Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38765982

RESUMO

Microglia are innate CNS immune cells that play key roles in supporting key CNS functions including brain plasticity. We now report a previously unknown role for microglia in regulating neuroplasticity within spinal phrenic motor neurons, the neurons driving diaphragm contractions and breathing. We demonstrate that microglia regulate phrenic long-term facilitation (pLTF), a form of respiratory memory lasting hours after repetitive exposures to brief periods of low oxygen (acute intermittent hypoxia; AIH) via neuronal/microglial fractalkine signaling. AIH-induced pLTF is regulated by the balance between competing intracellular signaling cascades initiated by serotonin vs adenosine, respectively. Although brainstem raphe neurons release the relevant serotonin, the cellular source of adenosine is unknown. We tested a model in which hypoxia initiates fractalkine signaling between phrenic motor neurons and nearby microglia that triggers extracellular adenosine accumulation. With moderate AIH, phrenic motor neuron adenosine 2A receptor activation undermines serotonin-dominant pLTF; in contrast, severe AIH drives pLTF by a unique, adenosine-dominant mechanism. Phrenic motor neuron fractalkine knockdown, cervical spinal fractalkine receptor inhibition on nearby microglia, and microglial depletion enhance serotonin-dominant pLTF with moderate AIH but suppress adenosine-dominant pLTF with severe AIH. Thus, microglia play novel functions in the healthy spinal cord, regulating hypoxia-induced neuroplasticity within the motor neurons responsible for breathing.

2.
J Physiol ; 599(15): 3771-3797, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34142718

RESUMO

KEY POINTS: While respiratory complications following opioid use are mainly mediated via activation of mu opioid receptors, long-latency off-target signalling via innate immune toll-like receptor 4 (TLR4) may impair other essential elements of breathing control such as respiratory motor plasticity. In adult rats, pre-treatment with a single dose of morphine blocked long-term facilitation (LTF) of phrenic motor output via a long-latency TLR4-dependent mechanism. In the phrenic motor nucleus, morphine triggered TLR4-dependent activation of microglial p38 MAPK - a key enzyme that orchestrates inflammatory signalling and is known to undermine phrenic LTF. Morphine-induced LTF loss may destabilize breathing, potentially contributing to respiratory side effects. Therefore, we suggest minimizing TLR-4 signalling may improve breathing stability during opioid therapy. ABSTRACT: Opioid-induced respiratory dysfunction is a significant public health burden. While respiratory effects are mediated via mu opioid receptors, long-latency off-target opioid signalling through innate immune toll-like receptor 4 (TLR4) may modulate essential elements of breathing control, particularly respiratory motor plasticity. Plasticity in respiratory motor circuits contributes to the preservation of breathing in the face of destabilizing influences. For example, respiratory long-term facilitation (LTF), a well-studied model of respiratory motor plasticity triggered by acute intermittent hypoxia, promotes breathing stability by increasing respiratory motor drive to breathing muscles. Some forms of respiratory LTF are exquisitely sensitive to inflammation and are abolished by even a mild inflammation triggered by TLR4 activation (e.g. via systemic lipopolysaccharides). Since opioids induce inflammation and TLR4 activation, we hypothesized that opioids would abolish LTF through a TLR4-dependent mechanism. In adult Sprague Dawley rats, pre-treatment with a single systemic injection of the prototypical opioid agonist morphine blocks LTF expression several hours later in the phrenic motor system - the motor pool driving diaphragm muscle contractions. Morphine blocked phrenic LTF via TLR4-dependent mechanisms because pre-treatment with (+)-naloxone - the opioid inactive stereoisomer and novel small molecule TLR4 inhibitor - prevented impairment of phrenic LTF in morphine-treated rats. Morphine triggered TLR4-dependent activation of microglial p38 MAPK within the phrenic motor system - a key enzyme that orchestrates inflammatory signalling and undermines phrenic LTF. Morphine-induced LTF loss may destabilize breathing, potentially contributing to respiratory side effects. We suggest minimizing TLR-4 signalling may improve breathing stability during opioid therapy by restoring endogenous mechanisms of plasticity within respiratory motor circuits.


