Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Proc Natl Acad Sci U S A ; 114(30): 8095-8100, 2017 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-28698373

RESUMO

Vocalization in young mice is an innate response to isolation or mechanical stimulation. Neuronal circuits that control vocalization and breathing overlap and rely on motor neurons that innervate laryngeal and expiratory muscles, but the brain center that coordinates these motor neurons has not been identified. Here, we show that the hindbrain nucleus tractus solitarius (NTS) is essential for vocalization in mice. By generating genetically modified newborn mice that specifically lack excitatory NTS neurons, we show that they are both mute and unable to produce the expiratory drive required for vocalization. Furthermore, the muteness of these newborns results in maternal neglect. We also show that neurons of the NTS directly connect to and entrain the activity of spinal (L1) and nucleus ambiguus motor pools located at positions where expiratory and laryngeal motor neurons reside. These motor neurons control expiratory pressure and laryngeal tension, respectively, thereby establishing the essential biomechanical parameters used for vocalization. In summary, our work demonstrates that the NTS is an obligatory component of the neuronal circuitry that transforms breaths into calls.


Assuntos
Núcleo Solitário/fisiologia , Vocalização Animal/fisiologia , Animais , Animais Recém-Nascidos , Feminino , Músculos Laríngeos/fisiologia , Comportamento Materno , Camundongos , Neurônios Motores/fisiologia , Gravidez , Respiração
2.
Nat Commun ; 13(1): 3284, 2022 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-35672398

RESUMO

While apneas are associated with multiple pathological and fatal conditions, the underlying molecular mechanisms remain elusive. We report that a mutated form of the transcription factor Mafa (Mafa4A) that prevents phosphorylation of the Mafa protein leads to an abnormally high incidence of breath holding apneas and death in newborn Mafa4A/4A mutant mice. This apneic breathing is phenocopied by restricting the mutation to central GABAergic inhibitory neurons and by activation of inhibitory Mafa neurons while reversed by inhibiting GABAergic transmission centrally. We find that Mafa activates the Gad2 promoter in vitro and that this activation is enhanced by the mutation that likely results in increased inhibitory drives onto target neurons. We also find that Mafa inhibitory neurons are absent from respiratory, sensory (primary and secondary) and pontine structures but are present in the vicinity of the hypoglossal motor nucleus including premotor neurons that innervate the geniohyoid muscle, to control upper airway patency. Altogether, our data reveal a role for Mafa phosphorylation in regulation of GABAergic drives and suggest a mechanism whereby reduced premotor drives to upper airway muscles may cause apneic breathing at birth.


Assuntos
Apneia , Neurônios Motores , Animais , Fatores de Transcrição Maf Maior , Camundongos , Neurônios Motores/fisiologia , Fosforilação , Regiões Promotoras Genéticas
3.
Respir Physiol Neurobiol ; 165(1): 40-8, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18977317

RESUMO

A mouse strain with a deleted acetylcholinesterase (AChE) gene (AChE knockout) shows a decreased inspiration time and increased tidal volume and ventilation .To investigate the respective roles of AChE in brain and muscle, we recorded respiration by means of whole-body plethysmography in knockout mice with tissue selective deletions in AChE expression. A mouse strain with the anchoring domains of AChE deleted (del E5+6 knockout mice) has very low activity in the brain and neuromuscular junction, but increased monomeric AChE in serum. A mouse strain with deletion of the muscle specific region of AChE (del i1RR knockout mice) exhibits no expression in muscle, but unaltered expression in the central nervous system. Neither strain exhibits the pronounced phenotypic traits observed in the complete AChE knockout strain. A third strain lacking the anchor molecule PRiMA, has no functional AChE and butyrylcholinesterase (BChE) in brain and an unaltered respiratory function. BChE inhibition by bambuterol decreases tidal volume and body temperature in del E5+6 and i1RR knockout strains, but not in PRiMA deletion or wild-type controls. We find that: (1) deletion of the full AChE gene is required for a pronounced alteration in respiratory phenotype, (2) BChE is involved in respiratory muscles contraction and temperature control in del E5+6 and i1RR knockout mice, and (3) AChE expression requiring a gene product splice to either exons 5 and 6 or regulated by intron1 influences temperature control.


