Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
J Mol Cell Biol ; 13(3): 210-224, 2021 07 06.
Article in English | MEDLINE | ID: mdl-33475140

ABSTRACT

Breathing is an integrated motor behavior that is driven and controlled by a network of brainstem neurons. Zfhx4 is a zinc finger transcription factor and our results showed that it was specifically expressed in several regions of the mouse brainstem. Mice lacking Zfhx4 died shortly after birth from an apparent inability to initiate respiration. We also found that the electrical rhythm of brainstem‒spinal cord preparations was significantly depressed in Zfhx4-null mice compared to wild-type mice. Immunofluorescence staining revealed that Zfhx4 was coexpressed with Phox2b and Math1 in the brainstem and that Zfhx4 ablation greatly decreased the expression of these proteins, especially in the retrotrapezoid nucleus. Combined ChIP‒seq and mRNA expression microarray analysis identified Phox2b as the direct downstream target gene of Zfhx4, and this finding was validated by ChIP‒qPCR. Previous studies have reported that both Phox2b and Math1 play key roles in the development of the respiratory center, and Phox2b and Math1 knockout mice are neonatal lethal due to severe central apnea. On top of this, our study revealed that Zfhx4 is a critical regulator of Phox2b expression and essential for perinatal breathing.


Subject(s)
Apnea , Homeodomain Proteins/genetics , Respiratory Center , Animals , Apnea/metabolism , Apnea/mortality , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/metabolism , Brain Stem/metabolism , Gene Expression Regulation , Homeodomain Proteins/metabolism , Mice , Mice, Knockout/genetics , Neurons/metabolism , Respiration , Respiratory Center/embryology , Respiratory Center/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Wiley Interdiscip Rev Dev Biol ; 9(3): e366, 2020 05.
Article in English | MEDLINE | ID: mdl-31816185

ABSTRACT

The respiratory circuit is comprised of over a dozen functionally and anatomically segregated brainstem nuclei that work together to control respiratory rhythms. These respiratory rhythms emerge prenatally but only acquire vital importance at birth, which is the first time the respiratory circuit faces the sole responsibility for O2 /CO2 homeostasis. Hence, the respiratory circuit has little room for trial-and-error-dependent fine tuning and relies on a detailed genetic blueprint for development. This blueprint is provided by transcription factors that have specific spatiotemporal expression patterns along the rostral-caudal or dorsal-ventral axis of the developing brainstem, in proliferating precursor cells and postmitotic neurons. Studying these transcription factors in mice has provided key insights into the functional segregation of respiratory control and the vital importance of specific respiratory nuclei. Many studies converge on just two respiratory nuclei that each have rhythmogenic properties during the prenatal period: the preBötzinger complex (preBötC) and retrotrapezoid nucleus/parafacial nucleus (RTN/pF). Here, we discuss the transcriptional regulation that guides the development of these nuclei. We also summarize evidence showing that normal preBötC development is necessary for neonatal survival, and that neither the preBötC nor the RTN/pF alone is sufficient to sustain normal postnatal respiratory rhythms. Last, we highlight several studies that use intersectional genetics to assess the necessity of transcription factors only in subregions of their expression domain. These studies independently demonstrate that lack of RTN/pF neurons weakens the respiratory circuit, yet these neurons are not necessary for neonatal survival because developmentally related populations can compensate for abnormal RTN/pF function at birth. This article is categorized under: Nervous System Development > Vertebrates: Regional Development.


Subject(s)
Neurogenesis , Respiratory Center/metabolism , Animals , Gene Expression Regulation, Developmental , Humans , Respiratory Center/embryology , Respiratory Center/physiology
3.
Neuroscience ; 357: 160-171, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28583412

ABSTRACT

The central command for breathing arises mainly from two interconnected rhythmogenic hindbrain networks, the parafacial respiratory group (pFRG or epF at embryonic stages) and the preBötzinger complex (preBötC), which are comprised of a limited number of neurons located in confined regions of the ventral medulla. In rodents, both networks become active toward the end of gestation but little is known about the signaling pathways involved in their anatomical and functional establishment during embryogenesis. During embryonic development, epF and preBötC neurons migrate from their territories of origin to their final positions in ventral brainstem areas. Planar Cell Polarity (PCP) signaling, including the molecule Scrib, is known to control the developmental migration of several hindbrain neuronal groups. Accordingly, a homozygous mutation of Scrib leads to severe disruption of hindbrain anatomy and function. Here, we aimed to determine whether Scrib is also involved in the prenatal development of the hindbrain nuclei controlling breathing. We combined immunostaining, calcium imaging and electrophysiological recordings of neuronal activity in isolated in vitro preparations. In the Scrib mutant, despite severe neural tube defects, epF and preBötC neurons settled at their expected hindbrain positions. Furthermore, both networks remained capable of generating rhythmically organized, respiratory-related activities and exhibited normal sensitivity to pharmacological agents known to modify respiratory circuit function. Thus Scrib is not required for the proper migration of epF and preBötC neurons during mouse embryogenesis. Our findings thus further illustrate the robustness and specificity of the developmental processes involved in the establishment of hindbrain respiratory circuits.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Respiration , Respiratory Center/embryology , Respiratory Center/metabolism , Rhombencephalon/embryology , Rhombencephalon/metabolism , Animals , Calcium/metabolism , Cations, Divalent/metabolism , Cell Movement/physiology , Intracellular Signaling Peptides and Proteins/genetics , Mice, Transgenic , Mutation , Neural Pathways/drug effects , Neural Pathways/embryology , Neural Pathways/metabolism , Neural Pathways/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Respiration/drug effects , Respiratory Center/drug effects , Respiratory Center/pathology , Respiratory System Agents/pharmacology , Rhombencephalon/drug effects , Rhombencephalon/pathology , Tissue Culture Techniques
4.
Elife ; 52016 07 19.
Article in English | MEDLINE | ID: mdl-27434668

ABSTRACT

Breathing is a vital rhythmic behavior generated by hindbrain neuronal circuitry, including the preBötzinger complex network (preBötC) that controls inspiration. The emergence of preBötC network activity during prenatal development has been described, but little is known regarding inspiratory neurons expressing pacemaker properties at embryonic stages. Here, we combined calcium imaging and electrophysiological recordings in mouse embryo brainstem slices together with computational modeling to reveal the existence of heterogeneous pacemaker oscillatory properties relying on distinct combinations of burst-generating INaP and ICAN conductances. The respective proportion of the different inspiratory pacemaker subtypes changes during prenatal development. Concomitantly, network rhythmogenesis switches from a purely INaP/ICAN-dependent mechanism at E16.5 to a combined pacemaker/network-driven process at E18.5. Our results provide the first description of pacemaker bursting properties in embryonic preBötC neurons and indicate that network rhythmogenesis undergoes important changes during prenatal development through alterations in both circuit properties and the biophysical characteristics of pacemaker neurons.


Subject(s)
Biological Clocks , Brain Stem/embryology , Brain Stem/physiology , Neurons/physiology , Respiratory Center/embryology , Respiratory Center/physiology , Animals , Functional Neuroimaging , Mice , Patch-Clamp Techniques
5.
Med Sci (Paris) ; 29(10): 875-82, 2013 Oct.
Article in French | MEDLINE | ID: mdl-24148126

ABSTRACT

From birth onwards, rhythmic breathing is required for blood oxygenation and survival in mammals. During their lifespan, human or mouse or elephant will spontaneously produce several hundreds of millions of respiratory movements. The central nervous command responsible for these spontaneous rhythmic movements is elaborated by a complex neural network extending within the brainstem. In the medulla, a special part of this network contains respiratory pacemaker neurons that play a crucial role in respiratory rhythmogenesis: the pre-Bötzinger complex. This review summarizes and discusses the main electrophysiological, molecular and genetic mechanisms contributing to the function and the perinatal maturation of the pre-Bötzinger complex.


Subject(s)
Electrophysiological Phenomena , Respiration/genetics , Respiratory Center , Adult , Animals , Humans , Infant, Newborn , Mammals , Mice , Motor Neurons/cytology , Motor Neurons/physiology , Periodicity , Respiratory Center/embryology , Respiratory Center/growth & development , Respiratory Center/physiology
6.
Respir Physiol Neurobiol ; 178(1): 146-55, 2011 Aug 31.
Article in English | MEDLINE | ID: mdl-21527363

ABSTRACT

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.


Subject(s)
Embryo, Mammalian/physiology , Respiratory Center/embryology , Animals , Mice
7.
J Neurosci Res ; 88(16): 3555-65, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20936702

ABSTRACT

Although extracellular calcium ionic concentration ([Ca](o) ) is known to increase during late gestation and to drop after parturition, little is known about the influence of [Ca](o) on fetal brain function. We have investigated the influence of [Ca](o) , calcium-sensing receptors/nonselective cation currents (CaSR/NSCC), and GABAergic inhibitions on maturation of brainstem-spinal motor activities: the primary low-frequency embryonic rhythm [LF; silent since embryonic day (E)16] and the fetal respiratory rhythm (RR; emerging at E14-E15). Using in vitro isolated brainstem-spinal cord preparations of mice at different fetal and postnatal (P) stages (E16-P1), we demonstrate that reducing fetal [Ca](o) from 1.2 mM to 0.7 mM at E16-E18 or blocking GABA(A) receptors at E16-P0 reactivates LF and reveals LF-related disturbance of RR at E16-E18. This LF is stopped by adding gadolinium or spermidine (CaSR/NSCC agonists) at E18-P0 or GABA(A) receptor agonists at E16-E18. In contrast, [Ca](o) -induced slowing of RR at E16-E18 is not reproduced by gadolinium and spermidine. We conclude that perinatal CaSR/NSCC and GABA(A) inhibition allow quiescence of the LF, thereby improving functional maturation of the RR.


Subject(s)
Biological Clocks/physiology , Calcium Signaling/physiology , Motor Neurons/metabolism , Receptors, Calcium-Sensing/metabolism , Receptors, GABA-A/metabolism , Respiratory Center/embryology , Animals , Brain Stem/cytology , Brain Stem/embryology , Brain Stem/metabolism , Brain Waves/physiology , Calcium/metabolism , Cations , Embryo, Mammalian , Fetal Development/physiology , In Vitro Techniques , Ion Channels/metabolism , Mice , Respiratory Center/cytology , Respiratory Center/metabolism , Respiratory Mechanics/physiology , Spinal Cord/cytology , Spinal Cord/embryology , Spinal Cord/metabolism
8.
J Neurosci ; 29(47): 14836-46, 2009 Nov 25.
Article in English | MEDLINE | ID: mdl-19940179

ABSTRACT

The retrotrapezoid nucleus (RTN) is a group of neurons in the rostral medulla, defined here as Phox2b-, Vglut2-, neurokinin1 receptor-, and Atoh1-expressing cells in the parafacial region, which have been proposed to function both as generators of respiratory rhythm and as central respiratory chemoreceptors. The present study was undertaken to assess these two putative functions using genetic tools. We generated two conditional Phox2b mutations, which target different subsets of Phox2b-expressing cells, but have in common a massive depletion of RTN neurons. In both conditional mutants as well as in the previously described Phox2b(27Ala) mutants, in which the RTN is also compromised, the respiratory-like rhythmic activity normally seen in the parafacial region of fetal brainstem preparations was completely abrogated. Rhythmic motor bursts were recorded from the phrenic nerve roots in the mutants, but their frequency was markedly reduced. Both the rhythmic activity in the RTN region and the phrenic nerve discharges responded to a low pH challenge in control, but not in the mutant embryos. Together, our results provide genetic evidence for the essential role of the Phox2b-expressing RTN neurons both in establishing a normal respiratory rhythm before birth and in providing chemosensory drive.


Subject(s)
Chemoreceptor Cells/metabolism , Homeodomain Proteins/genetics , Respiration , Respiratory Center/metabolism , Rhombencephalon/metabolism , Transcription Factors/genetics , Action Potentials/physiology , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Nerve Net/embryology , Nerve Net/metabolism , Nerve Net/physiopathology , Organ Culture Techniques , Phrenic Nerve/physiology , Respiratory Center/embryology , Respiratory Center/physiopathology , Rhombencephalon/embryology , Rhombencephalon/physiopathology
10.
Nat Neurosci ; 12(8): 1028-35, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19578380

ABSTRACT

The hindbrain transcription factors Phox2b and Egr2 (also known as Krox20) are linked to the development of the autonomic nervous system and rhombomere-related regulation of breathing, respectively. Mutations in these proteins can lead to abnormal breathing behavior as a result of an alteration in an unidentified neuronal system. We characterized a bilateral embryonic parafacial (e-pF) population of rhythmically bursting neurons at embryonic day (E) 14.5 in mice. These cells expressed Phox2b, were derived from Egr2-expressing precursors and their development was dependent on the integrity of the Egr2 gene. Silencing or eliminating the e-pF oscillator, but not the putative inspiratory oscillator (preBötzinger complex, preBötC), led to an abnormally slow rhythm, demonstrating that the e-pF controls the respiratory rhythm. The e-pF oscillator, the only one active at E14.5, entrained and then coupled with the preBötC, which emerged independently at E15.5. These data establish the dual organization of the respiratory rhythm generator at the time of its inception, when it begins to drive fetal breathing.


Subject(s)
Biological Clocks/genetics , Brain Stem/embryology , Brain Stem/metabolism , Gene Expression Regulation, Developmental/genetics , Respiratory Center/embryology , Respiratory Center/metabolism , Animals , Brain Stem/cytology , Cell Differentiation/genetics , Early Growth Response Protein 2/genetics , Early Growth Response Protein 2/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Inhalation/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Nerve Net/cytology , Nerve Net/embryology , Nerve Net/metabolism , Neurogenesis/genetics , Neurons/cytology , Neurons/metabolism , Periodicity , Respiratory Center/cytology , Respiratory Physiological Phenomena/genetics , Reticular Formation/cytology , Reticular Formation/embryology , Reticular Formation/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Respir Physiol Neurobiol ; 168(1-2): 86-91, 2009 Aug 31.
Article in English | MEDLINE | ID: mdl-19560563

ABSTRACT

Breathing is vital for life ex utero and therefore requires that the respiratory rhythm generator (RRG), the central neural network generating the continuous rhythmic motor command, be functional at birth. The RRG, located in the brainstem, appears to comprise two interacting respiratory oscillators: the parafacial respiratory group (pFRG), and the preBötzinger complex (preBötC). Data on the establishment of these respiratory oscillators during embryonic and foetal periods are beginning to be produced. The present paper provides a short review of the current knowledge regarding: (i) the emergence of activity in the two respiratory oscillators and (ii) their functional coupling during prenatal development in rodents.


Subject(s)
Brain Stem/embryology , Periodicity , Respiration , Respiratory Center/embryology , Animals , Brain Stem/cytology , Brain Stem/growth & development , Neural Pathways/cytology , Neural Pathways/embryology , Neural Pathways/growth & development , Neurons/physiology , Respiratory Center/physiology , Rodentia/physiology
12.
J Comp Neurol ; 506(5): 775-90, 2008 Feb 10.
Article in English | MEDLINE | ID: mdl-18076058

ABSTRACT

Serotonin receptors (5-HTRs) are known to be involved in the regulation of breathing behavior and to mediate neurotrophic actions that exert a significant function in network formation during development. We studied neuronal 5-HT(4(a))R-immunoreactivity (-IR) at developmental ages from E14 to P10. Within the pre-Bötzinger complex (pre-BötC), a part of the respiratory network important for rhythmogenesis, 5-HT(4(a))R-IR was most extensive in rats at an age of E18. The 5-HT(4(a))-IR was found predominantly in the neuropil, whereas somatic staining was sporadic at late embryonic (E18-E20) stages. At birth, we observed a dramatic change to a predominantly somatic staining, and neuropil staining was greatly reduced and disappeared at an age of P4. In all developmental stages, 5-HT(4(a)) and mu-opioid receptors were strongly coexpressed in neurons of the pre-BötC, whereas 5-HT(4(a))R expression was absent in neurons within the dorsal horn. Nestin, a marker for CNS progenitor cells, was used to obtain information about the degree of pre-BötC differentiation. Nestin-positive cells did not appear within the pre-BötC before age E20. At E16, nestin-expressing cells were absent in the nucleus ambiguus (NA) and its ventral periphery. The number of nestin-positive cells increased after birth within and outside the pre-BötC, the majority of cells being glial. Coexpression of nestin and 5-HT(4(a))R was localized predominantly within the NA and appeared only sporadically within the pre-BötC. We conclude that 5-HT(4(a))Rs are important not only for neuromodulation of cellular excitability but also for respiratory network formation.


Subject(s)
Gene Expression Regulation, Developmental , Neurons/metabolism , Receptors, Serotonin, 5-HT4/metabolism , Respiratory Center/metabolism , Animals , Circadian Rhythm/physiology , Immunohistochemistry , In Vitro Techniques , Intermediate Filament Proteins/metabolism , Medulla Oblongata/embryology , Medulla Oblongata/growth & development , Medulla Oblongata/metabolism , Nerve Tissue Proteins/metabolism , Nestin , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/metabolism , Respiratory Center/embryology , Respiratory Center/growth & development , Stem Cells , Tissue Distribution
13.
Neuroscience ; 148(1): 140-50, 2007 Aug 10.
Article in English | MEDLINE | ID: mdl-17629626

ABSTRACT

Using voltage-sensitive dye recording, we surveyed neural responses related to the vagus nerve in the embryonic chick brainstem. In our previous studies, we identified four vagus nerve-related response areas in the brainstem. On the stimulated side, they included (1) the nucleus of the tractus solitarius (NTS: the primary sensory nucleus) and (2) the dorsal motor nucleus of the vagus nerve (DMNV), whereas on the contralateral side, they corresponded to (3) the parabrachial nucleus (PBN: the second/higher-ordered nucleus) and (4) the medullary non-NTS region. In the present study, in addition to these areas, we identified another response area circumflex to the obex. The intensity of the optical signal in the response area was much smaller than that in the NTS/DMNV, and the spatio-temporal pattern could be discerned after signal averaging. The conduction rate to the response area was slower than that to the other four areas. Ontogenetically, the response area was distributed on the stimulated side at the 6-day embryonic stage, and it spread into the contralateral side in 7- and 8-day embryonic stages. These distribution patterns were consistent with projection patterns of vagal afferent fibers stained with a fluorescent tracer, suggesting that the response area included a primary sensory nucleus. In comparison with the functional development of the other four response areas, we traced the functional organization of vagus nerve-related nuclei in the embryonic brainstem.


Subject(s)
Afferent Pathways/embryology , Brain Stem/embryology , Neurons, Afferent/cytology , Vagus Nerve/embryology , Action Potentials/physiology , Afferent Pathways/physiology , Animals , Area Postrema/embryology , Area Postrema/physiology , Axons/physiology , Axons/ultrastructure , Brain Mapping/methods , Brain Stem/physiology , Carbocyanines , Chick Embryo , Electric Stimulation , Electrophysiology/methods , Excitatory Postsynaptic Potentials/physiology , Fluorescent Dyes , Fourth Ventricle/anatomy & histology , Functional Laterality/physiology , Neurons, Afferent/physiology , Optics and Photonics , Organ Culture Techniques , Respiratory Center/embryology , Respiratory Center/physiology , Solitary Nucleus/embryology , Solitary Nucleus/physiology , Staining and Labeling/methods , Vagus Nerve/physiology
14.
J Neurobiol ; 66(12): 1285-301, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16967510

ABSTRACT

How regional patterning of the neural tube in vertebrate embryos may influence the emergence and the function of neural networks remains elusive. We have begun to address this issue in the embryonic mouse hindbrain by studying rhythmogenic properties of different neural tube segments. We have isolated pre- and post-otic hindbrain segments and spinal segments of the mouse neural tube, when they form at embryonic day (E) 9, and grafted them into the same positions in stage-matched chick hosts. Three days after grafting, in vitro recordings of the activity in the cranial nerves exiting the grafts indicate that a high frequency (HF) rhythm (order: 10 bursts/min) is generated in post-otic segments while more anterior pre-otic and more posterior spinal territories generate a low frequency (LF) rhythm (order: 1 burst/min). Comparison with homo-specific grafting of corresponding chick segments points to conservation in mouse and chick of the link between the patterning of activities and the axial origin of the hindbrain segment. This HF rhythm is reminiscent of the respiratory rhythm known to appear at E15 in mice. We also report on pre-/post-otic interactions. The pre-otic rhombomere 5 prevents the emergence of the HF rhythm at E12. Although the nature of the interaction with r5 remains obscure, we propose that ontogeny of fetal-like respiratory circuits relies on: (i) a selective developmental program enforcing HF rhythm generation, already set at E9 in post-otic segments, and (ii) trans-segmental interactions with pre-otic territories that may control the time when this rhythm appears.


Subject(s)
Branchial Region/embryology , Efferent Pathways/embryology , Respiration , Respiratory Center/embryology , Rhombencephalon/embryology , Spinal Cord/embryology , Action Potentials/physiology , Animals , Body Patterning/physiology , Brain Tissue Transplantation/methods , Branchial Region/physiology , Chick Embryo , Cranial Nerves/embryology , Cranial Nerves/physiology , Efferent Pathways/physiology , Embryonic Development/physiology , Mice , Respiratory Center/physiology , Rhombencephalon/physiology , Species Specificity , Spinal Cord/physiology , Transplantation Chimera/embryology , Transplantation Chimera/physiology
15.
Brain Res ; 1090(1): 45-50, 2006 May 23.
Article in English | MEDLINE | ID: mdl-16643861

ABSTRACT

Adrenaline is a potent respiratory regulator. However, adrenergic contribution to the developing respiratory center has not been studied extensively. Adrenaline application on embryonic day 17 medulla-spinal cord block preparations abolished non-respiratory activity and enhanced respiratory frequency. Phentolamine application on neonatal blocks that produced stable neonatal respiration resulted in respiratory destabilization. These results suggest that central adrenergic modulation is involved in fetal respiratory development and maintenance of stable respiration.


Subject(s)
Efferent Pathways/embryology , Epinephrine/metabolism , Medulla Oblongata/embryology , Neurons/metabolism , Respiratory Center/embryology , Spinal Cord/embryology , Action Potentials/drug effects , Action Potentials/physiology , Adrenergic alpha-Antagonists/pharmacology , Aging/drug effects , Aging/physiology , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cell Differentiation/physiology , Efferent Pathways/growth & development , Efferent Pathways/metabolism , Epinephrine/pharmacology , Medulla Oblongata/growth & development , Medulla Oblongata/metabolism , Nerve Net/embryology , Nerve Net/growth & development , Nerve Net/metabolism , Neurons/drug effects , Organ Culture Techniques , Phentolamine/pharmacology , Rats , Rats, Wistar , Respiratory Center/growth & development , Respiratory Center/metabolism , Respiratory Physiological Phenomena/drug effects , Spinal Cord/growth & development , Spinal Cord/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
16.
J Physiol ; 570(Pt 3): 437-44, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16284077

ABSTRACT

By birth, the regulatory neural network responsible for respiratory control is capable of generating robust rhythm-driving ventilation that can adjust to homeostatic needs. The advent of in vitro models isolated from prenatal rodents has significantly advanced our understanding of these processes. In this topical review, we examine the development of medullary respiratory rhythm-generating centres and phrenic motoneurone-diaphragm properties during the prenatal period.


Subject(s)
Phrenic Nerve/embryology , Phrenic Nerve/physiology , Respiratory Center/embryology , Respiratory Center/physiology , Respiratory Mechanics/physiology , Animals , Fetus/physiology , Motor Neurons/physiology , Phrenic Nerve/cytology , Respiratory Center/cytology
17.
Respir Physiol Neurobiol ; 149(1-3): 17-27, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16203211

ABSTRACT

The survival of neonatal mammals requires a correct function of the respiratory rhythm generator (RRG), and therefore, the processes that control its prenatal maturation are of vital importance. In humans, lambs and rodents, foetal breathing movements (FBMs) occur early during gestation, are episodic, sensitive to bioamines, central hypoxia and inputs from CNS upper structures, and evolve with developmental age. In vitro, the foetal rodent RRG studied in preparations where the upper CNS structures are lacking continuously produces a rhythmic command, which is sensitive to hypoxia and bioaminergic inputs. The rhythm is slow with variable periods 4 days before birth. It becomes faster 2 days before birth, similar to the postnatal rhythm. Compelling evidence suggests that a region of the RRG called the preBötzinger complex (PBC) contains respiratory pacemaker neurones which play a primary role in perinatal rhythmogenesis. Although the RRG functions during early gestation, no pacemakers are found in the putative PBC area and its electrical stimulation and lesion do not affect the early foetal rhythm. To know whether the early foetal and perinatal rhythms originate from either pacemaker neurones or network connection properties, and to know which maturational processes might explain the appearance of PBC pacemakers and the rhythm increase during perinatal development, we computationally modelled maturing RRG. Our model shows that both network noise and persistent sodium conductance are crucial for rhythmogenesis and that a slight increase in the persistent sodium conductance can solve the pacemaker versus network dilemma in a noisy network.


Subject(s)
Computer Simulation , Periodicity , Respiratory Center/embryology , Respiratory Center/growth & development , Respiratory Physiological Phenomena , Animals , Animals, Newborn , Humans , Infant, Newborn
18.
Respir Physiol Neurobiol ; 149(1-3): 29-41, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-15914099

ABSTRACT

Compared with birds and mammals, very little is known about the development and regulation of respiratory rhythm generation in ectothermic vertebrates. The development and regulation of respiratory rhythm generation in ectothermic vertebrates (fish, amphibians and reptiles) should provide insight into the evolution of these mechanisms. One useful model for examining the development of respiratory rhythm generation in ectothermic vertebrates has emerged from studies with the North American bullfrog (Rana catesbeiana). A major advantage of bullfrogs as a comparative model for respiratory rhythm generation is that respiratory output may be measured at all stages of development, both in vivo and in vitro. An emerging view of recent studies in developing bullfrogs is that many of the mechanisms of respiratory rhythm generation are very similar to those seen in birds and mammals. The overall conclusion from these studies is that respiratory rhythm generation during development may be highly conserved during evolution. The development of respiratory rhythm generation in mammals may, therefore, reflect the antecedent mechanisms seen in ectothermic vertebrates. The main focus of this brief review is to discuss recent data on the development of respiratory rhythm generation in ectothermic vertebrates, with particular emphasis on the North American bullfrog (R. catesbeiana) as a model.


Subject(s)
Amphibians/physiology , Rana catesbeiana/physiology , Respiratory Physiological Phenomena , Animals , Periodicity , Respiratory Center/embryology , Respiratory Center/growth & development , Respiratory System/embryology , Respiratory System/growth & development
19.
J Neurosci ; 25(17): 4307-18, 2005 Apr 27.
Article in English | MEDLINE | ID: mdl-15858057

ABSTRACT

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.


Subject(s)
Brain Stem/cytology , Motor Neurons/physiology , Periodicity , Respiration , Respiratory Center/physiology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Age Factors , Analgesics, Opioid/pharmacology , Animals , Calcium/metabolism , Cholera Toxin/pharmacology , Diagnostic Imaging/methods , Dose-Response Relationship, Radiation , Electric Stimulation/methods , Embryo, Mammalian , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Female , Fluorescent Antibody Technique/methods , In Vitro Techniques , Male , Mice , Patch-Clamp Techniques/methods , Pregnancy , Receptors, Neurokinin-1/metabolism , Respiratory Center/drug effects , Respiratory Center/embryology , Substance P/pharmacology , gamma-Aminobutyric Acid/pharmacology
20.
J Neurosci ; 24(47): 10693-701, 2004 Nov 24.
Article in English | MEDLINE | ID: mdl-15564586

ABSTRACT

Na+, K+-ATPase 2 subunit gene (Atp1a2) knock-out homozygous mice (Atp1a2-/-) died immediately after birth resulting from lack of breathing. The respiratory-related neuron activity in Atp1a2-/- was investigated using a brainstem-spinal cord en bloc preparation. The respiratory motoneuron activity recorded from the fourth cervical ventral root (C4) was defective in Atp1a2-/- fetuses of embryonic day 18.5. The C4 response to electrical stimulation of the ventrolateral medulla (VLM) recovered more slowly in Atp1a2-/- than in wild type during superfusion with Krebs' solution, consistent with the high extracellular GABA in brain of Atp1a2-/-. Lack of inhibitory neural activities in VLM of Atp1a2-/- was observed by optical recordings. High intracellular Cl- concentrations in neurons of the VLM of Atp1a2-/- were detected in gramicidin-perforated patch-clamp recordings. The alpha2 subunit and a neuron-specific K-Cl cotransporter KCC2 were coimmunoprecipitated in a purified synaptic membrane fraction of wild-type fetuses. Based on these results, we propose a model for functional coupling between the Na+, K+-ATPase alpha2 subunit and KCC2, which excludes Cl- from the cytosol in respiratory center neurons.


Subject(s)
Chlorides/metabolism , Neurons/physiology , Respiratory Center/metabolism , Sodium-Potassium-Exchanging ATPase/physiology , Symporters/physiology , Amino Acid Sequence , Animals , Apnea/embryology , Apnea/genetics , Apnea/physiopathology , Electric Stimulation , Facial Nerve/physiology , Fetus/metabolism , Fetus/physiology , Homeostasis/physiology , Isoenzymes/physiology , Mice , Mice, Knockout , Molecular Sequence Data , Neural Inhibition/physiology , Neurons/metabolism , Patch-Clamp Techniques , Potassium/metabolism , Respiratory Center/cytology , Respiratory Center/embryology , Respiratory Center/physiology , Sodium-Potassium-Exchanging ATPase/genetics , Spinal Nerve Roots/physiology , Symporters/genetics , Symporters/metabolism , gamma-Aminobutyric Acid/physiology , K Cl- Cotransporters
SELECTION OF CITATIONS
SEARCH DETAIL
...