Assuntos
Morfina , Nervo Frênico , Receptor 4 Toll-Like , Animais , Hipóxia , Morfina/farmacologia , Plasticidade Neuronal , Ratos , Ratos Sprague-Dawley , Medula Espinal
3.
J Neurotrauma ; 38(9): 1292-1305, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33446048

RESUMO

"Low-dose" acute intermittent hypoxia (AIH; 3-15 episodes/day) is emerging as a promising therapeutic strategy to improve motor function after incomplete cervical spinal cord injury (cSCI). Conversely, chronic "high-dose" intermittent hypoxia (CIH; > 80-100 episodes/day) elicits multi-system pathology and is a hallmark of sleep apnea, a condition highly prevalent in individuals with cSCI. Whereas daily AIH (dAIH) enhances phrenic motor plasticity in intact rats, it is abolished by CIH. However, there have been no direct comparisons of prolonged dAIH versus CIH on phrenic motor outcomes after chronic cSCI. Thus, phrenic nerve activity and AIH-induced phrenic long-term facilitation (pLTF) were assessed in anesthetized rats. Experimental groups included: 1) intact rats exposed to 28 days of normoxia (Nx28; 21% O2; 8 h/day), and three groups with chronic C2 hemisection (C2Hx) exposed to either: 2) Nx28; 3) dAIH (dAIH28; 10, 5-min episodes of 10.5% O2/day; 5-min intervals); or 4) CIH (IH28-2/2; 2-min episodes; 2-min intervals; 8 h/day). Baseline ipsilateral phrenic nerve activity was reduced in injured versus intact rats but unaffected by dAIH28 or IH28-2/2. There were no group differences in contralateral phrenic activity. pLTF was enhanced bilaterally by dAIH28 versus Nx28 but unaffected by IH28-2/2. Whereas dAIH28 enhanced pLTF after cSCI, it did not improve baseline phrenic output. In contrast, unlike shorter protocols in intact rats, CIH28-2/2 did not abolish pLTF in chronic C2Hx. Mechanisms of differential responses to dAIH versus CIH are not yet known, particularly in the context of cSCI. Further, it remains unclear whether enhanced phrenic motor plasticity can improve breathing after cSCI.


Assuntos
Medula Cervical/lesões , Hipóxia/metabolismo , Plasticidade Neuronal/fisiologia , Nervo Frênico/metabolismo , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/terapia , Animais , Pressão Sanguínea/fisiologia , Frequência Cardíaca/fisiologia , Precondicionamento Isquêmico/métodos , Masculino , Neurônios Motores/metabolismo , Ratos , Ratos Sprague-Dawley
4.
J Neuroinflammation ; 18(1): 28, 2021 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-33468163

RESUMO

BACKGROUND: Inflammation undermines multiple forms of neuroplasticity. Although inflammation and its influence on plasticity in multiple neural systems has been extensively studied, its effects on plasticity of neural networks controlling vital life functions, such as breathing, are less understood. In this study, we investigated the signaling mechanisms whereby lipopolysaccharide (LPS)-induced systemic inflammation impairs plasticity within the phrenic motor system-a major spinal respiratory motor pool that drives contractions of the diaphragm muscle. Here, we tested the hypotheses that lipopolysaccharide-induced systemic inflammation (1) blocks phrenic motor plasticity by a mechanism that requires cervical spinal okadaic acid-sensitive serine/threonine protein phosphatase (PP) 1/2A activity and (2) prevents phosphorylation/activation of extracellular signal-regulated kinase 1/2 mitogen activated protein kinase (ERK1/2 MAPK)-a key enzyme necessary for the expression of phrenic motor plasticity. METHODS: To study phrenic motor plasticity, we utilized a well-characterized model for spinal respiratory plasticity called phrenic long-term facilitation (pLTF). pLTF is characterized by a long-lasting, progressive enhancement of inspiratory phrenic nerve motor drive following exposures to moderate acute intermittent hypoxia (mAIH). In anesthetized, vagotomized and mechanically ventilated adult Sprague Dawley rats, we examined the effect of inhibiting cervical spinal serine/threonine PP 1/2A activity on pLTF expression in sham-vehicle and LPS-treated rats. Using immunofluorescence optical density analysis, we compared mAIH-induced phosphorylation/activation of ERK 1/2 MAPK with and without LPS-induced inflammation in identified phrenic motor neurons. RESULTS: We confirmed that mAIH-induced pLTF is abolished 24 h following low-dose systemic LPS (100 µg/kg, i.p.). Cervical spinal delivery of the PP 1/2A inhibitor, okadaic acid, restored pLTF in LPS-treated rats. LPS also prevented mAIH-induced enhancement in phrenic motor neuron ERK1/2 MAPK phosphorylation. Thus, a likely target for the relevant okadaic acid-sensitive protein phosphatases is ERK1/2 MAPK or its upstream activators. CONCLUSIONS: This study increases our understanding of fundamental mechanisms whereby inflammation disrupts neuroplasticity in a critical population of motor neurons necessary for breathing, and highlights key roles for serine/threonine protein phosphatases and ERK1/2 MAPK kinase in the plasticity of mammalian spinal respiratory motor circuits.


Assuntos
Inflamação/metabolismo , Neurônios Motores/enzimologia , Plasticidade Neuronal/fisiologia , Fosfoproteínas Fosfatases/metabolismo , Nervo Frênico/enzimologia , Animais , Inflamação/induzido quimicamente , Lipopolissacarídeos/toxicidade , Potenciação de Longa Duração/fisiologia , Sistema de Sinalização das MAP Quinases , Masculino , Ratos , Ratos Sprague-Dawley , Fenômenos Fisiológicos Respiratórios
5.
J Appl Physiol (1985) ; 127(2): 432-443, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31219768

RESUMO

Serotonin (5-HT) is a key regulator of spinal respiratory motor plasticity. For example, spinal 5-HT receptor activation is necessary for the induction of phrenic long-term facilitation (pLTF), a form of respiratory motor plasticity triggered by moderate acute intermittent hypoxia (mAIH). mAIH-induced pLTF is blocked by cervical spinal application of the broad-spectrum 5-HT-receptor antagonist, methysergide. However, methysergide does not allow distinctions between the relative contributions of different 5-HT receptor subtypes. Intravenous administration of the Gq protein-coupled 5-HT2A/2C receptor antagonist ketanserin blocks mAIH-induced pLTF when administered before, but not after, mAIH; thus, 5-HT2 receptor activation is necessary for the induction but not maintenance of mAIH-induced pLTF. However, systemic ketanserin administration does not identify the site of the relevant 5-HT2A/2C receptors. Furthermore, this approach does not differentiate between the roles of 5-HT2A versus 5-HT2C receptors, nor does it preclude involvement of other Gq protein-coupled metabotropic 5-HT receptors capable of eliciting long-lasting phrenic motor facilitation, such as 5-HT2B receptors. Here we tested the hypothesis that mAIH-induced pLTF requires cervical spinal 5-HT2 receptor activation and determined which 5-HT2 receptor subtypes are involved. Anesthetized, paralyzed, and ventilated adult male Sprague Dawley rats were pretreated intrathecally with cervical (~C3-C5) spinal injections of subtype selective 5-HT2A/2C, 5-HT2B, or 5-HT2C receptor antagonists before mAIH. Whereas cervical spinal 5-HT2C receptor inhibition had no impact on mAIH-induced pLTF, pLTF was no longer observed after pretreatment with either 5-HT2A/2C or 5-HT2B receptor antagonists. Furthermore, spinal pretreatment with an MEK/ERK MAPK inhibitor blocked phrenic motor facilitation elicited by intrathecal injections of 5-HT2A but not 5-HT2B receptor agonists. Thus, mAIH-induced pLTF requires concurrent cervical spinal activation of both 5-HT2A and 5-HT2B receptors. However, these distinct receptor subtypes contribute to phrenic motor facilitation via distinct downstream signaling cascades that differ in their requirement for ERK MAPK signaling. The demonstration that both 5-HT2A and 5-HT2B receptors make unique contributions to mAIH-induced pLTF advances our understanding of mechanisms that underlie 5-HT-induced phrenic motor plasticity.NEW & NOTEWORTHY Moderate acute intermittent hypoxia (mAIH) triggers a persistent enhancement in phrenic motor output, an effect termed phrenic long-term facilitation (pLTF). mAIH-induced pLTF is blocked by cervical spinal application of the broad-spectrum serotonin (5-HT) receptor antagonist methysergide, demonstrating the need for spinal 5-HT receptor activation. However, the exact type of 5-HT receptors required for initiation of pLTF remains unknown. To the best of out knowledge, the present study is the first to demonstrate that 1) spinal coactivation of two distinct Gq protein-coupled 5-HT2 receptor subtypes is necessary for mAIH-induced pLTF, and 2) these receptors contribute to pLTF via cascades that differ in their requirement for ERK MAPK signaling.


Assuntos
Hipóxia/metabolismo , Potenciação de Longa Duração/fisiologia , Nervo Frênico/metabolismo , Receptores de Serotonina/metabolismo , Medula Espinal/metabolismo , Animais , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Nervo Frênico/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Serotonina/metabolismo , Antagonistas da Serotonina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Medula Espinal/efeitos dos fármacos
6.
Sci Rep ; 8(1): 12709, 2018 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-30139983

RESUMO

The respiratory control system is plastic. It has a working memory and is capable of retaining how respiratory stimuli affect breathing by regulating synaptic strength between respiratory neurons. For example, repeated airway obstructions trigger a form of respiratory plasticity that strengthens inspiratory activity of hypoglossal (XII) motoneurons. This form of respiratory plasticity is known as long-term facilitation (LTF) and requires noradrenaline released onto XII motoneurons. However, the brainstem regions responsible for this form of LTF remain unidentified. Here, we used electrophysiology, neuropharmacology and immunohistochemistry in adult rats to identify the brainstem regions involved in mediating LTF. First, we show that repeated airway obstructions induce LTF of XII motoneuron activity and that inactivation of the noradrenergic system prevents LTF. Second, we show that noradrenergic cells in the locus coeruleus (LC), which project to XII motoneurons, are recruited during LTF induction. Third, we show that targeted inactivation of noradrenergic LC cells during LTF induction prevents LTF. And lastly, we show that the nucleus tractus solitarius (NTS), which has known projections to the LC, is critical for LTF because its inactivation prevents LTF. Our results suggest that both the LC and NTS are involved in mediating apnea-induced LTF, and we hypothesize that a NTS → LC → XII circuit mechanism mediates this form of respiratory motor plasticity.


Assuntos
Apneia/metabolismo , Tronco Encefálico/metabolismo , Núcleo Celular/metabolismo , Animais , Apneia/fisiopatologia , Tronco Encefálico/fisiologia , Núcleo Celular/fisiologia , Eletrofisiologia , Nervo Hipoglosso/metabolismo , Nervo Hipoglosso/fisiologia , Potenciação de Longa Duração/fisiologia , Masculino , Neurônios Motores/citologia , Neurônios Motores/fisiologia , Plasticidade Neuronal/fisiologia , Ratos , Ratos Sprague-Dawley , Sistema Respiratório/metabolismo , Sistema Respiratório/fisiopatologia
7.
J Physiol ; 596(8): 1501-1512, 2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29388230

RESUMO

KEY POINTS: Although adenosine 2A (A2A ) receptor activation triggers specific cell signalling cascades, the ensuing physiological outcomes depend on the specific cell type expressing these receptors. Cervical spinal adenosine 2A (A2A ) receptor activation elicits a prolonged facilitation in phrenic nerve activity, which was nearly abolished following intrapleural A2A receptor siRNA injections. A2A receptor siRNA injections selectively knocked down A2A receptors in cholera toxin B-subunit-identified phrenic motor neurons, sparing cervical non-phrenic motor neurons. Collectively, our results support the hypothesis that phrenic motor neurons express the A2A receptors relevant to A2A receptor-induced phrenic motor facilitation. Upregulation of A2A receptor expression in the phrenic motor neurons per se may potentially be a useful approach to increase phrenic motor neuron excitability in conditions such as spinal cord injury. ABSTRACT: Cervical spinal adenosine 2A (A2A ) receptor activation elicits a prolonged increase in phrenic nerve activity, an effect known as phrenic motor facilitation (pMF). The specific cervical spinal cells expressing the relevant A2A receptors for pMF are unknown. This is an important question since the physiological outcome of A2A receptor activation is highly cell type specific. Thus, we tested the hypothesis that the relevant A2A receptors for pMF are expressed in phrenic motor neurons per se versus non-phrenic neurons of the cervical spinal cord. A2A receptor immunostaining significantly colocalized with NeuN-positive neurons (89 ± 2%). Intrapleural siRNA injections were used to selectively knock down A2A receptors in cholera toxin B-subunit-labelled phrenic motor neurons. A2A receptor knock-down was verified by a ∼45% decrease in A2A receptor immunoreactivity within phrenic motor neurons versus non-targeting siRNAs (siNT; P < 0.05). There was no evidence for knock-down in cervical non-phrenic motor neurons. In rats that were anaesthetized, subjected to neuromuscular blockade and ventilated, pMF induced by cervical (C3-4) intrathecal injections of the A2A receptor agonist CGS21680 was greatly attenuated in siA2A (21%) versus siNT treated rats (147%; P < 0.01). There were no significant effects of siA2A on phrenic burst frequency. Collectively, our results support the hypothesis that phrenic motor neurons express the A2A receptors relevant to A2A receptor-induced pMF.


Assuntos
Neurônios Motores/metabolismo , Nervo Frênico/metabolismo , Receptor A2A de Adenosina/metabolismo , Potenciais de Ação , Agonistas do Receptor A2 de Adenosina/farmacologia , Animais , Toxina da Cólera/farmacologia , Masculino , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/fisiologia , Nervo Frênico/citologia , Nervo Frênico/fisiologia , Ratos , Ratos Sprague-Dawley
8.
J Neurosci ; 30(50): 16886-95, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21159960

RESUMO

The respiratory control system is not just reflexive, it is smart, it learns, and, in fact, it has a memory. The respiratory system listens to and carefully remembers how previous stimuli affect breathing. Respiratory memory is laid down by adjusting synaptic strength between respiratory neurons. For example, repeated hypoxic bouts trigger a form of respiratory memory that functions to strengthen the ability of respiratory motoneurons to trigger contraction of breathing muscles. This type of respiratory plasticity is known as long-term facilitation (LTF). Although chemical feedback, such as hypoxia, initiates LTF, it is unknown whether natural modulation of mechanical feedback (from vagal inputs) also causes motor plasticity. Here, we used reverse microdialysis, electrophysiology, neuropharmacology, and histology to determine whether episodic modulation of vagally mediated mechanical feedback is able to induce respiratory LTF in anesthetized adult rats. We show that repeated obstructive apneas disrupt vagal feedback and trigger LTF of hypoglossal motoneuron activity and genioglossus muscle tone. This same stimulus does not cause LTF of diaphragm activity. Hypoxic episodes do not cause apnea-induced LTF; instead, LTF is triggered by modulation of vagal feedback. Unlike hypoxia-induced respiratory plasticity, vagus-induced LTF does not require 5-HT(2) receptors but instead relies on activation of α1-adrenergic receptors on hypoglossal motoneurons. In summary, we identify a novel form of hypoxia- and 5-HT-independent respiratory motor plasticity that is triggered by physiological modulation of vagal feedback and is mediated by α1-adrenergic receptor activation on (or near) hypoglossal motoneurons.


Assuntos
Retroalimentação Sensorial/fisiologia , Potenciação de Longa Duração/fisiologia , Neurônios Motores/fisiologia , Receptores Adrenérgicos alfa 1/fisiologia , Sistema Respiratório/inervação , Nervo Vago/fisiologia , Antagonistas de Receptores Adrenérgicos alfa 1/farmacologia , Animais , Apneia/fisiopatologia , Diafragma/fisiologia , Nervo Hipoglosso/efeitos dos fármacos , Hipóxia , Ketanserina/farmacologia , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Prazosina/análogos & derivados , Prazosina/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor 5-HT2A de Serotonina/fisiologia , Receptores de AMPA/agonistas , Antagonistas da Serotonina/farmacologia , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia
9.
Adv Exp Med Biol ; 669: 289-92, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20217368

RESUMO

Sleep loss leads to deficits in neuroplasticity that underlie important physiological functions such as learning and memory. However, the influence of sleep loss on respiratory motor plasticity is unclear. In this study, we examined the influence of sleep loss on plasticity of upper airway motor outflow induced by repeated obstructive apneas. Here, we demonstrate that repeated airway obstructions, as experienced during obstructive apnea (OSA), induce a long-term enhancement of upper airway respiratory muscle activity, and that short-term sleep deprivation (6 hours) reduces the magnitude of this response. Our results suggest that respiratory motor plasticity may be reduced or minimized in disease conditions that are characterized by abnormal sleep disturbances (e.g., OSA).


Assuntos
Mecânica Respiratória , Privação do Sono/fisiopatologia , Animais , Masculino , Ratos , Ratos Sprague-Dawley , Sono/fisiologia , Síndromes da Apneia do Sono/fisiopatologia , Fatores de Tempo
10.
Adv Exp Med Biol ; 669: 293-6, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20217369

RESUMO

The respiratory neural network is flexible and can undergo neuronal plasticity. Recent work suggests that neurotrophins and their high-affinity tyrosine kinase (Trk) receptors are involved in mediating plasticity of respiratory motor output elicited by intermittent hypoxia. We aimed to determine whether Trk receptor activation is required for plasticity of upper airway motor outflow induced by repeated obstructive apneas that mimic those experienced in obstructive sleep apnea (OSA). We show that Trk receptor inhibition on hypoglossal motor neurons prevents long-term enhancement of genioglossus muscle tone triggered by repeated airway obstructions in rats. Our result suggests that plasticity of upper airway motor outflow requires a functional neurotrophic signaling cascade. Triggering motor plasticity of upper airways via pharmacological mechanisms could be a potentially useful strategy for improving airway patency in OSA.


Assuntos
Fatores de Crescimento Neural/metabolismo , Mecânica Respiratória/fisiologia , Transdução de Sinais , Animais , Nervo Hipoglosso/efeitos dos fármacos , Nervo Hipoglosso/metabolismo , Nervo Hipoglosso/fisiopatologia , Masculino , Microdiálise , Perfusão , Ratos , Ratos Sprague-Dawley , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/metabolismo , Mecânica Respiratória/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Síndromes da Apneia do Sono/metabolismo , Síndromes da Apneia do Sono/fisiopatologia , Cloreto de Sódio
11.
Adv Exp Med Biol ; 669: 297-301, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20217370

RESUMO

The respiratory network is sensitive to both chemical and mechanosensory feedback and exhibits considerable plasticity when exposed to repeated respiratory perturbations. One example of respiratory plasticity is long-term facilitation (LTF) - a long-term enhancement in respiratory activity induced by intermittent hypoxia. Although intermittent hypoxia ranging from 5-min to 10-s has been shown to trigger respiratory LTF, it is unknown whether brief repeated airway obstruction as experience in obstructive sleep apnea could trigger LTF of respiratory motor outflow. Here we show that brief (15-s), repeated apneas trigger LTF of genioglossus muscle tone in rats. We also show that vagal feedback is important for the expression of apnea-induced LTF because it was not inducible when vagal feedback was impaired. Our results demonstrate a novel mechanism for triggering respiratory neuroplasticity and may have implications for maintenance of airway patency during sleep.


Assuntos
Potenciação de Longa Duração/fisiologia , Músculo Esquelético/fisiologia , Síndromes da Apneia do Sono/fisiopatologia , Animais , Diafragma/fisiologia , Inalação/fisiologia , Masculino , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
13.
J Physiol ; 585(Pt 2): 593-606, 2007 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-17932158

RESUMO

Episodic hypoxia causes repetitive inspiratory activation that induces a form of respiratory plasticity termed long-term facilitation (LTF). While LTF is a function of the hypoxic exposures and inspiratory activation, their relative importance in evoking LTF is unknown. The aims of this study were to: (1) dissociate the relative roles played by episodic hypoxia and respiratory activation in LTF; and (2) determine whether the magnitude of LTF varies as a function of hypoxic intensity. We did this by examining the effects of episodic hypoxia in postnatal rats (15-25 days old), which unlike adult rats exhibit a prominent hypoxia-induced respiratory depression. We quantified inspiratory phrenic nerve activity generated by the in situ working-heart brainstem before, during and for 60 min after episodic hypoxia. We demonstrate that episodic hypoxia evokes LTF despite the fact that it potently suppresses inspiratory activity during individual hypoxic exposures (P < 0.05). Specifically, we show that after episodic hypoxia (three 5 min periods of 10% O2) respiratory frequency increased to 40 +/- 3.3% above baseline values over the next 60 min (P < 0.001). Continuous hypoxia (15 min of 10% O2) had no lasting effects on respiratory frequency (P > 0.05). To determine if LTF magnitude was affected by hypoxic intensity, the episodic hypoxia protocol was repeated under three different O2 tensions. We demonstrate that the magnitude and time course of LTF depend on hypoxic severity, with more intense hypoxia inducing a more potent degree of LTF. We conclude that inspiratory activation is not required for LTF induction, and that hypoxia per se is the physiological stimulus for eliciting hypoxia-induced respiratory LTF.


Assuntos
Hipóxia/fisiopatologia , Inalação/fisiologia , Potenciação de Longa Duração/fisiologia , Plasticidade Neuronal/fisiologia , Fatores Etários , Animais , Dióxido de Carbono/farmacologia , Inalação/efeitos dos fármacos , Masculino , Neurônios Motores/fisiologia , Nervo Frênico/citologia , Nervo Frênico/fisiologia , Ratos , Ratos Sprague-Dawley , Índice de Gravidade de Doença
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...