Assuntos
Acetilcolinesterase/metabolismo , Encéfalo/enzimologia , Regulação da Expressão Gênica/fisiologia , Músculos/enzimologia , Respiração/genética , Acetilcolinesterase/deficiência , Análise de Variância , Animais , Temperatura Corporal/genética , Encéfalo/efeitos dos fármacos , Broncodilatadores/farmacologia , Butirilcolinesterase/metabolismo , Éxons/genética , Feminino , Regulação da Expressão Gênica/genética , Hipercapnia/genética , Hipercapnia/fisiopatologia , Hipóxia/genética , Hipóxia/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Músculos/efeitos dos fármacos , Pletismografia Total/métodos , Ventilação Pulmonar/genética , Respiração/efeitos dos fármacos , Deleção de Sequência/genética , Terbutalina/análogos & derivados , Terbutalina/farmacologia , Volume de Ventilação Pulmonar/genética
4.
Eur J Neurosci ; 28(3): 510-20, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18702723

RESUMO

Brain-derived neurotrophic factor (BDNF) is required during the prenatal period for normal development of the respiratory central command; however, the underlying mechanisms remain unknown. To approach this issue, the present study examined BDNF regulation of fetal respiratory rhythm generation in the preBötzinger complex (preBötC) of the mouse, using transverse brainstem slices obtained from prenatal day 16.5 animals. BDNF application (100 ng/mL, 15 min) increased the frequency of rhythmic population activity in the preBötC by 43%. This effect was not observed when preparations were exposed to nerve growth factor (100 ng/mL, 30 min) or pretreated with the tyrosine kinase inhibitor K252a (1 h, 200 nm), suggesting that BDNF regulation of preBötC activity requires activation of its cognate tyrosine receptor kinase, TrkB. Consistent with this finding, single-cell reverse transcription-polymerase chain reaction experiments showed that one third of the rhythmically active preBötC neurons analysed expressed TrkB mRNA. Moreover, 20% expressed BDNF mRNA, suggesting that the preBötC is both a target and a source of BDNF. At the network level, BDNF augmented activity of preBötC glutamatergic neurons and potentiated glutamatergic synaptic drives in respiratory neurons by 34%. At the cellular level, BDNF increased the activity frequency of endogenously bursting neurons by 53.3% but had no effect on basal membrane properties of respiratory follower neurons, including the Ih current. Our data indicate that BDNF signalling through TrkB can acutely modulate fetal respiratory rhythm in association with increased glutamatergic drive and bursting activity in the preBötC.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/farmacologia , Feto , Bulbo/anatomia & histologia , Respiração/efeitos dos fármacos , Centro Respiratório/efeitos dos fármacos , Centro Respiratório/fisiologia , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Animais , Animais Recém-Nascidos , Cálcio/metabolismo , Antagonistas de Aminoácidos Excitatórios/farmacologia , Feminino , Feto/anatomia & histologia , Feto/efeitos dos fármacos , Feto/fisiologia , Idade Gestacional , Ácido Glutâmico/metabolismo , Bulbo/efeitos dos fármacos , Camundongos , Neurônios/fisiologia , Técnicas de Patch-Clamp , Periodicidade , Gravidez , Receptor trkB/genética , Receptor trkB/metabolismo , Centro Respiratório/anatomia & histologia , Transdução de Sinais/fisiologia , Sinapses/metabolismo
5.
J Neurosci ; 25(17): 4307-18, 2005 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-15858057

RESUMO

To obtain insights into the emergence of rhythmogenic circuits supporting respiration, we monitored spontaneous activities in isolated brainstem and medullary transverse slice preparations of mouse embryos, combining electrophysiological and calcium imaging techniques. At embryonic day 15 (E15), in a restricted region ventral to the nucleus ambiguus, we observed the onset of a sustained high-frequency (HF) respiratory-like activity in addition to a preexisting low-frequency activity having a distinct initiation site, spatial extension, and susceptibility to gap junction blockers. At the time of its onset, the HF generator starts to express the neurokinin 1 receptor, is connected bilaterally, requires active AMPA/kainate glutamatergic synapses, and is modulated by substance P and the mu-opioid agonist D-Ala2-N-Me-Phe4-Glycol5-enkephalin. We conclude that a rhythm generator sharing the properties of the neonatal pre-Bötzinger complex becomes active during E15 in mice.


Assuntos
Tronco Encefálico/citologia , Neurônios Motores/fisiologia , Periodicidade , Respiração , Centro Respiratório/fisiologia , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Fatores Etários , Analgésicos Opioides/farmacologia , Animais , Cálcio/metabolismo , Toxina da Cólera/farmacologia , Diagnóstico por Imagem/métodos , Relação Dose-Resposta à Radiação , Estimulação Elétrica/métodos , Embrião de Mamíferos , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Feminino , Imunofluorescência/métodos , Técnicas In Vitro , Masculino , Camundongos , Técnicas de Patch-Clamp/métodos , Gravidez , Receptores da Neurocinina-1/metabolismo , Centro Respiratório/efeitos dos fármacos , Centro Respiratório/embriologia , Substância P/farmacologia , Ácido gama-Aminobutírico/farmacologia
6.
J Physiol Paris ; 100(5-6): 290-6, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17628454

RESUMO

In humans, several pathologies are associated with disturbances of the respiratory control, some of them including alteration in the brain-derived neurotrophic factor (BDNF) signalling pathway. BDNF has long been known as a neurotrophic factor involved in survival, differentiation and maintenance of neuronal populations in the peripheral and central nervous system. More recently BDNF has also been discovered to be a potent neuromodulator with acute effects on neuronal excitability and synaptic plasticity. Animals deleted for the gene encoding BDNF exhibit respiratory alteration suggesting an important but yet undefined role of the neurotrophin in respiratory rhythmogenesis either by a trophic and/or an acute action. The possibility that BDNF might exert an acute regulatory role on the rhythmic activity of the respiratory generator of the pre-Bötzinger complex has been recently examined in newborn mice in vitro. Results obtained, reviewed in the present paper, will help getting insights in respiratory rhythm regulatory mechanisms that involve BDNF signalling.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/fisiologia , Rede Nervosa/fisiologia , Periodicidade , Centro Respiratório/fisiologia , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Humanos , Técnicas In Vitro , Camundongos , Neurônios/fisiologia , Centro Respiratório/anatomia & histologia , Transdução de Sinais/fisiologia
7.
J Neurosci ; 23(20): 7685-9, 2003 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-12930808

RESUMO

Genetic loss of brain-derived neurotrophic factor (BDNF) severely disrupts brainstem control of respiratory rhythmogenesis in newborn mice; however, the sites at which BDNF acts to regulate respiratory rhythmogenesis are unknown. Using immunochemical and multiplex RT-PCR analysis in mouse brainstem slices, we report that the BDNF receptor, Tyrosine kinase B (TrkB), is strongly expressed in the pre-Bötzinger complex (PBC), the presumed site for rhythm generation, and colocalizes with neurokinin 1 (NK1), a marker of neurons critical for breathing. The period of the respiratory rhythm generated by PBC neurons in vitro was increased by 30% after BDNF treatment (100 ng/ml) and not by nerve growth factor (100 ng/ml) or BDNF (100 ng/ml) in the presence of the tyrosine kinase inhibitor K252a (200 nm). Both synaptic and voltage-dependent properties of PBC neurons were modified by BDNF. Synaptic currents underlying spontaneous rhythmic bursts and glutamate-evoked currents were enhanced by 66 and 33%, respectively. BDNF reduced the Ih current amplitude in rhythmic neurons by 46% and shifted its activation curve by -17 mV. All neurons expressing TrkB mRNA (n = 8) also expressed mRNAs for the Ih current [hyperpolarization-activated cyclic nucleotide-sensitive cation nonselective channel (HCN1)], and three of four NK1-positive neurons coexpressed TrkB and HCN mRNA. Six of 16 PBC neurons expressed BDNF mRNA, supporting the possibility of autocrine and paracrine actions of BDNF within the respiratory pattern generator. Our data demonstrate that BDNF can modulate respiratory network activity through TrkB signaling in rhythmic PBC neurons.


Assuntos
Neurônios/fisiologia , Receptor trkB/metabolismo , Centro Respiratório/metabolismo , Centro Respiratório/fisiologia , Animais , Animais Recém-Nascidos , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Células Cultivadas , Condutividade Elétrica , Potenciais Pós-Sinápticos Excitadores , Imuno-Histoquímica , Bulbo/anatomia & histologia , Bulbo/fisiologia , Camundongos , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Periodicidade , RNA Mensageiro/biossíntese , Receptor trkB/análise , Receptor trkB/genética , Centro Respiratório/citologia , Transcrição Gênica
8.
J Neurosci ; 24(42): 9383-90, 2004 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-15496674

RESUMO

Observations of knock-out mice suggest that breathing at birth requires correct development of a specific hindbrain territory corresponding to rhombomeres (r) 3 and 4. Focusing on this territory, we examined the development of a neuronal rhythm generator in the chick embryo. We show that rhythmic activity in r4 is inducible after developmental stage 10 through interaction with r3. Although the nature of this interaction remains obscure, we find that the expression of Krox20, a segmentation gene responsible for specifying r3 and r5, is sufficient to endow other rhombomeres with the capacity to induce rhythmic activity in r4. Induction is robust, because it can be reproduced with r2 and r6 instead of r4 and with any hindbrain territory that normally expresses Krox20 (r3, r5) or can be forced to do so (r1, r4). Interestingly, the interaction between r4 and r3/r5 that results in rhythm production can only take place through the anterior border of r4, revealing a heretofore unsuspected polarity in individual rhombomeres. The r4 rhythm generator appears to be homologous to a murine respiratory parafacial neuronal system developing in r4 under the control of Krox20 and Hoxa1. These results identify a late role for Krox20 at the onset of neurogenesis.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Rombencéfalo/embriologia , Fatores de Transcrição/fisiologia , Potenciais de Ação/fisiologia , Animais , Embrião de Galinha , Proteína 2 de Resposta de Crescimento Precoce , Eletroporação , Periodicidade , Plasmídeos , Proteínas Recombinantes , Respiração , Rombencéfalo/fisiologia
9.
Prog Biophys Mol Biol ; 84(2-3): 89-106, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-14769431

RESUMO

The respiratory rhythm is generated within the hindbrain reticular formation, rostrally in the vicinity of the facial nucleus and caudally within the vagal/glossopharyngeal domain. This is probably one of the best models to understand how genes have been selected and conserved to control adaptive behaviour in vertebrates. The para-facial region is well understood with respect to the transcription factors that underlie antero-posterior specification of neural progenitors in the embryo. Hox paralogs and Hox-regulating genes kreisler and Krox-20 govern transient formation of developmental compartments, the rhombomeres, in which rhythmic neuronal networks develop. Hox are master genes selecting and coordinating the developmental fate of reticular and motor neurons thereby specifying patterns of motor activities operating throughout life. Neuronal function and development are also tightly linked in the vagal/glossopharyngeal domain. At this level, bdnf acts as a neurotrophin of peripheral chemoafferent neural populations and as a neuromodulator of the central rhythmogenic respiratory circuits. A general view is now emerging on the role of developmental transcription and trophic factors allowing the coordinated integration of different neuronal types to produce, and eventually refine, respiratory rhythmic pattern in a use-dependent manner.


Assuntos
Proteínas Aviárias , Tronco Encefálico/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Oncogênicas , Animais , Fator Neurotrófico Derivado do Encéfalo/fisiologia , Embrião de Galinha , Proteínas de Ligação a DNA/fisiologia , Proteína 2 de Resposta de Crescimento Precoce , Humanos , Fator de Transcrição MafB , Camundongos , Modelos Biológicos , Neurônios/metabolismo , Fenótipo , Rombencéfalo/fisiologia , Fatores de Tempo , Fatores de Transcrição/fisiologia
10.
Brain Res ; 1042(2): 133-43, 2005 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-15854585

RESUMO

To understand how nicotinic cholinergic receptors may participate in epileptic seizures, we tested the effects of nicotine and of the competitive nicotinic antagonists dihydro-beta-erythroidine and alpha-bungarotoxin on synaptic paroxysmal depolarization shifts (PDSs) and intrinsic bursts of action potentials recorded in slices from rats presenting a cortical status epilepticus. This model named GABA-withdrawal syndrome (GWS) appears consecutive to the interruption of a prolonged intracortical GABA infusion. Effects of both nicotinic antagonists suggest a distinct involvement of alpha4-beta2 and alpha7 subunits in shaping individual PDSs and patterning repetitive bursts. On one hand, in GWS rats, an increase of PDS latency and prolongation of PDS and bursts were induced by nicotine and reduced by dihydro-beta-erythroidine, but not by alpha-bungarotoxin. The K+ blocker tetraethylammonium also increased duration without changing latency. Thus, dihydro-beta-erythroidine-sensitive receptors exert distinct controls on the presynaptic generation of PDS and on the process which terminates PDSs and bursts. On the other hand, alpha-bungarotoxin depolarized neurons and generated rhythmic discharges of clustered bursts. Clustered bursts were also observed in slices obtained from GWS rats treated with the acetylcholinesterase inhibitor eserine. We suggest that both dihydro-beta-erythroidine and alpha-bungarotoxin-sensitive sites control paroxysmic activities in GWS and could be involved in some human and animal epilepsies presenting mutations of nicotinic cholinergic receptors.


Assuntos
Epilepsia/metabolismo , Receptores Nicotínicos/metabolismo , Síndrome de Abstinência a Substâncias/metabolismo , Ácido gama-Aminobutírico/farmacologia , Animais , Masculino , Nicotina/farmacologia , Antagonistas Nicotínicos/farmacologia , Ratos , Ratos Wistar
11.
Pharmacol Biochem Behav ; 80(1): 53-61, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15652380

RESUMO

We investigated the contributions of acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) inhibition to the respiratory dysfunction produced by organophosphates in mice which were adapted or not to low AChE activity. Effects of acute selective inhibition of AChE and BChE on ventilation measured by whole-body plethysmography were compared in mice with either normal AChE activity (wild-type), or mice adapted to a null AChE activity (homozygotes for AChE gene deletion) or adapted to an intermediate level of activity (heterozygotes). In wild-type mice acute reduction of AChE by Huperzine A (1 mg/kg) to the level found in asymptomatic heterozygotes, induced tremors but no respiratory depression, whereas the same dose of Huperzine in heterozygote animals further reduced AChE activity, increased tidal volume (V(T)) and decreased breathing frequency (f(R)). A lethal dose of Huperzine in wild-type mice augmented these respiratory effects, but was ineffective in homozygotes. BChE inhibition by bambuterol was ineffective in wild-type mice and heterozygotes, decreased V(T) in homozygotes adapted to null AChE activity but increased V(T) in wild-type mice acutely treated with Huperzine, also aggravating the cholinergic syndrome. We conclude that: (1) Huperzine does not perturb respiration at a dose inhibiting 40% of AChE, and at a lethal dose does not affect any other enzyme important for respiration; (2) Respiratory function is more sensitive to anticholinesterases in heterozygotes than in wild-type mice; (3) BChE may play distinct roles in respiratory function, because its inhibition has opposite effects on tidal volume depending on whether the mouse has adapted to null AChE or whether AChE has been lowered acutely; (4) BChE inhibition may contribute to the respiratory toxicity of organophosphates.


Assuntos
Acetilcolinesterase/metabolismo , Butirilcolinesterase/metabolismo , Inibidores da Colinesterase/farmacologia , Respiração/efeitos dos fármacos , Terbutalina/análogos & derivados , Acetilcolinesterase/deficiência , Acetilcolinesterase/genética , Animais , Feminino , Masculino , Camundongos , Camundongos Knockout , Terbutalina/farmacologia
12.
Mol Neurobiol ; 28(3): 277-94, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14709790

RESUMO

Respiration is a rhythmic motor behavior that appears in the fetus and acquires a vital importance at birth. It is generated within central pattern-generating neuronal networks of the hindbrain. This region of the brain is of particular interest since it is the most understood part with respect to the cellular and molecular mechanisms that underlie its development. Hox paralogs and Hox-regulating genes kreisler/mafB and Krox20 are required for the normal formation of rhombomeres in vertebrate embryos. From studies of rhombomeres r3 and r4, the authors review mechanisms whereby these developmental genes may govern the early embryonic development of para-facial neuronal networks and specify patterns of motor activities operating throughout life. A model whereby the regional identity of progenitor cells can be abnormally specified in r3 and r4 after a mutation of these genes is proposed. Novel neuronal circuits may develop from some of these misspecified progenitors while others are eliminated, eventually affecting respiration and survival after birth.


Assuntos
Padronização Corporal/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Centro Respiratório/embriologia , Centro Respiratório/fisiologia , Rombencéfalo/embriologia , Rombencéfalo/fisiologia , Animais , Genes Homeobox/genética , Humanos , Recém-Nascido , Malformações do Sistema Nervoso/genética , Malformações do Sistema Nervoso/patologia , Malformações do Sistema Nervoso/fisiopatologia , Centro Respiratório/citologia , Síndrome do Desconforto Respiratório do Recém-Nascido/genética , Síndrome do Desconforto Respiratório do Recém-Nascido/patologia , Síndrome do Desconforto Respiratório do Recém-Nascido/fisiopatologia , Rombencéfalo/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo
13.
Neurosci Lett ; 323(2): 89-92, 2002 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-11950500

RESUMO

We investigated a possible involvement of the prion protein in ventilatory control in four groups of mice, those deficient for the prion protein (PrP(c)), those overexpressing the prion protein, and two groups of genetically and age-matched controls. Ventilatory patterns of unrestrained mice were measured in a whole-body plethysmograph. Between each genotype and its control, we compared ventilation at rest and the ventilatory response to moderate hypoxia (10-12% O2), hyperoxia and hyperoxic hypercapnia. Mice lacking or overexpressing PrP(c) and their respective controls showed similar ventilatory patterns at rest and similar chemosensory responses when awake and under urethane anesthesia. Our results do not support the view that PrP(c) may play any significant role in basal ventilation or in the chemosensory ventilatory control of adult mice.


Assuntos
Proteínas PrPC/biossíntese , Proteínas PrPC/genética , Ventilação Pulmonar/genética , Animais , Feminino , Hipóxia Encefálica/genética , Hipóxia Encefálica/metabolismo , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas PrPC/deficiência , Ventilação Pulmonar/fisiologia
14.
Respir Physiol Neurobiol ; 139(1): 91-5, 2003 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-14637315

RESUMO

Respiratory neuronal networks in vertebrates appear to be able to generate a variety of rhythmic patterns in vivo, leading to the biological diversity of eupneic patterns as well as to life-threatening dyspneic patterns. Eupnea is best viewed as the collection of respiratory strategies preventing potential dyspneas, the major (and perhaps the only) criterion for a definition being that eupnea allows survival. Specific criteria can then be derived from the physiological identification of neurobiological mechanisms underlying identified dyspneic patterns, by exaggerating (pro-dyspneic mechanisms) or suppressing them (anti-dyspneic mechanisms). Because eupnea is vital, and one of the major targets of evolutionary pressure, identification of dyspnea-related neuronal systems seems to be important to understand the normal biological organization of the respiratory neuronal system.


Assuntos
Encéfalo/fisiologia , Neurobiologia , Respiração , Fenômenos Fisiológicos Respiratórios , Adaptação Fisiológica , Animais , Apneia/fisiopatologia , Dispneia/fisiopatologia , Humanos , Hipóxia/fisiopatologia
15.
Respir Physiol Neurobiol ; 139(3): 237-45, 2004 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-15122990

RESUMO

Acetylcholine (ACh) acting through muscarinic receptors is thought to be involved in the control of breathing, notably in central and peripheral chemosensory afferents and in regulations related to sleep-wake states. By using whole-body plethysmography, we compared baseline breathing at rest and ventilatory responses to acute exposure (5 min) to moderate hypoxia (10% O(2)) and hypercapnia (3 and 5% CO(2)) in mice lacking either the M(1) or the M(3) muscarinic receptor, and in wild-type matched controls. M(1) knockout mice showed normal minute ventilation (V(E)) but elevated tidal volume (V(T)) at rest, and normal chemosensory ventilatory responses to hypoxia and hypercapnia. M(3) knockout mice had elevated V(E) and V(T) at rest, a reduced V(T) response slope to hypercapnia, and blunted V(E) and frequency responses to hypoxia. The results suggest that M(1) and M(3) muscarinic receptors play significant roles in the regulation of tidal volume at rest and that the afferent pathway originating from peripheral chemoreceptors involves M(3) receptors.


Assuntos
Hipercapnia/fisiopatologia , Hipóxia/fisiopatologia , Ventilação Pulmonar/fisiologia , Receptor Muscarínico M1/fisiologia , Receptor Muscarínico M3/fisiologia , Respiração/genética , Animais , Temperatura Corporal/genética , Temperatura Corporal/fisiologia , Peso Corporal/genética , Peso Corporal/fisiologia , Dióxido de Carbono/farmacologia , Relação Dose-Resposta a Droga , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Oxigênio/farmacologia , Pletismografia Total/métodos , Ventilação Pulmonar/efeitos dos fármacos , Receptor Muscarínico M1/genética , Receptor Muscarínico M3/genética , Respiração/efeitos dos fármacos , Volume de Ventilação Pulmonar/genética , Volume de Ventilação Pulmonar/fisiologia
16.
Respir Physiol Neurobiol ; 135(2-3): 121-32, 2003 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-12809613

RESUMO

The present paper presents some of the molecular switches that may operate at early embryonic stages to make development of the brainstem respiratory rhythm generator a robust and irreversible process. We concentrate on the role of transient Hox-related gene expression patterns in register with the regionalisation of the rhombencephalic neural tube along the antero-posterior axis. Using different recording and isolation procedures in chick embryos, we show that the hindbrain is subdivided at E1 into developmental units (rhombomeres) intrinsically able to produce rhythm generating neuronal circuits active at E5. At E6, intrinsic cues also allow a progressive maturation of episodic rhythm generators that persists after isolation of the hindbrain in vitro and requires odd/even rhombomeric interactions at E1. From these results and from respiratory pathologies observed in transgenic mice, we are beginning to understand that, despite diversity of breathing patterns and adaptations, there are links between developmental control genes and adult respiration.


Assuntos
Sistema Nervoso Central/embriologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Genes de Troca/genética , Respiração/genética , Animais , Sistema Nervoso Central/anatomia & histologia , Sistema Nervoso Central/crescimento & desenvolvimento , Eletrofisiologia/métodos , Embrião de Mamíferos , Embrião não Mamífero , Genes Homeobox , Genes de Troca/fisiologia , Modelos Biológicos
17.
Respir Physiol Neurobiol ; 131(1-2): 5-13, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12106991

RESUMO

We are investigating neuronal circuits resulting from conservative developmental mechanisms orchestrating the segmentation of the vertebrates hindbrain into compartments called rhombomeres (r). Segmentation transcription factors Hoxa1, Krox20 and kreisler are expressed in the future rhombomeres r4-r5, r3 and r5, r5-r6, respectively. In mice, the in vivo and in vitro analysis of neuronal groups after inactivation of these three genes revealed distinct postnatal respiratory phenotypes associated with defects of central respiratory controls resulting from deletion, neoformation or reconfiguration of modular circuits. In chick and mice, we have found neuronal rhythm generators that conform to the rhombomeric anatomical pattern as early as at the end of the segmentation. By isolating chick hindbrain segments in vitro, we have also identified rhombomeric motifs allowing the formation or deletion of a specific (GABAergic) rhythm-promoting module. Therefore, primordial rhombomeric organization of the hindbrain seems to determine a modular organization of the rhythmogenic network, thereby influencing later function of brainstem respiratory control networks.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Rede Nervosa/embriologia , Rede Nervosa/fisiologia , Mecânica Respiratória/genética , Animais , Embrião de Galinha , Camundongos , Camundongos Mutantes , Mecânica Respiratória/fisiologia , Rombencéfalo/embriologia , Rombencéfalo/fisiologia
18.
PLoS One ; 7(2): e31140, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22363567

RESUMO

The proper development and maturation of neuronal circuits require precise migration of component neurons from their birthplace (germinal zone) to their final positions. Little is known about the effects of aberrant neuronal position on the functioning of organized neuronal groups, especially in mammals. Here, we investigated the formation and properties of brainstem respiratory neurons in looptail (Lp) mutant mice in which facial motor neurons closely apposed to some respiratory neurons fail to migrate due to loss of function of the Wnt/Planar Cell Polarity (PCP) protein Vangl2. Using calcium imaging and immunostaining on embryonic hindbrain preparations, we found that respiratory neurons constituting the embryonic parafacial oscillator (e-pF) settled at the ventral surface of the medulla in Vangl2(Lp/+) and Vangl2(Lp/Lp) embryos despite the failure of tangential migration of its normally adjacent facial motor nucleus. Anatomically, the e-pF neurons were displaced medially in Lp/+ embryos and rostro-medially Lp/Lp embryos. Pharmacological treatments showed that the e-pF oscillator exhibited characteristic network properties in both Lp/+ and Lp/Lp embryos. Furthermore, using hindbrain slices, we found that the other respiratory oscillator, the preBötzinger complex, was also anatomically and functionally established in Lp mutants. Importantly, the displaced e-pF oscillator established functional connections with the preBötC oscillator in Lp/+ mutants. Our data highlight the robustness of the developmental processes that assemble the neuronal networks mediating an essential physiological function.


Assuntos
Relógios Biológicos , Tronco Encefálico/patologia , Movimento Celular , Polaridade Celular , Neurônios/patologia , Respiração , Proteínas Wnt/metabolismo , Animais , Embrião de Mamíferos/patologia , Face , Feminino , Proteínas de Homeodomínio/metabolismo , Concentração de Íons de Hidrogênio , Proteínas com Homeodomínio LIM/metabolismo , Camundongos , Camundongos Mutantes , Modelos Biológicos , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Neurônios/metabolismo , Rombencéfalo/metabolismo , Rombencéfalo/patologia , Fatores de Transcrição/metabolismo
19.
Respir Physiol Neurobiol ; 178(1): 146-55, 2011 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-21527363

RESUMO

Foetal breathing in mice results from prenatal activity of the two coupled hindbrain oscillators considered to be responsible for respiratory rhythm generation after birth: the pre-Bötzinger complex (preBötC) is active shortly before the onset of foetal breathing; the parafacial respiratory group (e-pF in embryo) starts activity one day earlier. Transcription factors have been identified that are essential to specify neural progenitors and lineages forming each of these oscillators during early development of the neural tube: Hoxa1, Egr2 (Krox20), Phox2b, Lbx1 and Atoh1 for the e-pF; Dbx1 and Evx1 for the preBötC which eventually grow contralateral axons requiring expression of Robo3. Inactivation of the genes encoding these factors leads to mis-specification of these neurons and distinct breathing abnormalities: apneic patterns and loss of central chemosensitivity for the e-pF (central congenital hypoventilation syndrome, CCHS, in humans), complete loss of breathing for the preBötC, right-left desynchronized breathing in Robo3 mutants. Mutations affecting development in more rostral (pontine) respiratory territories change the shape of the inspiratory drive without affecting the rhythm. Other (primordial) embryonic oscillators start in the mouse three days before the e-pF, to generate low frequency (LF) rhythms that are probably required for activity-dependent development of neurones at embryonic stages; in the foetus, however, they are actively silenced to avoid detrimental interaction with the on-going respiratory rhythm. Altogether, these observations provide a strong support to the previously proposed hypothesis that the functional organization of the respiratory generator is specified at early stages of development and is dual in nature, comprising two serially non-homologous oscillators.


Assuntos
Embrião de Mamíferos/fisiologia , Centro Respiratório/embriologia , Animais , Camundongos
20.
Nat Neurosci ; 13(9): 1066-74, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20680010

RESUMO

Breathing is a bilaterally synchronous behavior that relies on a respiratory rhythm generator located in the brainstem. An essential component of this generator is the preBötzinger complex (preBötC), which paces inspirations. Little is known about the developmental origin of the interneuronal populations forming the preBötC oscillator network. We found that the homeobox gene Dbx1 controls the fate of glutamatergic interneurons required for preBötC rhythm generation in the mouse embryo. We also found that a conditional inactivation in Dbx1-derived cells of the roundabout homolog 3 (Robo3) gene, which is necessary for axonal midline crossing, resulted in left-right de-synchronization of the preBötC oscillator. Together, these findings identify Dbx1-derived interneurons as the core rhythmogenic elements of the preBötC oscillator and indicate that Robo3-dependent guidance signaling in these cells is required for bilaterally synchronous activity.


Assuntos
Axônios/fisiologia , Interneurônios/fisiologia , Respiração , Rombencéfalo/embriologia , Rombencéfalo/fisiologia , Animais , Lateralidade Funcional , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Neurônios Motores/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Vias Neurais/embriologia , Vias Neurais/fisiologia , Periodicidade , Receptores de Superfície Celular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA