Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(4): e2212338120, 2023 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-36649421

RESUMO

To fertilize an oocyte, the membrane potential of both mouse and human sperm must hyperpolarize (become more negative inside). Determining the molecular mechanisms underlying this hyperpolarization is vital for developing new contraceptive methods and detecting causes of idiopathic male infertility. In mouse sperm, hyperpolarization is caused by activation of the sperm-specific potassium (K+) channel SLO3 [C. M. Santi et al., FEBS Lett. 584, 1041-1046 (2010)]. In human sperm, it has long been unclear whether hyperpolarization depends on SLO3 or the ubiquitous K+ channel SLO1 [N. Mannowetz, N. M. Naidoo, S. A. S. Choo, J. F. Smith, P. V. Lishko, Elife 2, e01009 (2013), C. Brenker et al., Elife 3, e01438 (2014), and S. A. Mansell, S. J. Publicover, C. L. R. Barratt, S. M. Wilson, Mol. Hum. Reprod. 20, 392-408 (2014)]. In this work, we identified the first selective inhibitor for human SLO3-VU0546110-and showed that it completely blocked heterologous SLO3 currents and endogenous K+ currents in human sperm. This compound also prevented sperm from hyperpolarizing and undergoing hyperactivated motility and induced acrosome reaction, which are necessary to fertilize an egg. We conclude that SLO3 is the sole K+ channel responsible for hyperpolarization and significantly contributes to the fertilizing ability of human sperm. Moreover, SLO3 is a good candidate for contraceptive development, and mutation of this gene is a possible cause of idiopathic male infertility.


Assuntos
Infertilidade Masculina , Canais de Potássio Ativados por Cálcio de Condutância Alta , Humanos , Masculino , Canais de Potássio Ativados por Cálcio de Condutância Alta/antagonistas & inibidores , Potenciais da Membrana/fisiologia , Sêmen , Espermatozoides/fisiologia
2.
Mol Pharmacol ; 101(5): 357-370, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35246480

RESUMO

Heteromeric Kir4.1/Kir5.1 (KCNJ10/KCNJ16) inward rectifier potassium (Kir) channels play key roles in the brain and kidney, but pharmacological tools for probing their physiology and therapeutic potential have not been developed. Here, we report the discovery, in a high-throughput screening of 80,475 compounds, of the moderately potent and selective inhibitor VU0493690, which we selected for characterization and chemical optimization. VU0493690 concentration-dependently inhibits Kir4.1/5.1 with an IC50 of 0.96 µM and exhibits at least 10-fold selectivity over Kir4.1 and ten other Kir channels. Multidimensional chemical optimization of VU0493690 led to the development of VU6036720, the most potent (IC50 = 0.24 µM) and selective (>40-fold over Kir4.1) Kir4.1/5.1 inhibitor reported to date. Cell-attached patch single-channel recordings revealed that VU6036720 inhibits Kir4.1/5.1 activity through a reduction of channel open-state probability and single-channel current amplitude. Elevating extracellular potassium ion by 20 mM shifted the IC50 6.8-fold, suggesting that VU6036720 is a pore blocker that binds in the ion-conduction pathway. Mutation of the "rectification controller" asparagine 161 to glutamate (N161E), which is equivalent to small-molecule binding sites in other Kir channels, led to a strong reduction of inhibition by VU6036720. Renal clearance studies in mice failed to show a diuretic response that would be consistent with inhibition of Kir4.1/5.1 in the renal tubule. Drug metabolism and pharmacokinetics profiling revealed that high VU6036720 clearance and plasma protein binding may prevent target engagement in vivo. In conclusion, VU6036720 represents the current state-of-the-art Kir4.1/5.1 inhibitor that should be useful for probing the functions of Kir4.1/5.1 in vitro and ex vivo. SIGNIFICANCE STATEMENT: Heteromeric inward rectifier potassium (Kir) channels comprising Kir4.1 and Kir5.1 subunits play important roles in renal and neural physiology and may represent inhibitory drug targets for hypertension and edema. Herein, we employ high-throughput compound library screening, patch clamp electrophysiology, and medicinal chemistry to develop and characterize the first potent and specific in vitro inhibitor of Kir4.1/5.1, VU6036720, which provides proof-of-concept that drug-like inhibitors of this channel may be developed.


Assuntos
Canais de Potássio Corretores do Fluxo de Internalização , Animais , Biblioteca Gênica , Ensaios de Triagem em Larga Escala , Camundongos , Potássio/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo
3.
Mol Psychiatry ; 26(9): 4670-4686, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33558651

RESUMO

Genetic abnormalities in synaptic proteins are common in individuals with autism; however, our understanding of the cellular and molecular mechanisms disrupted by these abnormalities is limited. SHANK3 is a postsynaptic scaffolding protein of excitatory synapses that has been found mutated or deleted in most patients with 22q13 deletion syndrome and about 2% of individuals with idiopathic autism and intellectual disability. Here, we generated CRISPR/Cas9-engineered human pluripotent stem cells (PSCs) with complete hemizygous SHANK3 deletion (SHANK3+/-), which is the most common genetic abnormality in patients, and investigated the synaptic and morphological properties of SHANK3-deficient PSC-derived cortical neurons engrafted in the mouse prefrontal cortex. We show that human PSC-derived neurons integrate into the mouse cortex by acquiring appropriate cortical layer identities and by receiving and sending anatomical projections from/to multiple different brain regions. We also demonstrate that SHANK3-deficient human neurons have reduced AMPA-, but not NMDA- or GABA-mediated synaptic transmission and exhibit impaired dendritic arbors and spines, as compared to isogenic control neurons co-engrafted in the same brain region. Together, this study reveals specific synaptic and morphological deficits caused by SHANK3 hemizygosity in human cortical neurons at different developmental stages under physiological conditions and validates the use of co-engrafted control and mutant human neurons as a new platform for studying connectivity deficits in genetic neurodevelopmental disorders associated with autism.


Assuntos
Proteínas do Tecido Nervoso , Transmissão Sináptica , Animais , Humanos , Camundongos , Proteínas dos Microfilamentos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Córtex Pré-Frontal/metabolismo , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico
4.
Elife ; 92020 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-33206596

RESUMO

The nucleus of the solitary tract (NTS) is critical for the central integration of signals from visceral organs and contains preproglucagon (PPG) neurons, which express leptin receptors in the mouse and send direct projections to the paraventricular nucleus of the hypothalamus (PVH). Here, we visualized projections of PPG neurons in leptin-deficient Lepob/ob mice and found that projections from PPG neurons are elevated compared with controls, and PPG projections were normalized by targeted rescue of leptin receptors in LepRbTB/TB mice, which lack functional neuronal leptin receptors. Moreover, Lepob/ob and LepRbTB/TB mice displayed increased levels of neuronal activation in the PVH following vagal stimulation, and whole-cell patch recordings of GLP-1 receptor-expressing PVH neurons revealed enhanced excitatory neurotransmission, suggesting that leptin acts cell autonomously to suppress representation of excitatory afferents from PPG neurons, thereby diminishing the impact of visceral sensory information on GLP-1 receptor-expressing neurons in the PVH.


Assuntos
Peptídeo 1 Semelhante ao Glucagon/metabolismo , Leptina/metabolismo , Núcleo Hipotalâmico Paraventricular/crescimento & desenvolvimento , Núcleo Hipotalâmico Paraventricular/metabolismo , Animais , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Proglucagon/metabolismo , Receptores para Leptina/genética , Receptores para Leptina/metabolismo , Núcleo Solitário/metabolismo
5.
ACS Chem Neurosci ; 11(21): 3658-3671, 2020 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-33143429

RESUMO

Malignant migrating partial seizures of infancy is a rare, devastating form of epilepsy most commonly associated with gain-of-function mutations in the potassium channel, Slack. Not only is this condition almost completely pharmacoresistant, there are not even selective drug-like tools available to evaluate whether inhibition of these overactivated, mutant Slack channels may represent a viable path forward toward new antiepileptic therapies. Therefore, we used a high-throughput thallium flux assay to screen a drug-like, 100 000-compound library in search of inhibitors of both wild-type and a disease-associated mutant Slack channel. Using this approach, we discovered VU0606170, a selective Slack channel inhibitor with low micromolar potency. Critically, VU0606170 also proved effective at significantly decreasing the firing rate in overexcited, spontaneously firing cortical neuron cultures. Taken together, our data provide compelling evidence that selective inhibition of Slack channel activity can be achieved with small molecules and that inhibition of Slack channel activity in neurons produces efficacy consistent with an antiepileptic effect. Thus, the identification of VU0606170 provides a much-needed tool for advancing our understanding of the role of the Slack channel in normal physiology and disease as well as its potential as a target for therapeutic intervention.


Assuntos
Sinalização do Cálcio , Proteínas do Tecido Nervoso , Canais de Potássio Ativados por Sódio , Células Cultivadas , Células HEK293 , Humanos , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Canais de Potássio Ativados por Sódio/antagonistas & inibidores , Canais de Potássio Ativados por Sódio/metabolismo
6.
Neurobiol Dis ; 127: 449-461, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30885793

RESUMO

The amyloid precursor protein (APP) has been extensively studied because of its association with Alzheimer's disease (AD). However, APP distribution across different subcellular membrane compartments and its function in neurons remains unclear. We generated an APP fusion protein with a pH-sensitive green fluorescent protein at its ectodomain and a pH-insensitive blue fluorescent protein at its cytosolic domain and used it to measure APP's distribution, subcellular trafficking, and cleavage in live neurons. This reporter, closely resembling endogenous APP, revealed only a limited correlation between synaptic activities and APP trafficking. However, the synaptic surface fraction of APP was increased by a reduction in membrane cholesterol levels, a phenomenon that involves APP's cholesterol-binding motif. Mutations at or near binding sites not only reduced both the surface fraction of APP and membrane cholesterol levels in a dominant negative manner, but also increased synaptic vulnerability to moderate membrane cholesterol reduction. Our results reveal reciprocal modulation of APP and membrane cholesterol levels at synaptic boutons.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Colesterol/metabolismo , Neurônios/metabolismo , Sinapses/metabolismo , Membranas Sinápticas/metabolismo , Doença de Alzheimer/metabolismo , Animais , Membrana Celular/metabolismo , Hipocampo/metabolismo , Imagem Óptica , Transporte Proteico/fisiologia , Ratos
7.
Bio Protoc ; 8(2)2018 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-29552593

RESUMO

Release and recycling of synaptic vesicles are essential for neurotransmission and synaptic plasticity. To gain mechanistic understanding of these processes, direct measurements of vesicle release and retrieval is indispensable. Styryl dyes like FM1-43 and FM4-64 have been widely used for this purpose and their loading and unloading are reliable measurements for synaptic vesicle release and retrieval in cultured neurons. This protocol describes in detail the procedure of using styryl dyes to label and measure synaptic vesicle uptake and release in cultured rat hippocampal neurons. We also include a brief description of hippocampal culture. In the end, we briefly discuss the commonality and difference among FM dye, pH-sensitive fluorescent proteins and quantum dots in terms of measuring synaptic vesicle behavior.

8.
Nat Commun ; 9(1): 796, 2018 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-29476054

RESUMO

Graphene possesses extraordinary properties that promise great potential in biomedicine. However, fully leveraging these properties requires close contact with the cell surface, raising the concern of unexpected biological consequences. Computational models have demonstrated that graphene preferentially interacts with cholesterol, a multifunctional lipid unique to eukaryotic membranes. Here we demonstrate an interaction between graphene and cholesterol. We find that graphene increases cell membrane cholesterol and potentiates neurotransmission, which is mediated by increases in the number, release probability, and recycling rate of synaptic vesicles. In fibroblasts grown on graphene, we also find an increase in cholesterol, which promotes the activation of P2Y receptors, a family of receptor regulated by cholesterol. In both cases, direct manipulation of cholesterol levels elucidates that a graphene-induced cholesterol increase underlies the observed potentiation of each cell signaling pathway. These findings identify cholesterol as a mediator of graphene's cellular effects, providing insight into the biological impact of graphene.


Assuntos
Membrana Celular/metabolismo , Colesterol/metabolismo , Grafite/química , Animais , Membrana Celular/química , Eletrofisiologia , Feminino , Fibroblastos/química , Fibroblastos/metabolismo , Grafite/metabolismo , Masculino , Camundongos , Células NIH 3T3 , Neurônios/química , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Purinérgicos P2Y/metabolismo
9.
Sci Rep ; 7(1): 5061, 2017 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-28698583

RESUMO

Genetically encoded pH-sensors are widely used in studying cell membrane trafficking and membrane protein turnover because they render exo-/endocytosis-associated pH changes to fluorescent signals. For imaging and analysis purposes, high concentration ammonium chloride is routinely used to alkalize intracellular membrane compartments under the assumption that it does not cause long-term effects on cellular processes being studied like neurotransmission. However, pathological studies about hyperammonemia have shown that ammonium is toxic to brain cells especially astrocytes and neurons. Here, we focus on ammonium's physiological impacts on neurons including membrane potential, cytosolic Ca2+ and synaptic vesicles. We have found that extracellularly applied ammonium chloride as low as 5 mM causes intracellular Ca2+-increase and a reduction of vesicle release even after washout. The often-used 50 mM ammonium chloride causes more extensive and persistent changes, including membrane depolarization, prolonged elevation of intracellular Ca2+ and diminution of releasable synaptic vesicles. Our findings not only help to bridge the discrepancies in previous studies about synaptic vesicle release using those pH-sensors or other vesicle specific reporters, but also suggest an intriguing relationship between intracellular pH and neurotransmission.


Assuntos
Cloreto de Amônio/farmacologia , Neurônios/fisiologia , Vesículas Sinápticas/metabolismo , Animais , Cálcio/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Camundongos , Neurônios/efeitos dos fármacos , Ratos Sprague-Dawley , Vesículas Sinápticas/efeitos dos fármacos
10.
Science ; 348(6240): 1255-60, 2015 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-26068853

RESUMO

Blood gas and tissue pH regulation depend on the ability of the brain to sense CO2 and/or H(+) and alter breathing appropriately, a homeostatic process called central respiratory chemosensitivity. We show that selective expression of the proton-activated receptor GPR4 in chemosensory neurons of the mouse retrotrapezoid nucleus (RTN) is required for CO2-stimulated breathing. Genetic deletion of GPR4 disrupted acidosis-dependent activation of RTN neurons, increased apnea frequency, and blunted ventilatory responses to CO2. Reintroduction of GPR4 into RTN neurons restored CO2-dependent RTN neuronal activation and rescued the ventilatory phenotype. Additional elimination of TASK-2 (K(2P)5), a pH-sensitive K(+) channel expressed in RTN neurons, essentially abolished the ventilatory response to CO2. The data identify GPR4 and TASK-2 as distinct, parallel, and essential central mediators of respiratory chemosensitivity.


Assuntos
Dióxido de Carbono/fisiologia , Canais de Potássio de Domínios Poros em Tandem/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Respiração , Corpo Trapezoide/fisiologia , Acidose Respiratória/genética , Acidose Respiratória/fisiopatologia , Animais , Feminino , Deleção de Genes , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Neurônios/metabolismo , Neurônios/fisiologia , Canais de Potássio de Domínios Poros em Tandem/genética , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/genética , Corpo Trapezoide/citologia , Corpo Trapezoide/metabolismo
11.
Stem Cells Transl Med ; 4(8): 887-93, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26025981

RESUMO

The exocytotic release of dopamine is one of the most characteristic but also one of the least appreciated processes in dopaminergic neurotransmission. Fluorescence imaging has yielded rich information about the properties of synaptic vesicles and the release of neurotransmitters in excitatory and inhibitory neurons. In contrast, imaging-based studies for in-depth understanding of synaptic vesicle behavior in dopamine neurons are lagging largely because of a lack of suitable preparations. Midbrain culture has been one of the most valuable preparations for the subcellular investigation of dopaminergic transmission; however, the paucity and fragility of cultured dopaminergic neurons limits their use for live cell imaging. Recent developments in stem cell technology have led to the successful production of dopamine neurons from embryonic or induced pluripotent stem cells. Although the dopaminergic identity of these stem cell-derived neurons has been characterized in different ways, vesicle-mediated dopamine release from their axonal terminals has been barely assessed. We report a more efficient procedure to reliably generate dopamine neurons from embryonic stem cells, and it yields more dopamine neurons with more dopaminergic axon projections than midbrain culture does. Using a collection of functional measurements, we show that stem cell-derived dopamine neurons are indistinguishable from those in midbrain culture. Taking advantage of this new preparation, we simultaneously tracked the turnover of hundreds of synaptic vesicles individually using pH-sensitive quantum dots. By doing so, we revealed distinct fusion kinetics of the dopamine-secreting vesicles, which is consistent within both preparations.


Assuntos
Dopamina/metabolismo , Células-Tronco Embrionárias/citologia , Transmissão Sináptica , Vesículas Sinápticas/metabolismo , Animais , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Células-Tronco Embrionárias/metabolismo , Mesencéfalo/citologia , Camundongos , Neurotransmissores/metabolismo , Vesículas Sinápticas/patologia
12.
J Gen Physiol ; 139(3): 219-34, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22371363

RESUMO

Here, we describe a new mechanism by which glutamate (Glu) and trace metals reciprocally modulate activity of the Ca(v)2.3 channel by profoundly shifting its voltage-dependent gating. We show that zinc and copper, at physiologically relevant concentrations, occupy an extracellular binding site on the surface of Ca(v)2.3 and hold the threshold for activation of these channels in a depolarized voltage range. Abolishing this binding by chelation or the substitution of key amino acid residues in IS1-IS2 (H111) and IS2-IS3 (H179 and H183) loops potentiates Ca(v)2.3 by shifting the voltage dependence of activation toward more negative membrane potentials. We demonstrate that copper regulates the voltage dependence of Ca(v)2.3 by affecting gating charge movements. Thus, in the presence of copper, gating charges transition into the "ON" position slower, delaying activation and reducing the voltage sensitivity of the channel. Overall, our results suggest a new mechanism by which Glu and trace metals transiently modulate voltage-dependent gating of Ca(v)2.3, potentially affecting synaptic transmission and plasticity in the brain.


Assuntos
Canais de Cálcio Tipo R/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Sítios de Ligação , Fenômenos Biofísicos , Canais de Cálcio Tipo N/química , Canais de Cálcio Tipo N/genética , Canais de Cálcio Tipo N/metabolismo , Canais de Cálcio Tipo R/química , Canais de Cálcio Tipo R/genética , Proteínas de Transporte de Cátions/agonistas , Proteínas de Transporte de Cátions/química , Proteínas de Transporte de Cátions/genética , Cobre/farmacologia , Ácido Glutâmico/farmacologia , Glicina/análogos & derivados , Glicina/farmacologia , Células HEK293 , Humanos , Técnicas In Vitro , Ativação do Canal Iônico/efeitos dos fármacos , Potenciais da Membrana , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Ratos , Ratos Transgênicos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Oligoelementos/farmacologia
13.
J Phys Chem C Nanomater Interfaces ; 116(30): 16319-16324, 2012 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-26644813

RESUMO

It is well-known that ionic surfactant coated single-walled carbon nanotubes (SWNTs) possess higher near-infrared fluorescence (NIRF) quantum yield than nonionic polymer functionalized SWNTs. However, the influence of surface functionalization on the magnetic properties of SWNTs for T2-weighted magnetic resonance imaging (MRI) has not been reported. Here, we demonstrate that SWNTs functionalized by nonionic polymers display superior T2 relaxivity for MRI as compared to those coated by ionic surfactants. This difference may indicate that micelle structures formed by ionic surfactants are sufficiently tight to partially exclude water protons from the iron catalysts attached to the ends of SWNTs. On the basis of the different effects of the two types of suspension agents on NIRF and MRI of functionalized SWNTs, we further explore the competitive surface functionalization between ionic surfactants and nonionic polymers by stepwise replacing ionic surfactant molecules in a nanotube suspension with nonionic polymers. The superior NIRF of ionic surfactant coated SWNTs gradually quenches whereas no improvement on T2 relaxivity is observed during this replacement process. This result may indicate that nonionic polymers wrap around the outside of micelle structures to form small nanotube bundles rather than replacing ionic surfactants in the micelle structures to directly interact with the SWNT surface. Finally, we demonstrate the feasibility of dual-modality NIRF and MRI of nonionic polymer functionalized SWNTs in brain cells.

14.
Cereb Cortex ; 21(3): 666-82, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20705896

RESUMO

Fast-spiking (FS) cells are a prominent subtype of neocortical γ-aminobutyric acidergic interneurons that mediate feed-forward inhibition and the temporal sculpting of information transfer in neural circuits, maintain excitation/inhibition balance, and contribute to network oscillations. FS cell dysfunction may be involved in the pathogenesis of disorders such as epilepsy, autism, and schizophrenia. Mature FS cells exhibit coordinated molecular and cellular specializations that facilitate rapid responsiveness, including brief spikes and sustained high-frequency discharge. We show that these features appear during the second and third postnatal weeks driven by upregulation of K(+) channel subunits of the Kv3 subfamily. The low membrane resistance and fast time constant characteristic of FS cells also appears during this time, driven by expression of a K(+) leak current mediated by K(ir)2 subfamily inward rectifier K(+) channels and TASK subfamily 2-pore K(+) channels. Blockade of this leak produces dramatic depolarization of FS cells suggesting the possibility for potent neuromodulation. Finally, the frequency of FS cell membrane potential oscillations increases during development and is markedly slower in TASK-1/3 knockout mice, suggesting that TASK channels regulate FS cell rhythmogenesis. Our findings imply that some of the effects of acidosis and/or anesthetics on brain function may be due to blockade of TASK channels in FS cells.


Assuntos
Interneurônios/citologia , Neocórtex/crescimento & desenvolvimento , Proteínas do Tecido Nervoso/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Animais , Western Blotting , Separação Celular , Citometria de Fluxo , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/deficiência , Neurogênese/fisiologia , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp , Canais de Potássio de Domínios Poros em Tandem/deficiência , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Respir Physiol Neurobiol ; 175(2): 283-7, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21145990

RESUMO

The retrotrapezoid nucleus (RTN), located at the ventral surface of the medulla oblongata, contains glutamatergic Phox2b-expressing interneurons that have central respiratory chemoreceptor properties. RTN also operates as a relay for hypothalamic pathways that regulate breathing, one of which probably originates from the orexinergic neurons (Dias et al., 2009. J. Physiol. 587, 2059-2067). The present study explores this hypothesis at the cellular level. Using immunohistochemistry in adult Phox2b-eGFP transgenic mice, we demonstrate the presence of numerous close appositions between orexin-containing axonal varicosities and RTN chemoreceptor neurons. Using electrophysiological recordings in slices from neonatal (P6-P10) Phox2b-eGFP mice, we show that orexin A produces a robust dose-dependent excitation of the acid-sensitive RTN neurons (ED(50) ∼250nM). These data support the idea that RTN neurons are a point of convergence for several groups of CNS neurons that contribute to respiratory chemoreflexes, now including serotonergic and orexinergic neurons.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Bulbo/fisiologia , Neurônios/fisiologia , Neuropeptídeos/fisiologia , Neurotransmissores/fisiologia , Animais , Células Quimiorreceptoras/fisiologia , Feminino , Proteínas de Homeodomínio/fisiologia , Interneurônios/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Orexinas , Respiração , Centro Respiratório/fisiologia , Fatores de Transcrição/fisiologia
16.
J Neurosci ; 30(27): 9324-34, 2010 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-20610767

RESUMO

At surgical depths of anesthesia, inhalational anesthetics cause a loss of motor response to painful stimuli (i.e., immobilization) that is characterized by profound inhibition of spinal motor circuits. Yet, although clearly depressed, the respiratory motor system continues to provide adequate ventilation under these same conditions. Here, we show that isoflurane causes robust activation of CO(2)/pH-sensitive, Phox2b-expressing neurons located in the retrotrapezoid nucleus (RTN) of the rodent brainstem, in vitro and in vivo. In brainstem slices from Phox2b-eGFP mice, the firing of pH-sensitive RTN neurons was strongly increased by isoflurane, independent of prevailing pH conditions. At least two ionic mechanisms contributed to anesthetic activation of RTN neurons: activation of an Na(+)-dependent cationic current and inhibition of a background K(+) current. Single-cell reverse transcription-PCR analysis of dissociated green fluorescent protein-labeled RTN neurons revealed expression of THIK-1 (TWIK-related halothane-inhibited K(+) channel, K(2P)13.1), a channel that shares key properties with the native RTN current (i.e., suppression by inhalational anesthetics, weak rectification, inhibition by extracellular Na(+), and pH-insensitivity). Isoflurane also increased firing rate of RTN chemosensitive neurons in urethane-anesthetized rats, again independent of CO(2) levels. In these animals, isoflurane transiently enhanced activity of the respiratory system, an effect that was most prominent at low levels of respiratory drive and mediated primarily by an increase in respiratory frequency. These data indicate that inhalational anesthetics cause activation of RTN neurons, which serve an important integrative role in respiratory control; the increased drive provided by enhanced RTN neuronal activity may contribute, in part, to maintaining respiratory motor activity under immobilizing anesthetic conditions.


Assuntos
Anestésicos Inalatórios/farmacologia , Células Quimiorreceptoras/efeitos dos fármacos , Isoflurano/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Inibição Neural/efeitos dos fármacos , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Centro Respiratório/citologia , Análise de Variância , Animais , Animais Recém-Nascidos , Pressão Sanguínea/efeitos dos fármacos , Relação Dose-Resposta a Droga , Estimulação Elétrica/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Fluorescência Verde/genética , Proteínas de Homeodomínio/genética , Concentração de Íons de Hidrogênio , Técnicas In Vitro , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/genética , Camundongos , Camundongos Transgênicos , Técnicas de Patch-Clamp/métodos , Nervo Frênico/efeitos dos fármacos , Nervo Frênico/fisiologia , Respiração/efeitos dos fármacos , Fatores de Transcrição/genética
17.
J Neurosci ; 30(22): 7691-704, 2010 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-20519544

RESUMO

General anesthetics cause sedation, hypnosis, and immobilization via CNS mechanisms that remain incompletely understood; contributions of particular anesthetic targets in specific neural pathways remain largely unexplored. Among potential molecular targets for mediating anesthetic actions, members of the TASK subgroup [TASK-1 (K2P3.1) and TASK-3 (K2P9.1)] of background K(+) channels are appealing candidates since they are expressed in CNS sites relevant to anesthetic actions and activated by clinically relevant concentrations of inhaled anesthetics. Here, we used global and conditional TASK channel single and double subunit knock-out mice to demonstrate definitively that TASK channels account for motoneuronal, anesthetic-activated K(+) currents and to test their contributions to sedative, hypnotic, and immobilizing anesthetic actions. In motoneurons from all knock-out mice lines, TASK-like currents were reduced and cells were less sensitive to hyperpolarizing effects of halothane and isoflurane. In an immobilization assay, higher concentrations of both halothane and isoflurane were required to render TASK knock-out animals unresponsive to a tail pinch; in assays of sedation (loss of movement) and hypnosis (loss-of-righting reflex), TASK knock-out mice showed a modest decrease in sensitivity, and only for halothane. In conditional knock-out mice, with TASK channel deletion restricted to cholinergic neurons, immobilizing actions of the inhaled anesthetics and sedative effects of halothane were reduced to the same extent as in global knock-out lines. These data indicate that TASK channels in cholinergic neurons are molecular substrates for select actions of inhaled anesthetics; for immobilization, which is spinally mediated, these data implicate motoneurons as the likely neuronal substrates.


Assuntos
Anestésicos Inalatórios/farmacologia , Resposta de Imobilidade Tônica/efeitos dos fármacos , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Análise de Variância , Animais , Animais Recém-Nascidos , Fenômenos Biofísicos/efeitos dos fármacos , Fenômenos Biofísicos/genética , Tronco Encefálico/citologia , Linhagem Celular Transformada , Colina O-Acetiltransferase/metabolismo , Relação Dose-Resposta a Droga , Estimulação Elétrica , Feminino , Deleção de Genes , Halotano/farmacologia , Humanos , Concentração de Íons de Hidrogênio , Técnicas In Vitro , Isoflurano/farmacologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/deficiência , Técnicas de Patch-Clamp , Canais de Potássio de Domínios Poros em Tandem/deficiência
18.
J Comp Neurol ; 517(1): 69-86, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19711410

RESUMO

The retrotrapezoid nucleus (RTN) contains noncholinergic noncatecholaminergic glutamatergic neurons that express the transcription factor Phox2b (chemically coded or "cc" RTN neurons). These cells regulate breathing and may be central chemoreceptors. Here we explore their ultrastructure and their acid sensitivity by using two novel BAC eGFP-Phox2b transgenic mice (B/G, GENSAT JX99) in which, respectively, 36% and 100% of the cc RTN neurons express the transgene in complete or partial anatomical isolation from other populations of eGFP neurons. All but one of the eGFP-labeled RTN neurons recorded in these mice were acid activated in slices. These cells contained VGLUT2 mRNA, and 50% contained preprogalanin mRNA (determined by single-cell PCR in the B/G mouse). Two neuronal subgroups were revealed, which differed in discharge rate at pH 7.3 (type I approximately 2; type II approximately 4 Hz) and the degree of alkalization that silenced the cells (type I 7.4-7.6, type II 7.8-8.0). Medial to the RTN, C1 neurons recorded in a tyrosine hydroxylase-GFP mouse were pH insensitive between pH 6.9 and pH 7.5. Ultrastructural studies demonstrated that eGFP-labeled RTN neurons were surrounded by numerous capillaries and were often in direct contact with glial cells, pericytes, and the basement membrane of capillaries. Terminals contacting large proximal eGFP dendrites formed mainly symmetric, likely inhibitory, synapses. Terminals on more distal eGFP dendrites formed preferentially asymmetric, presumably excitatory, synapses. In sum, C1 cells are pH insensitive, whereas cc RTN neurons are uniformly acid sensitive. The RTN neurons receive inhibitory and excitatory synaptic inputs and may have unfettered biochemical interactions with glial cells and the local microvasculature.


Assuntos
Proteínas de Homeodomínio/metabolismo , Bulbo/fisiologia , Bulbo/ultraestrutura , Neurônios/fisiologia , Neurônios/ultraestrutura , Fatores de Transcrição/metabolismo , Potenciais de Ação/fisiologia , Animais , Membrana Basal/fisiologia , Membrana Basal/ultraestrutura , Tronco Encefálico/irrigação sanguínea , Tronco Encefálico/fisiologia , Tronco Encefálico/ultraestrutura , Cromossomos Artificiais Bacterianos , Dendritos/fisiologia , Dendritos/ultraestrutura , Galanina/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Homeodomínio/genética , Concentração de Íons de Hidrogênio , Técnicas In Vitro , Bulbo/irrigação sanguínea , Camundongos , Camundongos Transgênicos , Neuroglia/ultraestrutura , Pericitos/ultraestrutura , RNA Mensageiro/metabolismo , Sinapses/fisiologia , Sinapses/ultraestrutura , Fatores de Transcrição/genética , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA