismo , Caspase 3/metabolismo , Proteínas do Domínio Duplacortina , Células HEK293 , Humanos , Técnicas In Vitro , Integrina beta1/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/genética , Molécula L1 de Adesão de Célula Nervosa/metabolismo , Neuropeptídeos/metabolismo , Técnicas de Cultura de Órgãos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ácidos Siálicos/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo
5.
J Neurosci ; 34(2): 434-45, 2014 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-24403143

RESUMO

The auxiliary subunit α2δ3 modulates the expression and function of voltage-gated calcium channels. Here we show that α2δ3 mRNA is expressed in spiral ganglion neurons and auditory brainstem nuclei and that the protein is required for normal acoustic responses. Genetic deletion of α2δ3 led to impaired auditory processing, with reduced acoustic startle and distorted auditory brainstem responses. α2δ3(-/-) mice learned to discriminate pure tones, but they failed to discriminate temporally structured amplitude-modulated tones. Light and electron microscopy analyses revealed reduced levels of presynaptic Ca(2+) channels and smaller auditory nerve fiber terminals contacting cochlear nucleus bushy cells. Juxtacellular in vivo recordings of sound-evoked activity in α2δ3(-/-) mice demonstrated impaired transmission at these synapses. Together, our results identify a novel role for the α2δ3 auxiliary subunit in the structure and function of specific synapses in the mammalian auditory pathway and in auditory processing disorders.


Assuntos
Transtornos da Percepção Auditiva/metabolismo , Canais de Cálcio/metabolismo , Nervo Coclear/metabolismo , Aprendizagem por Discriminação/fisiologia , Sinapses/metabolismo , Animais , Transtornos da Percepção Auditiva/genética , Transtornos da Percepção Auditiva/fisiopatologia , Tronco Encefálico/metabolismo , Tronco Encefálico/patologia , Canais de Cálcio/genética , Nervo Coclear/patologia , Eletrofisiologia , Potenciais Evocados Auditivos do Tronco Encefálico/fisiologia , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Gânglio Espiral da Cóclea/metabolismo , Gânglio Espiral da Cóclea/fisiologia , Sinapses/patologia , Transmissão Sináptica/fisiologia
6.
Proc Natl Acad Sci U S A ; 109(49): 20107-12, 2012 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-23169657

RESUMO

Connexins have been implicated in the regulation of precursor cell migration and proliferation during embryonic development of the mammalian brain. However, their function in postnatal neurogenesis is unclear. Here we demonstrate that connexin (Cx) 45 is expressed in transit-amplifying cells and neuroblasts in the postnatal subventricular zone (SVZ) and modulated the proliferation of SVZ-derived precursor cells in vivo. Thus, overexpression of Cx45 by retroviral injections increased the proliferation of Mash-1-positive transit-amplifying precursor cells in the SVZ. Conversely, conditional deletion of Cx45 in precursor cells decreased proliferation. Finally, we established that Cx45 positively influences cell cycle reentry via ATP signaling that involves intracellular calcium stores and ERK1/2 signaling.


Assuntos
Conexinas/metabolismo , Ventrículos Laterais/citologia , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Bromodesoxiuridina , Proliferação de Células , Ventrículos Laterais/metabolismo , Camundongos
7.
Hum Mol Genet ; 21(17): 3896-909, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22678062

RESUMO

Hearing impairment represents the most common sensory deficit in humans. Genetic mutations contribute significantly to this disorder. Mostly, only malfunction of the ear is considered. Here, we assessed the role of the peripheral deafness gene Cacna1d, encoding the L-type channel Ca(v)1.3, in downstream processing of acoustic information. To this end, we generated a mouse conditional Cacna1d-eGFP(flex) allele. Upon pairing with Egr2::Cre mice, Ca(v)1.3 was ablated in the auditory brainstem, leaving the inner ear intact. Structural assessment of the superior olivary complex (SOC), an essential auditory brainstem center, revealed a dramatic volume reduction (43-47%) of major nuclei in young adult Egr2::Cre;Cacna1d-eGFP(flex) mice. This volume decline was mainly caused by a reduced cell number (decline by 46-56%). Abnormal formation of the lateral superior olive was already present at P4, demonstrating an essential perinatal role of Ca(v)1.3 in the SOC. Measurements of auditory brainstem responses demonstrated a decreased amplitude in the auditory nerve between 50 and 75 dB stimulation in Egr2::Cre;Cacna1d-eGFP(flex) knockout mice and increased amplitudes in central auditory processing centers. Immunohistochemical studies linked the amplitude changes in the central auditory system to reduced expression of K(v)1.2. No changes were observed for K(v)1.1, KCC2, a determinant of inhibitory neurotransmission, and choline acetyltransferase, a marker of efferent olivocochlear neurons. Together, these analyses identify a crucial retrocochlear role of Ca(v)1.3 and demonstrate that mutations in deafness genes can affect sensory cells and neurons alike. As a corollary, hearing aids have to address central auditory processing deficits as well.


Assuntos
Canais de Cálcio Tipo L/genética , Cóclea/patologia , Surdez/genética , Alelos , Animais , Cóclea/metabolismo , Cruzamentos Genéticos , Surdez/fisiopatologia , Proteína 2 de Resposta de Crescimento Precoce/metabolismo , Potenciais Evocados Auditivos do Tronco Encefálico , Feminino , Deleção de Genes , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Imuno-Histoquímica , Integrases/metabolismo , Masculino , Camundongos , Camundongos Knockout , Núcleo Olivar/metabolismo , Núcleo Olivar/patologia , Núcleo Olivar/fisiopatologia , Superfamília Shaker de Canais de Potássio/metabolismo , Simportadores/metabolismo , Cotransportadores de K e Cl-
8.
J Neurosci ; 32(46): 16345-59, 2012 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-23152617

RESUMO

Mouse chromaffin cells (MCCs) fire spontaneous action potentials (APs) at rest. Ca(v)1.3 L-type calcium channels sustain the pacemaker current, and their loss results in depolarized resting potentials (V(rest)), spike broadening, and remarkable switches into depolarization block after BayK 8644 application. A functional coupling between Ca(v)1.3 and BK channels has been reported but cannot fully account for the aforementioned observations. Here, using Ca(v)1.3(-/-) mice, we investigated the role of Ca(v)1.3 on SK channel activation and how this functional coupling affects the firing patterns induced by sustained current injections. MCCs express SK1-3 channels whose tonic currents are responsible for the slow irregular firing observed at rest. Percentage of frequency increase induced by apamin was found inversely correlated to basal firing frequency. Upon stimulation, MCCs build-up Ca(v)1.3-dependent SK currents during the interspike intervals that lead to a notable degree of spike frequency adaptation (SFA). The major contribution of Ca(v)1.3 to the subthreshold Ca(2+) charge during an AP-train rather than a specific molecular coupling to SK channels accounts for the reduced SFA of Ca(v)1.3(-/-) MCCs. Low adaptation ratios due to reduced SK activation associated with Ca(v)1.3 deficiency prevent the efficient recovery of Na(V) channels from inactivation. This promotes a rapid decline of AP amplitudes and facilitates early onset of depolarization block following prolonged stimulation. Thus, besides serving as pacemaker, Ca(v)1.3 slows down MCC firing by activating SK channels that maintain Na(V) channel availability high enough to preserve stable AP waveforms, even upon high-frequency stimulation of chromaffin cells during stress responses.


Assuntos
Adaptação Fisiológica/fisiologia , Canais de Cálcio Tipo L/fisiologia , Células Cromafins/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Baixa/fisiologia , Potenciais de Ação/fisiologia , Adaptação Fisiológica/efeitos dos fármacos , Animais , Apamina/farmacologia , Cálcio/farmacologia , Canais de Cálcio/efeitos dos fármacos , Canais de Cálcio/fisiologia , Canais de Cálcio Tipo L/efeitos dos fármacos , Células Cromafins/efeitos dos fármacos , DNA Complementar/biossíntese , DNA Complementar/genética , Fenômenos Eletrofisiológicos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Técnicas de Patch-Clamp , RNA/biossíntese , RNA/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real , Canais de Potássio Ativados por Cálcio de Condutância Baixa/efeitos dos fármacos , Canais de Sódio/efeitos dos fármacos
9.
J Neurosci ; 32(25): 8545-53, 2012 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-22723694

RESUMO

The precision of sound information transmitted to the brain depends on the transfer characteristics of the inner hair cell (IHC) ribbon synapse and its multiple contacting auditory fibers. We found that brain derived neurotrophic factor (BDNF) differentially influences IHC characteristics in the intact and injured cochlea. Using conditional knock-out mice (BDNF(Pax2) KO) we found that resting membrane potentials, membrane capacitance and resting linear leak conductance of adult BDNF(Pax2) KO IHCs showed a normal maturation. Likewise, in BDNF(Pax2) KO membrane capacitance (ΔC(m)) as a function of inward calcium current (I(Ca)) follows the linear relationship typical for normal adult IHCs. In contrast the maximal ΔC(m), but not the maximal size of the calcium current, was significantly reduced by 45% in basal but not in apical cochlear turns in BDNF(Pax2) KO IHCs. Maximal ΔC(m) correlated with a loss of IHC ribbons in these cochlear turns and a reduced activity of the auditory nerve (auditory brainstem response wave I). Remarkably, a noise-induced loss of IHC ribbons, followed by reduced activity of the auditory nerve and reduced centrally generated wave II and III observed in control mice, was prevented in equally noise-exposed BDNF(Pax2) KO mice. Data suggest that BDNF expressed in the cochlea is essential for maintenance of adult IHC transmitter release sites and that BDNF upholds opposing afferents in high-frequency turns and scales them down following noise exposure.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/fisiologia , Células Ciliadas Auditivas Internas/fisiologia , Perda Auditiva Provocada por Ruído/genética , Sinapses/fisiologia , Animais , Northern Blotting , Western Blotting , Fator Neurotrófico Derivado do Encéfalo/genética , Contagem de Células , Cóclea/crescimento & desenvolvimento , Cóclea/fisiologia , Potenciais Evocados Auditivos do Tronco Encefálico/fisiologia , Exocitose/genética , Exocitose/fisiologia , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Ruído/efeitos adversos , Emissões Otoacústicas Espontâneas , Fator de Transcrição PAX2/genética , beta-Galactosidase/metabolismo
10.
J Physiol ; 590(20): 5053-73, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22826131

RESUMO

Mouse chromaffin cells (MCCs) express high densities of L-type Ca2+ channels (LTCCs), which control pacemaking activity and catecholamine secretion proportionally to their density of expression. In vivo phosphorylation of LTCCs by cAMP-PKA and cGMP­PKG, regulate LTCC gating in two opposing ways: the cAMP-PKA pathway potentiates while the cGMP­PKG cascade inhibits LTCCs. Despite this, no attempts have been made to answer three key questions related to the two Cav1 isoforms expressed in MCCs (Cav1.2 and Cav1.3): (i) how much are the two Cav1 channels basally modulated by PKA and PKG?, (ii) to what extent can Cav1.2 and Cav1.3 be further regulated by PKA or PKG activation?, and (iii) are the effects of both kinases cumulative when simultaneously active? Here, by comparing the size of L-type currents of wild-type (WT; Cav1.2+Cav1.3) and Cav1.3−/− KO (Cav1.2) MCCs, we provide new evidence that both PKA and PKG pathways affect Cav1.2 and Cav1.3 to the same extent either under basal conditions or induced stimulation. Inhibition of PKA by H89 (5 µM) reduced the L-type current in WT and KO MCCs by∼60%,while inhibition of PKG by KT 5823 (1 µM) increased by∼40% the same current in both cell types. Given that Cav1.2 and Cav1.3 carry the same quantity of Ca2+ currents, this suggests equal sensitivity of Cav1.2 and Cav1.3 to the two basal modulatory pathways. Maximal stimulation of cAMP­PKA by forskolin (100 µM) and activation of cGMP­PKG by pCPT-cGMP (1mM) uncovered a∼25% increase of L-type currents in the first case and∼65% inhibition in the second case in both WT and KO MCCs, suggesting equal sensitivity of Cav1.2 and Cav1.3 during maximal PKA or PKG stimulation. The effects of PKA and PKG were cumulative and most evident when one pathway was activated and the other was inhibited. The two extreme combinations(PKA activation­PKG inhibition vs. PKG activation-PKA inhibition) varied the size of L-type currents by one order of magnitude (from 180% to 18% of control size). Taken together our data suggest that: (i) Cav1.2 and Cav1.3 are equally sensitive to PKA and PKG action under both basal conditions and maximal stimulation, and (ii) PKA and PKG act independently on both Cav1.2 and Cav1.3, producing cumulative effects when opposingly activated. These extreme Cav1 channel modulations may occur either during high-frequency sympathetic stimulation to sustain prolonged catecholamine release (maximal L-type current) or following activation of the NO­cGMP­PKG signalling pathway (minimal L-type current) to limit the steady release of catecholamines.


Assuntos
Canais de Cálcio Tipo L/fisiologia , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , Animais , Células Cultivadas , Células Cromafins/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
11.
Mol Cell Neurosci ; 44(3): 246-59, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20363327

RESUMO

Neurotransmitter release and spontaneous action potentials during cochlear inner hair cell (IHC) development depend on the activity of Ca(v)1.3 voltage-gated L-type Ca(2+) channels. Their voltage- and Ca(2+)-dependent inactivation kinetics are slower than in other tissues but the underlying molecular mechanisms are not yet understood. We found that Rab3-interacting molecule-2alpha (RIM2alpha) mRNA is expressed in immature cochlear IHCs and the protein co-localizes with Ca(v)1.3 in the same presynaptic compartment of IHCs. Expression of RIM proteins in tsA-201 cells revealed binding to the beta-subunit of the channel complex and RIM-induced slowing of both Ca(2+)- and voltage-dependent inactivation of Ca(v)1.3 channels. By inhibiting inactivation, RIM induced a non-inactivating current component typical for IHC Ca(v)1.3 currents which should allow these channels to carry a substantial window current during prolonged depolarizations. These data suggest that RIM2 contributes to the stabilization of Ca(v)1.3 gating kinetics in immature IHCs.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Células Ciliadas Auditivas Internas/fisiologia , Ativação do Canal Iônico/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Isoformas de Proteínas/metabolismo , Proteínas rab3 de Ligação ao GTP/metabolismo , Potenciais de Ação/fisiologia , Processamento Alternativo , Animais , Canais de Cálcio Tipo L/genética , Células Cultivadas , Proteínas de Ligação ao GTP/genética , Células Ciliadas Auditivas Internas/citologia , Humanos , Camundongos , Proteínas do Tecido Nervoso/genética , Técnicas de Patch-Clamp , Isoformas de Proteínas/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Técnicas do Sistema de Duplo-Híbrido , Proteínas rab3 de Ligação ao GTP/genética
12.
Mol Neurobiol ; 53(8): 5607-27, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-26476841

RESUMO

For all sensory organs, the establishment of spatial and temporal cortical resolution is assumed to be initiated by the first sensory experience and a BDNF-dependent increase in intracortical inhibition. To address the potential of cortical BDNF for sound processing, we used mice with a conditional deletion of BDNF in which Cre expression was under the control of the Pax2 or TrkC promoter. BDNF deletion profiles between these mice differ in the organ of Corti (BDNF (Pax2) -KO) versus the auditory cortex and hippocampus (BDNF (TrkC) -KO). We demonstrate that BDNF (Pax2) -KO but not BDNF (TrkC) -KO mice exhibit reduced sound-evoked suprathreshold ABR waves at the level of the auditory nerve (wave I) and inferior colliculus (IC) (wave IV), indicating that BDNF in lower brain regions but not in the auditory cortex improves sound sensitivity during hearing onset. Extracellular recording of IC neurons of BDNF (Pax2) mutant mice revealed that the reduced sensitivity of auditory fibers in these mice went hand in hand with elevated thresholds, reduced dynamic range, prolonged latency, and increased inhibitory strength in IC neurons. Reduced parvalbumin-positive contacts were found in the ascending auditory circuit, including the auditory cortex and hippocampus of BDNF (Pax2) -KO, but not of BDNF (TrkC) -KO mice. Also, BDNF (Pax2) -WT but not BDNF (Pax2) -KO mice did lose basal inhibitory strength in IC neurons after acoustic trauma. These findings suggest that BDNF in the lower parts of the auditory system drives auditory fidelity along the entire ascending pathway up to the cortex by increasing inhibitory strength in behaviorally relevant frequency regions. Fidelity and inhibitory strength can be lost following auditory nerve injury leading to diminished sensory outcome and increased central noise.


Assuntos
Córtex Auditivo/patologia , Córtex Auditivo/fisiopatologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Ruído , Animais , Córtex Auditivo/metabolismo , Limiar Auditivo , Cóclea/metabolismo , Potenciais Evocados Auditivos do Tronco Encefálico , Deleção de Genes , Audição , Colículos Inferiores/patologia , Colículos Inferiores/fisiopatologia , Integrases/metabolismo , Camundongos Knockout , Regiões Promotoras Genéticas/genética , Receptor trkC/metabolismo , Fatores de Risco
13.
PLoS One ; 8(3): e57247, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23516401

RESUMO

Tinnitus is proposed to be caused by decreased central input from the cochlea, followed by increased spontaneous and evoked subcortical activity that is interpreted as compensation for increased responsiveness of central auditory circuits. We compared equally noise exposed rats separated into groups with and without tinnitus for differences in brain responsiveness relative to the degree of deafferentation in the periphery. We analyzed (1) the number of CtBP2/RIBEYE-positive particles in ribbon synapses of the inner hair cell (IHC) as a measure for deafferentation; (2) the fine structure of the amplitudes of auditory brainstem responses (ABR) reflecting differences in sound responses following decreased auditory nerve activity and (3) the expression of the activity-regulated gene Arc in the auditory cortex (AC) to identify long-lasting central activity following sensory deprivation. Following moderate trauma, 30% of animals exhibited tinnitus, similar to the tinnitus prevalence among hearing impaired humans. Although both tinnitus and no-tinnitus animals exhibited a reduced ABR wave I amplitude (generated by primary auditory nerve fibers), IHCs ribbon loss and high-frequency hearing impairment was more severe in tinnitus animals, associated with significantly reduced amplitudes of the more centrally generated wave IV and V and less intense staining of Arc mRNA and protein in the AC. The observed severe IHCs ribbon loss, the minimal restoration of ABR wave size, and reduced cortical Arc expression suggest that tinnitus is linked to a failure to adapt central circuits to reduced cochlear input.


Assuntos
Adaptação Fisiológica , Cóclea/fisiopatologia , Potenciais Evocados Auditivos do Tronco Encefálico , Ruído/efeitos adversos , Zumbido/etiologia , Animais , Córtex Auditivo/metabolismo , Limiar Auditivo , Comportamento Animal , Cóclea/metabolismo , Proteínas do Citoesqueleto/metabolismo , Feminino , Células Ciliadas Auditivas Internas/metabolismo , Perda Auditiva Provocada por Ruído/fisiopatologia , Imuno-Histoquímica , Proteínas do Tecido Nervoso/metabolismo , Ratos , Zumbido/metabolismo
14.
Mol Neurobiol ; 47(1): 261-79, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23154938

RESUMO

Increasing evidence shows that hearing loss is a risk factor for tinnitus and hyperacusis. Although both often coincide, a causal relationship between tinnitus and hyperacusis has not been shown. Currently, tinnitus and hyperacusis are assumed to be caused by elevated responsiveness in subcortical circuits. We examined both the impact of different degrees of cochlear damage and the influence of stress priming on tinnitus induction. We used (1) a behavioral animal model for tinnitus designed to minimize stress, (2) ribbon synapses in inner hair cells (IHCs) as a measure for deafferentation, (3) the integrity of auditory brainstem responses (ABR) to detect differences in stimulus-evoked neuronal activity, (4) the expression of the activity-regulated cytoskeletal protein, Arc, to identify long-lasting changes in network activity within the basolateral amygdala (BLA), hippocampal CA1, and auditory cortex (AC), and (5) stress priming to investigate the influence of corticosteroid on trauma-induced brain responses. We observed that IHC ribbon loss (deafferentation) leads to tinnitus when ABR functions remain reduced and Arc is not mobilized in the hippocampal CA1 and AC. If, however, ABR waves are functionally restored and Arc is mobilized, tinnitus does not occur. Both central response patterns were found to be independent of a profound threshold loss and could be shifted by the corticosterone level at the time of trauma. We, therefore, discuss the findings in the context of a history of stress that can trigger either an adaptive or nonadaptive brain response following injury.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Células Ciliadas Auditivas Internas/patologia , Proteínas do Tecido Nervoso/metabolismo , Ruído/efeitos adversos , Zumbido/metabolismo , Zumbido/patologia , Estimulação Acústica , Animais , Córtex Auditivo/metabolismo , Córtex Auditivo/patologia , Córtex Auditivo/fisiopatologia , Limiar Auditivo , Proteínas do Citoesqueleto/genética , Potenciais Evocados Auditivos do Tronco Encefálico , Feminino , Células Ciliadas Auditivas Internas/metabolismo , Perda Auditiva/complicações , Perda Auditiva/metabolismo , Perda Auditiva/patologia , Perda Auditiva/fisiopatologia , Modelos Biológicos , Proteínas do Tecido Nervoso/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Estresse Psicológico/complicações , Estresse Psicológico/patologia , Estresse Psicológico/fisiopatologia , Zumbido/complicações , Zumbido/fisiopatologia
15.
Front Mol Neurosci ; 6: 20, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23950737

RESUMO

Voltage-gated L-type Ca(2+) channels (L-VGCCs) like CaV1.2 are assumed to play a crucial role for controlling release of trophic peptides including brain-derived neurotrophic factor (BDNF). In the inner ear of the adult mouse, besides the well-described L-VGCC CaV1.3, CaV1.2 is also expressed. Due to lethality of constitutive CaV1.2 knock-out mice, the function of this ion channel as well as its putative relationship to BDNF in the auditory system is entirely elusive. We recently described that BDNF plays a differential role for inner hair cell (IHC) vesicles release in normal and traumatized condition. To elucidate a presumptive role of CaV1.2 during this process, two tissue-specific conditional mouse lines were generated. To distinguish the impact of CaV1.2 on the cochlea from that on feedback loops from higher auditory centers CaV1.2 was deleted, in one mouse line, under the Pax2 promoter (CaV1.2(Pax2)) leading to a deletion in the spiral ganglion neurons, dorsal cochlear nucleus, and inferior colliculus. In the second mouse line, the Egr2 promoter was used for deleting CaV1.2 (CaV1.2(Egr2)) in auditory brainstem nuclei. In both mouse lines, normal hearing threshold and equal number of IHC release sites were observed. We found a slight reduction of auditory brainstem response wave I amplitudes in the CaV1.2(Pax2) mice, but not in the CaV1.2(Egr2) mice. After noise exposure, CaV1.2(Pax2) mice had less-pronounced hearing loss that correlated with maintenance of ribbons in IHCs and less reduced activity in auditory nerve fibers, as well as in higher brain centers at supra-threshold sound stimulation. As reduced cochlear BDNF mRNA levels were found in CaV1.2(Pax2) mice, we suggest that a CaV1.2-dependent step may participate in triggering part of the beneficial and deteriorating effects of cochlear BDNF in intact systems and during noise exposure through a pathway that is independent of CaV1.2 function in efferent circuits.

16.
Cell Stem Cell ; 10(1): 76-87, 2012 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-22226357

RESUMO

The subventricular zone (SVZ) of the lateral ventricles is the largest neurogenic niche of the postnatal brain. New SVZ-generated neurons migrate via the rostral migratory stream to the olfactory bulb (OB) where they functionally integrate into preexisting neuronal circuits. Nonsynaptic GABA signaling was previously shown to inhibit SVZ-derived neurogenesis. Here we identify the endogenous protein diazepam binding inhibitor (DBI) as a positive modulator of SVZ postnatal neurogenesis by regulating GABA activity in transit-amplifying cells. We performed DBI loss- and gain-of-function experiments in vivo at the peak of postnatal OB neuron generation in mice and demonstrate that DBI enhances proliferation by preventing SVZ progenitors to exit the cell cycle. Furthermore, we provide evidence that DBI exerts its effect on SVZ progenitors via its octadecaneuropeptide proteolytic product (ODN) by inhibiting GABA-induced currents. Together our data reveal a regulatory mechanism by which DBI counteracts the inhibitory effect of nonsynaptic GABA signaling on subventricular neuronal proliferation.


Assuntos
Ciclo Celular/fisiologia , Inibidor da Ligação a Diazepam/metabolismo , Neurônios GABAérgicos/metabolismo , Neuropeptídeos/metabolismo , Bulbo Olfatório/metabolismo , Fragmentos de Peptídeos/metabolismo , Transdução de Sinais/fisiologia , Células-Tronco/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Neurônios GABAérgicos/citologia , Camundongos , Camundongos Transgênicos , Bulbo Olfatório/citologia , Células-Tronco/citologia , Ácido gama-Aminobutírico/genética
17.
Trends Pharmacol Sci ; 32(6): 366-75, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21450352

RESUMO

Within the family of voltage-gated calcium channels (VGCCs), L-type channels (L-VGCCs) represent a well-established therapeutic target for calcium channel blockers, which are widely used to treat hypertension and myocardial ischemia. L-VGCCs outside the cardiovascular system also control key physiological processes such as neuronal plasticity, sensory cell function (e.g. in the inner ear and retina) and endocrine function (e.g. in pancreatic beta cells and adrenal chromaffin cells). Research into L-VGCCs was stimulated by the discovery that the known L-VGCC isoforms (Ca(V)1.1, Ca(V)1.2, Ca(V)1.3 and Ca(V)1.4) possess different biophysical properties. However, no L-VGCC-isoform-selective drugs have yet been identified. In this review, we examine Ca(V)1.2 and Ca(V)1.3 isoforms at the level of genetic structure, splice variants, post-translational modifications and functional protein coupling. We discuss candidate Ca(V)1.2- and Ca(V)1.3-specific characteristics as future therapeutic targets in individual organs.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L , Proteínas do Tecido Nervoso/fisiologia , Processamento Alternativo , Sequência de Aminoácidos , Animais , Cálcio/metabolismo , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Regulação da Expressão Gênica , Humanos , Camundongos , Proteínas do Tecido Nervoso/genética , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Subunidades Proteicas , Alinhamento de Sequência , Homologia de Sequência , Transdução de Sinais
18.
Mol Neurobiol ; 42(3): 185-98, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21088933

RESUMO

L-type Ca(2+) channels (LTCCs, Ca(v)1) open readily during membrane depolarization and allow Ca(2+) to enter the cell. In this way, LTCCs regulate cell excitability and trigger a variety of Ca(2+)-dependent physiological processes such as: excitation-contraction coupling in muscle cells, gene expression, synaptic plasticity, neuronal differentiation, hormone secretion, and pacemaker activity in heart, neurons, and endocrine cells. Among the two major isoforms of LTCCs expressed in excitable tissues (Ca(v)1.2 and Ca(v)1.3), Ca(v)1.3 appears suitable for supporting a pacemaker current in spontaneously firing cells. It has steep voltage dependence and low threshold of activation and inactivates slowly. Using Ca(v)1.3(-/-) KO mice and membrane current recording techniques such as the dynamic and the action potential clamp, it has been possible to resolve the time course of Ca(v)1.3 pacemaker currents that regulate the spontaneous firing of dopaminergic neurons and adrenal chromaffin cells. In several cell types, Ca(v)1.3 is selectively coupled to BK channels within membrane nanodomains and controls both the firing frequency and the action potential repolarization phase. Here we review the most critical aspects of Ca(v)1.3 channel gating and its coupling to large conductance BK channels recently discovered in spontaneously firing neurons and neuroendocrine cells with the aim of furnishing a converging view of the role that these two channel types play in the regulation of cell excitability.


Assuntos
Potenciais de Ação/fisiologia , Canais de Cálcio Tipo L/metabolismo , Ativação do Canal Iônico/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Animais , Relógios Biológicos/fisiologia , Canais de Cálcio Tipo L/genética , Células Cromafins/fisiologia , Ritmo Circadiano/fisiologia , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Alta/genética , Camundongos , Camundongos Knockout , Miocárdio/citologia , Miocárdio/metabolismo , Técnicas de Patch-Clamp , Núcleo Supraquiasmático/metabolismo
19.
Gene ; 430(1-2): 17-29, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19000751

RESUMO

RACK1 (Receptor for Activated C Kinase 1) is a scaffold protein for different kinases and membrane receptors. Previously, we characterized an age-dependent decline of RACK1 protein expression which could be counteracted with DHEA (dehydroepiandrosterone) [Corsini, E., et al. 2002. In vivo dehydroepiandrosterone restores age-associated defects in the protein kinase C signal transduction pathway and related functional responses. J. Immunol. 168, 1753-1758. and Corsini, E., et al. 2005. Age-related decline in RACK-1 expression in human leukocytes is correlated to plasma levels of dehydroepiandrosterone. J. Leukoc. Biol. 77, 247-256.]. Hypothesizing a direct control of RACK1 expression by DHEA we studied the not yet characterized human promoter region of its coding gene GNB2L1. The FLOE (Fluorescently Labeled Oligonucleotide Extension) was used to map the transcription start site and a novel Gateway luciferase vector (GW luc basic; Del Vecchio, I., Zuccotti, A., Canneva, F., Lenzken, S.C., Racchi, M., 2007. Development of the first Gateway firefly luciferase vector and use of reverse transcriptase in FLOE (Fluorescently Labeled Oligonucleotide Extension) reactions. Plasmid 58, 269-274.) to obtain promoter region mutants. Human SH-SY5Y, THP1 and lymphoblastoid cells were used for transient transfections and treatments with lipopolysaccharide (LPS), phorbol myristate acetate (PMA), DHEA and cortisol (the first two molecules to differently activate NF-kB, a transcription complex able to regulate the murine Gnb2l1 gene expression, whereas DHEA and cortisol since they are known to be imbalanced during the aging and possess counteracting actions on the immune function). The primer extension demonstrated the existence of two alternative start sites of transcription respectively located at about 230 and 300 nt 5' of the Genbank mRNA entry for GNB2L1. Moreover, as a result of the luciferase study we were able to demonstrate that a little region of approximately 300 nt conserved sufficient elements for reporter expression. We also reported that the DHEA modulation of GNB2L1 endogenous expression could not be recapitulated with the luciferase assays. Indeed, the promoter was significantly modulated by means of LPS and PMA treatments but not using DHEA. Differently the use of cortisol led us to demonstrate a biologically significant decrease of luciferase activity only in the presence of a binding site for nuclear receptors of glucocorticoids. Interestingly, other binding sites for transcriptional factors were identified in silico: different c-Rel (NF-kB) and some cardiomyocitic specific cis-acting elements. All this data suggest that the DHEA mediated GNB2L1 regulation is modulated by distant elements (enhancers/silencers), whereas LPS, PMA and cortisol effect can act directly on the mapped GNB2L1 promoter. In conclusion we hypothesize that the imbalance between DHEA and cortisol during aging could be important in the previously demonstrated recovery of the RACK1 expression.


Assuntos
Mapeamento Cromossômico , Proteínas de Ligação ao GTP/genética , Proteínas de Neoplasias/genética , Fases de Leitura Aberta/genética , Regiões Promotoras Genéticas/genética , Receptores de Superfície Celular/genética , Sequência de Bases , Linhagem Celular , Biologia Computacional , Primers do DNA/metabolismo , Desidroepiandrosterona/farmacologia , Corantes Fluorescentes/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Hidrocortisona/farmacologia , Lipopolissacarídeos/farmacologia , Luciferases/metabolismo , Dados de Sequência Molecular , Proteínas de Neoplasias/metabolismo , Estabilidade Proteica/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Quinase C Ativada , Receptores de Superfície Celular/metabolismo , Deleção de Sequência , Acetato de Tetradecanoilforbol/farmacologia , Sítio de Iniciação de Transcrição , Transcrição Gênica/efeitos dos fármacos
20.
Plasmid ; 58(3): 269-74, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17707908

RESUMO

To study promoters we usually use primer extension to map the transcription start site and a panel of PCR generated deletion mutants. This strategy is complex and time-consuming. Therefore, we decided to improve it by using Gateway and FLOE (Fluorescently Labeled Oligonucleotide Extension). In this report we developed the first luciferase reporter "destination vector" (GW luc basic) for the Gateway technology and tested its efficacy, accuracy and background level by transfecting two distant cell lines (THP1 monocytic and SH-SY5Y neural cells). This vector is a real advantage for the cloning of many PCR fragments and sustains reporter activity also in THP1 cells, which are known to be problematic for transfection/expression. FLOE is a straightforward method to map transcription start sites but a bias in the capillary electrophoretic migration pattern of ROX weight markers has been reported: ROX markers migrated as if they were some bp longer. We hypothesized that this could depend on the use of different enzymes for the two principal reactions (DNA polymerase for the dideoxy chain terminated reaction on DNA and reverse transcriptase for the primer extension on RNA). Therefore, we used the same reverse transcriptase enzyme on both reactions, demonstrating that the reported bias is not due to the use of different enzymes but is an intrinsic feature of the ROX markers. The proposed procedure is important not only because of the timeliness but also for the global impact on the study of the first layer of the gene regulation.


Assuntos
Vetores Genéticos , Luciferases de Vaga-Lume/genética , Regiões Promotoras Genéticas/genética , DNA Polimerase Dirigida por RNA/metabolismo , Transfecção , Animais , Linhagem Celular , Vaga-Lumes , Regulação da Expressão Gênica , Luciferases de Vaga-Lume/metabolismo , Dados de Sequência Molecular , Oligonucleotídeos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA