Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
PLoS Biol ; 17(2): e2006094, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30789900

RESUMO

Inspiratory breathing movements depend on pre-Bötzinger complex (preBötC) interneurons that express calcium (Ca2+)-activated nonselective cationic current (ICAN) to generate robust neural bursts. Hypothesized to be rhythmogenic, reducing ICAN is predicted to slow down or stop breathing; its contributions to motor pattern would be reflected in the magnitude of movements (output). We tested the role(s) of ICAN using reverse genetic techniques to diminish its putative ion channels Trpm4 or Trpc3 in preBötC neurons in vivo. Adult mice transduced with Trpm4-targeted short hairpin RNA (shRNA) progressively decreased the tidal volume of breaths yet surprisingly increased breathing frequency, often followed by gasping and fatal respiratory failure. Mice transduced with Trpc3-targeted shRNA survived with no changes in breathing. Patch-clamp and field recordings from the preBötC in mouse slices also showed an increase in the frequency and a decrease in the magnitude of preBötC neural bursts in the presence of Trpm4 antagonist 9-phenanthrol, whereas the Trpc3 antagonist pyrazole-3 (pyr-3) showed inconsistent effects on magnitude and no effect on frequency. These data suggest that Trpm4 mediates ICAN, whose influence on frequency contradicts a direct role in rhythm generation. We conclude that Trpm4-mediated ICAN is indispensable for motor output but not the rhythmogenic core mechanism of the breathing central pattern generator.


Assuntos
Interneurônios/metabolismo , Atividade Motora , Respiração , Canais de Cátion TRPM/metabolismo , Envelhecimento/fisiologia , Animais , Comportamento Animal , Feminino , Masculino , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Canais de Cátion TRPC/genética , Canais de Cátion TRPC/metabolismo , Canais de Cátion TRPM/genética , Vigília
2.
J Physiol ; 591(10): 2393-401, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23440965

RESUMO

A key feature of neurodegenerative disease is the pathological loss of neurons that participate in generating behaviour. To investigate network properties of neural circuits and provide a complementary tool to study neurodegeneration in vitro or in situ, we developed an automated cell-specific laser detection and ablation system. The instrument consists of a two-photon and visible-wavelength confocal imaging setup, controlled by executive software, that identifies neurons in preparations based on genetically encoded fluorescent proteins or Ca(2+) imaging, and then sequentially ablates cell targets while monitoring network function concurrently. Pathological changes in network function can be directly attributed to ablated cells, which are logged in real time. Here, we investigated brainstem respiratory circuits to demonstrate single-cell precision in ablation during physiological network activity, but the technique could be applied to interrogate network properties in neural systems that retain network functionality in reduced preparations in vitro or in situ.


Assuntos
Tronco Encefálico/fisiologia , Terapia a Laser , Neurônios/fisiologia , Software , Algoritmos , Compostos de Anilina , Animais , Animais Recém-Nascidos , Cálcio/fisiologia , Feminino , Corantes Fluorescentes , Proteínas de Homeodomínio/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Confocal , Gravidez , Xantenos
3.
J Physiol ; 591(10): 2687-703, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23459755

RESUMO

Breathing in mammals depends on an inspiratory-related rhythm that is generated by glutamatergic neurons in the pre-Bötzinger complex (preBötC) of the lower brainstem. A substantial subset of putative rhythm-generating preBötC neurons derive from a single genetic line that expresses the transcription factor Dbx1, but the cellular mechanisms of rhythmogenesis remain incompletely understood. To elucidate these mechanisms, we carried out a comparative analysis of Dbx1-expressing neurons (Dbx1(+)) and non-Dbx1-derived (Dbx1(-)) neurons in the preBötC. Whole-cell recordings in rhythmically active newborn mouse slice preparations showed that Dbx1(+) neurons activate earlier in the respiratory cycle and discharge greater magnitude inspiratory bursts compared with Dbx1(-) neurons. Furthermore, Dbx1(+) neurons required less input current to discharge spikes (rheobase) in the context of network activity. The expression of intrinsic membrane properties indicative of A-current (IA) and hyperpolarization-activated current (Ih) tended to be mutually exclusive in Dbx1(+) neurons. In contrast, there was no such relationship in the expression of currents IA and Ih in Dbx1(-) neurons. Confocal imaging and digital morphological reconstruction of recorded neurons revealed dendritic spines on Dbx1(-) neurons, but Dbx1(+) neurons were spineless. The morphology of Dbx1(+) neurons was largely confined to the transverse plane, whereas Dbx1(-) neurons projected dendrites to a greater extent in the parasagittal plane. The putative rhythmogenic nature of Dbx1(+) neurons may be attributable, in part, to a higher level of intrinsic excitability in the context of network synaptic activity. Furthermore, Dbx1(+) neuronal morphology may facilitate temporal summation and integration of local synaptic inputs from other Dbx1(+) neurons, taking place largely in the dendrites, which could be important for initiating and maintaining bursts and synchronizing activity during the inspiratory phase.


Assuntos
Tronco Encefálico/fisiologia , Proteínas de Homeodomínio/fisiologia , Neurônios/fisiologia , Respiração , Animais , Animais Recém-Nascidos , Tronco Encefálico/citologia , Técnicas In Vitro , Camundongos , Camundongos Transgênicos
4.
Cell Rep ; 42(8): 113000, 2023 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-37590134

RESUMO

Inspiration is the inexorable active phase of breathing. The brainstem pre-Bötzinger complex (preBötC) gives rise to inspiratory neural rhythm, but its underlying cellular and ionic bases remain unclear. The long-standing "pacemaker hypothesis" posits that the persistent Na+ current (INaP) that gives rise to bursting-pacemaker properties in preBötC interneurons is essential for rhythmogenesis. We tested the pacemaker hypothesis by conditionally knocking out and knocking down the Scn8a (Nav1.6 [voltage-gated sodium channel 1.6]) gene in core rhythmogenic preBötC neurons. Deleting Scn8a substantially decreases the INaP and abolishes bursting-pacemaker activity, which slows inspiratory rhythm in vitro and negatively impacts the postnatal development of ventilation. Diminishing Scn8a via genetic interference has no impact on breathing in adult mice. We argue that the Scn8a-mediated INaP is not obligatory but that it influences the development and rhythmic function of the preBötC. The ubiquity of the INaP in respiratory brainstem interneurons could underlie breathing-related behaviors such as neonatal phonation or rhythmogenesis in different physiological conditions.


Assuntos
Tronco Encefálico , Respiração , Animais , Camundongos , Interneurônios , Neurônios , Taxa Respiratória , Canal de Sódio Disparado por Voltagem NAV1.6
5.
Sci Rep ; 12(1): 2923, 2022 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-35190626

RESUMO

Breathing depends on interneurons in the preBötzinger complex (preBötC) derived from Dbx1-expressing precursors. Here we investigate whether rhythm- and pattern-generating functions reside in discrete classes of Dbx1 preBötC neurons. In a slice model of breathing with ~ 5 s cycle period, putatively rhythmogenic Type-1 Dbx1 preBötC neurons activate 100-300 ms prior to Type-2 neurons, putatively specialized for output pattern, and 300-500 ms prior to the inspiratory motor output. We sequenced Type-1 and Type-2 transcriptomes and identified differential expression of 123 genes including ionotropic receptors (Gria3, Gabra1) that may explain their preinspiratory activation profiles and Ca2+ signaling (Cracr2a, Sgk1) involved in inspiratory and sigh bursts. Surprisingly, neuropeptide receptors that influence breathing (e.g., µ-opioid and bombesin-like peptide receptors) were only sparsely expressed, which suggests that cognate peptides and opioid drugs exert their profound effects on a small fraction of the preBötC core. These data in the public domain help explain the neural origins of breathing.


Assuntos
Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Interneurônios/fisiologia , Neurônios Motores/fisiologia , Transcriptoma/genética , Animais , Animais Recém-Nascidos , Fenômenos Eletrofisiológicos , Expressão Gênica , Camundongos , Camundongos Transgênicos , Respiração
6.
Sci Data ; 9(1): 457, 2022 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-35907922

RESUMO

Neurons in the brainstem preBötzinger complex (preBötC) generate the rhythm and rudimentary motor pattern for inspiratory breathing movements. We performed whole-cell patch-clamp recordings from inspiratory neurons in the preBötC of neonatal mouse slices that retain breathing-related rhythmicity in vitro. We classified neurons based on their electrophysiological properties and genetic background, and then aspirated their cellular contents for single-cell RNA sequencing (scRNA-seq). This data set provides the raw nucleotide sequences (FASTQ files) and annotated files of nucleotide sequences mapped to the mouse genome (mm10 from Ensembl), which includes the fragment counts, gene lengths, and fragments per kilobase of transcript per million mapped reads (FPKM). These data reflect the transcriptomes of the neurons that generate the rhythm and pattern for inspiratory breathing movements.


Assuntos
Neurônios , Centro Respiratório , Transcriptoma , Animais , Animais Recém-Nascidos , Camundongos , Neurônios/fisiologia , Técnicas de Patch-Clamp , Respiração , Centro Respiratório/citologia , Centro Respiratório/fisiologia , Análise de Célula Única
7.
J Neurosci ; 30(44): 14883-95, 2010 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-21048147

RESUMO

A subset of preBötzinger Complex (preBötC) neurokinin 1 receptor (NK1R) and somatostatin peptide (SST)-expressing neurons are necessary for breathing in adult rats, in vivo. Their developmental origins and relationship to other preBötC glutamatergic neurons are unknown. Here we show, in mice, that the "core" of preBötC SST(+)/NK1R(+)/SST 2a receptor(+) (SST2aR) neurons, are derived from Dbx1-expressing progenitors. We also show that Dbx1-derived neurons heterogeneously coexpress NK1R and SST2aR within and beyond the borders of preBötC. More striking, we find that nearly all non-catecholaminergic glutamatergic neurons of the ventrolateral medulla (VLM) are also Dbx1 derived. PreBötC SST(+) neurons are born between E9.5 and E11.5 in the same proportion as non-SST-expressing neurons. Additionally, preBötC Dbx1 neurons are respiratory modulated and show an early inspiratory phase of firing in rhythmically active slice preparations. Loss of Dbx1 eliminates all glutamatergic neurons from the respiratory VLM including preBötC NK1R(+)/SST(+) neurons. Dbx1 mutant mice do not express any spontaneous respiratory behaviors in vivo. Moreover, they do not generate rhythmic inspiratory activity in isolated en bloc preparations even after acidic or serotonergic stimulation. These data indicate that preBötC core neurons represent a subset of a larger, more heterogeneous population of VLM Dbx1-derived neurons. These data indicate that Dbx1-derived neurons are essential for the expression and, we hypothesize, are responsible for the generation of respiratory behavior both in vitro and in vivo.


Assuntos
Diferenciação Celular/genética , Proteínas de Homeodomínio/genética , Neurogênese/genética , Neurônios/citologia , Neurônios/fisiologia , Centro Respiratório/crescimento & desenvolvimento , Animais , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Homeodomínio/fisiologia , Camundongos , Camundongos Transgênicos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurônios/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Receptores da Neurocinina-1/fisiologia , Receptores de Somatostatina/genética , Receptores de Somatostatina/fisiologia , Centro Respiratório/citologia , Centro Respiratório/efeitos dos fármacos , Fenômenos Fisiológicos Respiratórios/genética , Somatostatina/metabolismo , Somatostatina/fisiologia
8.
eNeuro ; 7(1)2020.
Artigo em Inglês | MEDLINE | ID: mdl-31888961

RESUMO

The preBötzinger complex (preBötC) generates the rhythm and rudimentary motor pattern for inspiratory breathing movements. Here, we test "burstlet" theory (Kam et al., 2013a), which posits that low amplitude burstlets, subthreshold from the standpoint of inspiratory bursts, reflect the fundamental oscillator of the preBötC. In turn, a discrete suprathreshold process transforms burstlets into full amplitude inspiratory bursts that drive motor output, measurable via hypoglossal nerve (XII) discharge in vitro We recap observations by Kam and Feldman in neonatal mouse slice preparations: field recordings from preBötC demonstrate bursts and concurrent XII motor output intermingled with lower amplitude burstlets that do not produce XII motor output. Manipulations of excitability affect the relative prevalence of bursts and burstlets and modulate their frequency. Whole-cell and photonic recordings of preBötC neurons suggest that burstlets involve inconstant subsets of rhythmogenic interneurons. We conclude that discrete rhythm- and pattern-generating mechanisms coexist in the preBötC and that burstlets reflect its fundamental rhythmogenic nature.


Assuntos
Respiração , Centro Respiratório , Animais , Animais Recém-Nascidos , Interneurônios , Camundongos , Neurônios
9.
Sci Data ; 4: 170097, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28763053

RESUMO

The relationship between neuron morphology and function is a perennial issue in neuroscience. Information about synaptic integration, network connectivity, and the specific roles of neuronal subpopulations can be obtained through morphological analysis of key neurons within a microcircuit. Here we present morphologies of two classes of brainstem respiratory neurons. First, interneurons derived from Dbx1-expressing precursors (Dbx1 neurons) in the preBötzinger complex (preBötC) of the ventral medulla that generate the rhythm for inspiratory breathing movements. Second, Dbx1 neurons of the intermediate reticular formation that influence the motor pattern of pharyngeal and lingual movements during the inspiratory phase of the breathing cycle. We describe the image acquisition and subsequent digitization of morphologies of respiratory Dbx1 neurons from the preBötC and the intermediate reticular formation that were first recorded in vitro. These data can be analyzed comparatively to examine how morphology influences the roles of Dbx1 preBötC and Dbx1 reticular interneurons in respiration and can also be utilized to create morphologically accurate compartmental models for simulation and modeling of respiratory circuits.


Assuntos
Neurônios/citologia , Formação Reticular , Animais , Animais Recém-Nascidos , Tronco Encefálico , Proteínas de Homeodomínio , Interneurônios/citologia , Camundongos , Neurônios/metabolismo
10.
Sci Rep ; 7(1): 8669, 2017 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-28819234

RESUMO

We sequenced the transcriptome of brainstem interneurons in the specialized respiratory rhythmogenic site dubbed preBötzinger Complex (preBötC) from newborn mice. To distinguish molecular characteristics of the core oscillator we compared preBötC neurons derived from Dbx1-expressing progenitors that are respiratory rhythmogenic to neighbouring non-Dbx1-derived neurons, which support other respiratory and non-respiratory functions. Results in three categories are particularly salient. First, Dbx1 preBötC neurons express κ-opioid receptors in addition to µ-opioid receptors that heretofore have been associated with opiate respiratory depression, which may have clinical applications. Second, Dbx1 preBötC neurons express the hypoxia-inducible transcription factor Hif1a at levels three-times higher than non-Dbx1 neurons, which links core rhythmogenic microcircuits to O2-related chemosensation for the first time. Third, we detected a suite of transcription factors including Hoxa4 whose expression pattern may define the rostral preBötC border, Pbx3 that may influence ipsilateral connectivity, and Pax8 that may pertain to a ventrally-derived subset of Dbx1 preBötC neurons. These data establish the transcriptomic signature of the core respiratory oscillator at a perinatal stage of development.


Assuntos
Proteínas de Homeodomínio/genética , Neurônios/metabolismo , Transcriptoma , Animais , Animais Recém-Nascidos , Biomarcadores , Feminino , Expressão Gênica , Perfilação da Expressão Gênica , Genes Reporter , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Neurotransmissores/metabolismo , Peptídeos/metabolismo
11.
Elife ; 3: e02265, 2014 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-24842997

RESUMO

All motor behaviors require precise temporal coordination of different muscle groups. Breathing, for example, involves the sequential activation of numerous muscles hypothesized to be driven by a primary respiratory oscillator, the preBötzinger Complex, and at least one other as-yet unidentified rhythmogenic population. We tested the roles of Atoh1-, Phox2b-, and Dbx1-derived neurons (three groups that have known roles in respiration) in the generation and coordination of respiratory output. We found that Dbx1-derived neurons are necessary for all respiratory behaviors, whereas independent but coupled respiratory rhythms persist from at least three different motor pools after eliminating or silencing Phox2b- or Atoh1-expressing hindbrain neurons. Without Atoh1 neurons, however, the motor pools become temporally disorganized and coupling between independent respiratory oscillators decreases. We propose Atoh1 neurons tune the sequential activation of independent oscillators essential for the fine control of different muscles during breathing.DOI: http://dx.doi.org/10.7554/eLife.02265.001.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Lábio/citologia , Neurônios/fisiologia , Fenômenos Fisiológicos Respiratórios , Animais , Proteínas de Homeodomínio/fisiologia , Camundongos , Camundongos Transgênicos , Rombencéfalo/fisiologia , Medula Espinal/fisiologia
12.
Elife ; 3: e03427, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-25027440

RESUMO

To understand the neural origins of rhythmic behavior one must characterize the central pattern generator circuit and quantify the population size needed to sustain functionality. Breathing-related interneurons of the brainstem pre-Bötzinger complex (preBötC) that putatively comprise the core respiratory rhythm generator in mammals are derived from Dbx1-expressing precursors. Here, we show that selective photonic destruction of Dbx1 preBötC neurons in neonatal mouse slices impairs respiratory rhythm but surprisingly also the magnitude of motor output; respiratory hypoglossal nerve discharge decreased and its frequency steadily diminished until rhythm stopped irreversibly after 85±20 (mean ± SEM) cellular ablations, which corresponds to ∼15% of the estimated population. These results demonstrate that a single canonical interneuron class generates respiratory rhythm and contributes in a premotor capacity, whereas these functions are normally attributed to discrete populations. We also establish quantitative cellular parameters that govern network viability, which may have ramifications for respiratory pathology in disease states.


Assuntos
Proteínas de Homeodomínio/genética , Nervo Hipoglosso/fisiopatologia , Neurônios Motores/metabolismo , Centro Respiratório/fisiopatologia , Potenciais de Ação , Animais , Animais Recém-Nascidos , Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Inalação/fisiologia , Interneurônios/citologia , Interneurônios/fisiologia , Terapia a Laser , Camundongos , Camundongos Transgênicos , Neurônios Motores/patologia , Técnicas de Patch-Clamp , Centro Respiratório/lesões , Centro Respiratório/patologia , Taxa Respiratória , Técnicas de Cultura de Tecidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA