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.
Am J Physiol Regul Integr Comp Physiol ; 308(8): R700-7, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25673781

RESUMO

In many neural networks, mechanisms of compensatory plasticity respond to prolonged reductions in neural activity by increasing cellular excitability or synaptic strength. In the respiratory control system, a prolonged reduction in synaptic inputs to the phrenic motor pool elicits a TNF-α- and atypical PKC-dependent form of spinal plasticity known as inactivity-induced phrenic motor facilitation (iPMF). Although iPMF may be elicited by a prolonged reduction in respiratory neural activity, iPMF is more efficiently induced when reduced respiratory neural activity (neural apnea) occurs intermittently. Mechanisms giving rise to iPMF following intermittent neural apnea are unknown. The purpose of this study was to test the hypothesis that iPMF following intermittent reductions in respiratory neural activity requires spinal TNF-α and aPKC. Phrenic motor output was recorded in anesthetized and ventilated rats exposed to brief intermittent (5, ∼1.25 min), brief sustained (∼6.25 min), or prolonged sustained (30 min) neural apnea. iPMF was elicited following brief intermittent and prolonged sustained neural apnea, but not following brief sustained neural apnea. Unlike iPMF following prolonged neural apnea, spinal TNF-α was not required to initiate iPMF during intermittent neural apnea; however, aPKC was still required for its stabilization. These results suggest that different patterns of respiratory neural activity induce iPMF through distinct cellular mechanisms but ultimately converge on a similar downstream pathway. Understanding the diverse cellular mechanisms that give rise to inactivity-induced respiratory plasticity may lead to development of novel therapeutic strategies to treat devastating respiratory control disorders when endogenous compensatory mechanisms fail.


Assuntos
Hipocapnia/enzimologia , Plasticidade Neuronal , Neurônios/enzimologia , Nervo Frênico/enzimologia , Proteína Quinase C/metabolismo , Centro Respiratório/enzimologia , Músculos Respiratórios/inervação , Transdução de Sinais , Nervos Espinhais/enzimologia , Fator de Necrose Tumoral alfa/metabolismo , Potenciais de Ação , Animais , Modelos Animais de Doenças , Hipercapnia/enzimologia , Hipercapnia/fisiopatologia , Hipocapnia/sangue , Hipocapnia/fisiopatologia , Masculino , Nervo Frênico/fisiopatologia , Ratos Sprague-Dawley , Centro Respiratório/fisiopatologia , Nervos Espinhais/fisiopatologia , Fatores de Tempo
2.
J Physiol ; 591(22): 5585-98, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23878370

RESUMO

A prolonged reduction in central neural respiratory activity elicits a form of plasticity known as inactivity-induced phrenic motor facilitation (iPMF), a 'rebound' increase in phrenic burst amplitude apparent once respiratory neural activity is restored. iPMF requires atypical protein kinase C (aPKC) activity within spinal segments containing the phrenic motor nucleus to stabilize an early transient increase in phrenic burst amplitude and to form long-lasting iPMF following reduced respiratory neural activity. Upstream signal(s) leading to spinal aPKC activation are unknown. We tested the hypothesis that spinal tumour necrosis factor-α (TNFα) is necessary for iPMF via an aPKC-dependent mechanism. Anaesthetized, ventilated rats were exposed to a 30 min neural apnoea; upon resumption of respiratory neural activity, a prolonged increase in phrenic burst amplitude (42 ± 9% baseline; P < 0.05) was apparent, indicating long-lasting iPMF. Pretreatment with recombinant human soluble TNF receptor 1 (sTNFR1) in the intrathecal space at the level of the phrenic motor nucleus prior to neural apnoea blocked long-lasting iPMF (2 ± 8% baseline; P > 0.05). Intrathecal TNFα without neural apnoea was sufficient to elicit long-lasting phrenic motor facilitation (pMF; 62 ± 7% baseline; P < 0.05). Similar to iPMF, TNFα-induced pMF required spinal aPKC activity, as intrathecal delivery of a ζ-pseudosubstrate inhibitory peptide (PKCζ-PS) 35 min following intrathecal TNFα arrested TNFα-induced pMF (28 ± 8% baseline; P < 0.05). These data demonstrate that: (1) spinal TNFα is necessary for iPMF; and (2) spinal TNFα is sufficient to elicit pMF via a similar aPKC-dependent mechanism. These data are consistent with the hypothesis that reduced respiratory neural activity elicits iPMF via a TNFα-dependent increase in spinal aPKC activity.


Assuntos
Neurônios Motores/fisiologia , Nervo Frênico/metabolismo , Nervo Frênico/fisiologia , Medula Espinal/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Apneia/metabolismo , Apneia/fisiopatologia , Humanos , Masculino , Neurônios Motores/metabolismo , Proteína Quinase C/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores do Fator de Necrose Tumoral/metabolismo , Medula Espinal/fisiologia
3.
J Neurosci ; 32(46): 16510-20, 2012 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-23152633

RESUMO

The neural network controlling breathing must establish rhythmic motor output at a level adequate to sustain life. Reduced respiratory neural activity elicits a novel form of plasticity in circuits driving the diaphragm known as inactivity-induced phrenic motor facilitation (iPMF), a rebound increase in phrenic inspiratory output observed once respiratory neural drive is restored. The mechanisms underlying iPMF are unknown. Here, we demonstrate in anesthetized rats that spinal mechanisms give rise to iPMF and that iPMF consists of at least two mechanistically distinct phases: (1) an early, labile phase that requires atypical PKC (PKCζ and/or PKCι/λ) activity to transition to a (2) late, stable phase. Early (but not late) iPMF is associated with increased interactions between PKCζ/ι and the scaffolding protein ZIP (PKCζ-interacting protein)/p62 in spinal regions associated with the phrenic motor pool. Although PKCζ/ι activity is necessary for iPMF, spinal atypical PKC activity is not necessary for phrenic long-term facilitation (pLTF) following acute intermittent hypoxia, an activity-independent form of spinal respiratory plasticity. Thus, while iPMF and pLTF both manifest as prolonged increases in phrenic burst amplitude, they arise from distinct spinal cellular pathways. Our data are consistent with the hypotheses that (1) local mechanisms sense and respond to reduced respiratory-related activity in the phrenic motor pool and (2) inactivity-induced increases in phrenic inspiratory output require local PKCζ/ι activity to stabilize into a long-lasting iPMF. Although the physiological role of iPMF is unknown, we suspect that iPMF represents a compensatory mechanism, assuring adequate motor output in a physiological system in which prolonged inactivity ends life.


Assuntos
Nervo Frênico/fisiologia , Proteína Quinase C/metabolismo , Medula Espinal/enzimologia , Animais , Western Blotting , Tronco Encefálico/fisiologia , Dióxido de Carbono/metabolismo , Fenômenos Eletrofisiológicos , Hipóxia/fisiopatologia , Imunoprecipitação , Isoenzimas/metabolismo , Masculino , Neurônios Motores/fisiologia , Oxigênio/sangue , Ratos , Ratos Sprague-Dawley , Mecânica Respiratória/fisiologia
4.
Vet Surg ; 41(4): 455-7, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22380877

RESUMO

OBJECTIVE: To determine if the volume of injected local anesthetic solution affects cranial to caudal spread when performing ultrasound-guided transversus abdominis plane (TAP) blocks in dogs. STUDY DESIGN: Prospective experimental study. ANIMALS: Adult Beagle cadavers (n = 20) METHODS: Bilateral TAP blocks using ultrasound guidance was performed in 20 Beagle cadavers (mean ± SD weight, 9.3 ± 1.4 kg) using a 1:1 solution of methylene blue/bupivacaine injected at volumes of 0.25, 0.5, 0.75, and 1.0 mL/kg. Cadavers were dissected to determine injectate spread within the transversus abdominis fascial plane. RESULTS: The transversus abdominis fascial plane was adequately identified by ultrasonography, injected, and dissected in 38 beagle hemi-abdominal walls; injectate was not identified in 2 hemi-abdominal walls. Dermatomal spread (number of ventral nerve roots saturated by injected solution) was volume dependent (P = .026, Kruskal Wallis): 2.9 ± 0.74 nerve roots for 0.25 mL/kg; 3.4 ± 1.1 for 0.5 mL/kg; 4.0 ± 0.67 for 0.75 mL/kg; and 4.2 ± 1.2 for 1 mL/kg. CONCLUSION: In Beagle cadavers, the volume of injected local anesthetic solution significantly affects cranial to caudal spread within the TAP during ultrasound-guided TAP blocks. The volume of local anesthetic injected could potentially be used to augment the spread of analgesic coverage for a given surgical procedure in dogs.


Assuntos
Parede Abdominal/inervação , Anestésicos Locais/administração & dosagem , Bupivacaína/administração & dosagem , Cães/metabolismo , Bloqueio Nervoso/veterinária , Anestesia Local/métodos , Anestesia Local/veterinária , Anestésicos Locais/farmacocinética , Animais , Bupivacaína/farmacocinética , Cadáver , Corantes , Azul de Metileno , Bloqueio Nervoso/métodos , Estudos Prospectivos
5.
Respir Physiol Neurobiol ; 179(1): 48-56, 2011 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-21745601

RESUMO

Intermittent hypoxia-induced long-term facilitation (LTF) is variably expressed in the motor output of several inspiratory nerves, such as the phrenic and hypoglossal. Compared to phrenic LTF (pLTF), less is known about hypoglossal LTF (hLTF), although it is often assumed that cellular mechanisms are the same. While fundamental mechanisms appear to be similar, potentially important differences exist in the modulation of pLTF and hLTF. The primary objectives of this paper are to: (1) review similarities and differences in pLTF and hLTF, pointing out knowledge gaps and (2) present new data suggesting that reduced respiratory neural activity elicits differential plasticity in phrenic and hypoglossal output (inactivity-induced phrenic and hypoglossal motor facilitation, iPMF and iHMF), suggesting that these motor pool-specific differences are not unique to LTF. Differences in fundamental mechanisms or modulation of plasticity among motor pools may confer the capacity to mount a complex ventilatory response to specific challenges, particularly in motor pools with different "jobs" in the control of breathing.


Assuntos
Nervo Hipoglosso/fisiologia , Neurônios Motores/fisiologia , Nervo Frênico/fisiologia , Mecânica Respiratória/fisiologia , Animais , Humanos
6.
Vet Anaesth Analg ; 38(3): 267-71, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21492393

RESUMO

OBJECTIVE: To describe the ultrasound-guided technique to the transversus abdominis plane (TAP) block in the dog and evaluate the spread of a local anesthetic/methylene blue solution. STUDY DESIGN: Prospective experimental trial. ANIMALS: Ten adult Beagle cadavers weighing 11.1 ± 1.1 kg (mean ± SD). METHODS: Transversus abdominis plane (TAP) blocks were performed bilaterally by a single trained individual on unpreserved cadaver dogs using 10 mL of methylene blue/bupivacaine solution per site. Dissection of the abdominal wall was performed within 15-55 minutes of block to determine distribution of injectate and nerve involvement in the transversus abdominis fascial plane. RESULTS: The transversus abdominis fascial plane was adequately visualized via ultrasound and injected in twenty hemi-abdominal walls. Segmental branches of T11, T12, T13, L1, L2, and L3 were adequately stained in 20%, 60%, 100%, 100%, 90%, and 30% of injections, respectively. CONCLUSIONS AND CLINICAL RELEVANCE: This anatomical study suggests that the transversus abdominis plane (TAP) block would provide adequate regional anesthesia of the abdomen, potentially extending to the cranial and caudal limits of the abdomen. This supports the clinical potential of this block in veterinary medicine.


Assuntos
Parede Abdominal/diagnóstico por imagem , Parede Abdominal/inervação , Anestésicos Locais/farmacocinética , Bupivacaína/farmacocinética , Cães/cirurgia , Bloqueio Nervoso/veterinária , Animais , Azul de Metileno/administração & dosagem , Azul de Metileno/farmacocinética , Bloqueio Nervoso/instrumentação , Bloqueio Nervoso/métodos , Estudos Prospectivos , Ultrassonografia
7.
Respir Physiol Neurobiol ; 175(3): 303-9, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21167322

RESUMO

We hypothesized that reduced respiratory neural activity elicits compensatory mechanisms of plasticity that enhance respiratory motor output. In urethane-anesthetized and ventilated rats, we reversibly reduced respiratory neural activity for 25-30 min using: hypocapnia (end tidal CO(2)=30 mmHg), isoflurane (~1%) or high frequency ventilation (HFV; ~100 breaths/min). In all cases, increased phrenic burst amplitude was observed following restoration of respiratory neural activity (hypocapnia: 92±22%; isoflurane: 65±22%; HFV: 54±13% baseline), which was significantly greater than time controls receiving the same surgery, but no interruptions in respiratory neural activity (3±5% baseline, p<0.05). Hypocapnia also elicited transient increases in respiratory burst frequency (9±2 versus 1±1bursts/min, p<0.05). Our results suggest that reduced respiratory neural activity elicits a unique form of plasticity in respiratory motor control which we refer to as inactivity-induced phrenic motor facilitation (iPMF). iPMF may prevent catastrophic decreases in respiratory motor output during ventilatory control disorders associated with abnormal respiratory activity.


Assuntos
Nervo Frênico/fisiologia , Fenômenos Fisiológicos Respiratórios , Animais , Hipocapnia/fisiopatologia , Masculino , Ratos , Ratos Sprague-Dawley
8.
Respir Physiol Neurobiol ; 162(1): 8-17, 2008 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-18450525

RESUMO

Acute intermittent (AIH), but not acute sustained hypoxia (ASH) elicits a form of respiratory plasticity known as long-term facilitation (LTF). In anesthetized rats, LTF is expressed as increased respiratory-related nerve burst amplitude, with variable effects on burst frequency. We analyzed a large data set from multiple investigators using the same experimental protocol to determine factors influencing frequency LTF. Our meta-analysis revealed that AIH elicits both phrenic amplitude and frequency LTF in anesthetized and vagotomized rats, but frequency LTF is small in comparison with amplitude LTF (12% versus 60%, respectively). ASH elicits a small, but significant frequency and amplitude LTF (8% and 10%, respectively) that is not significantly different than controls. Similar to all published reports, analysis of this large data set confirms that phrenic amplitude LTF following AIH is significantly greater than ASH. Multiple regression analysis revealed a strong correlation between baseline burst frequency and frequency LTF. Variations in baseline burst frequency may contribute to variation in frequency LTF and may underlie the apparent effects of some drug treatments.


Assuntos
Hipóxia/fisiopatologia , Potenciação de Longa Duração/fisiologia , Nervo Frênico/fisiopatologia , Animais , Hipóxia/patologia , Masculino , Ratos , Ratos Sprague-Dawley , Análise de Regressão , Fatores de Tempo , Vagotomia/métodos
9.
J Neurosci ; 28(11): 2949-58, 2008 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-18337426

RESUMO

Phrenic long-term facilitation (pLTF) is a serotonin-dependent form of pattern-sensitive respiratory plasticity induced by intermittent hypoxia (IH), but not sustained hypoxia (SH). The mechanism(s) underlying pLTF pattern sensitivity are unknown. SH and IH may differentially regulate serine/threonine protein phosphatase activity, thereby inhibiting relevant protein phosphatases uniquely during IH and conferring pattern sensitivity to pLTF. We hypothesized that spinal protein phosphatase inhibition would relieve this braking action of protein phosphatases, thereby revealing pLTF after SH. Anesthetized rats received intrathecal (C4) okadaic acid (25 nm) before SH (25 min, 11% O(2)). Unlike (vehicle) control rats, SH induced a significant pLTF in okadaic acid-treated rats that was indistinguishable from rats exposed to IH (three 5 min episodes, 11% O(2)). IH and SH with okadaic acid may elicit pLTF by similar, serotonin-dependent mechanisms, because intravenous methysergide blocks pLTF in rats receiving IH or okadaic acid plus SH. Okadaic acid did not alter IH-induced pLTF. In summary, pattern sensitivity in pLTF may reflect differential regulation of okadaic acid-sensitive serine/threonine phosphatases; presumably, these phosphatases are less active during/after IH versus SH. The specific okadaic acid-sensitive phosphatase(s) constraining pLTF and their spatiotemporal dynamics during and/or after IH and SH remain to be determined.


Assuntos
Hipóxia/enzimologia , Potenciação de Longa Duração/fisiologia , Ácido Okadáico/farmacologia , Fosfoproteínas Fosfatases/fisiologia , Nervo Frênico/enzimologia , Animais , Hipóxia/fisiopatologia , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Fosfoproteínas Fosfatases/análise , Nervo Frênico/química , Nervo Frênico/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
10.
J Neurosci ; 28(9): 2033-42, 2008 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-18305238

RESUMO

Acute intermittent hypoxia elicits a form of spinal, brain-derived neurotrophic factor (BDNF)-dependent respiratory plasticity known as phrenic long-term facilitation. Ligands that activate G(s)-protein-coupled receptors, such as the adenosine 2a receptor, mimic the effects of neurotrophins in vitro by transactivating their high-affinity receptor tyrosine kinases, the Trk receptors. Thus, we hypothesized that A2a receptor agonists would elicit phrenic long-term facilitation by mimicking the effects of BDNF on TrkB receptors. Here we demonstrate that spinal A2a receptor agonists transactivate TrkB receptors in the rat cervical spinal cord near phrenic motoneurons, thus inducing long-lasting (hours) phrenic motor facilitation. A2a receptor activation increased phosphorylation and new synthesis of an immature TrkB protein, induced TrkB signaling through Akt, and strengthened synaptic pathways to phrenic motoneurons. RNA interference targeting TrkB mRNA demonstrated that new TrkB protein synthesis is necessary for A2a-induced phrenic motor facilitation. A2a receptor activation also increased breathing in unanesthetized rats, and improved breathing in rats with cervical spinal injuries. Thus, small, highly permeable drugs (such as adenosine receptor agonists) that transactivate TrkB receptors may provide an effective therapeutic strategy in the treatment of patients with ventilatory control disorders, such as obstructive sleep apnea, or respiratory insufficiency after spinal injury or during neurodegenerative diseases.


Assuntos
Neurônios Motores/fisiologia , Nervo Frênico/fisiologia , Receptor A2A de Adenosina/metabolismo , Medula Espinal/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Adenosina/análogos & derivados , Adenosina/farmacologia , Antagonistas do Receptor A2 de Adenosina , Animais , Anti-Hipertensivos/farmacologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Interações Medicamentosas , Ensaio de Imunoadsorção Enzimática/métodos , Masculino , Neurônios Motores/efeitos dos fármacos , Fenetilaminas/farmacologia , Nervo Frênico/efeitos dos fármacos , Pletismografia/métodos , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor trkB/genética , Receptor trkB/metabolismo , Medula Espinal/efeitos dos fármacos , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/fisiopatologia , Xantinas/farmacologia
11.
J Neurotrauma ; 22(2): 203-13, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15716627

RESUMO

Following chronic C2 spinal hemisection (C2HS), crossed spinal pathways to phrenic motoneurons exhibit a slow, spontaneous increase in efficacy by a serotonin (5-HT)-dependent mechanism associated with 5-HT2A receptor activation. Further, the spontaneous appearance of cross-phrenic activity following C2HS is accelerated and enhanced by exposure to chronic intermittent hypoxia (CIH). We hypothesized that chronic C2HS would increase 5-HT and 5-HT2A receptor expression in ventral cervical spinal segments containing phrenic motoneurons. In addition, we hypothesized that CIH exposure would further increase 5-HT and 5-HT2A receptor density in this region. Control, sham-operated, and C2HS Sprague-Dawley rats were studied following normoxia or CIH (11% O2-air; 5-min intervals; nights 7-14 post-surgery). At 2 weeks post-surgery, ventral spinal gray matter extending from C4 and C5 was isolated ipsilateral and contralateral to C2HS. Neither C2HS nor CIH altered 5-HT concentration measured with an ELISA on either side of the spinal cord. However, 5-HT2A receptor expression assessed with immunoblots increased in ipsilateral gray matter following C2HS, an effect independent of CIH. Immunocytochemistry revealed increased 5-HT2A receptor expression on identified phrenic motoneurons (p<0.05), as well as in the surrounding gray matter. Contralateral to injury, 5-HT2A receptor expression was elevated in CIH, but not normoxic C2HS rats (p<0.05). Our data are consistent with the hypothesis that spontaneous increase in 5-HT2A receptor expression on or near phrenic motoneurons contributes to strengthened crossed-spinal synaptic pathways to phrenic motoneurons following C2HS.


Assuntos
Hipóxia/metabolismo , Receptor 5-HT2A de Serotonina/metabolismo , Serotonina/metabolismo , Traumatismos da Medula Espinal/metabolismo , Medula Espinal/metabolismo , Animais , Vértebras Cervicais , Vias Eferentes/fisiologia , Hipóxia/complicações , Masculino , Neurônios Motores/metabolismo , Nervo Frênico/metabolismo , Ratos , Ratos Sprague-Dawley , Traumatismos da Medula Espinal/complicações , Regulação para Cima/fisiologia
12.
Nat Neurosci ; 7(1): 48-55, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14699417

RESUMO

Intermittent hypoxia causes a form of serotonin-dependent synaptic plasticity in the spinal cord known as phrenic long-term facilitation (pLTF). Here we show that increased synthesis of brain-derived neurotrophic factor (BDNF) in the spinal cord is necessary and sufficient for pLTF in adult rats. We found that intermittent hypoxia elicited serotonin-dependent increases in BDNF synthesis in ventral spinal segments containing the phrenic nucleus, and the magnitude of these BDNF increases correlated with pLTF magnitude. We used RNA interference (RNAi) to interfere with BDNF expression, and tyrosine kinase receptor inhibition to block BDNF signaling. These disruptions blocked pLTF, whereas intrathecal injection of BDNF elicited an effect similar to pLTF. Our findings demonstrate new roles and regulatory mechanisms for BDNF in the spinal cord and suggest new therapeutic strategies for treating breathing disorders such as respiratory insufficiency after spinal injury. These experiments also illustrate the potential use of RNAi to investigate functional consequences of gene expression in the mammalian nervous system in vivo.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/biossíntese , Hipóxia/metabolismo , Plasticidade Neuronal/fisiologia , Respiração , Medula Espinal/metabolismo , Animais , Masculino , Nervo Frênico/metabolismo , Ratos , Ratos Sprague-Dawley
13.
J Neurosci ; 22(14): 6239-46, 2002 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-12122082

RESUMO

Respiratory long-term facilitation (LTF) is a form of serotonin-dependent plasticity induced by intermittent hypoxia. LTF is manifested as a long-lasting increase in respiratory amplitude (and frequency) after the hypoxic episodes have ended. We tested the hypotheses that LTF of phrenic amplitude requires spinal serotonin receptor activation and spinal protein synthesis. A broad-spectrum serotonin receptor antagonist (methysergide) or protein synthesis inhibitors (emetine or cycloheximide) were injected intrathecally in the cervical spinal cord of anesthetized rats. Control rats, injected with vehicle (artificial CSF), exhibited an augmented phrenic burst amplitude after three 5 min episodes of hypoxia (78 +/- 15% above baseline, 60 min after hypoxia; p < 0.05), indicating LTF. Pretreatment with methysergide, emetine, or cycloheximide attenuated or abolished phrenic LTF (20 +/- 4, 0.2 +/- 11, and 20 +/- 2%, respectively; all p > 0.05). With protein synthesis inhibitors, phrenic LTF differed from control by 15 min after intermittent hypoxia. As an internal control against unintended drug distribution, we measured respiratory LTF in hypoglossal (XII) motor output. At 60 min after intermittent hypoxia, all treatment groups exhibited similar XII LTF (artificial CSF, 44 +/- 10%; methysergide, 40 +/- 5%; emetine, 35 +/- 9%; and cycloheximide, 57 +/- 29%; all p < 0.05), suggesting that drugs were restricted at effective doses to the spinal cord. We conclude that phrenic LTF requires spinal serotonin receptor activation and protein synthesis. Serotonin receptors on phrenic motoneuron dendrites may induce new protein synthesis, thereby giving rise to phrenic LTF.


Assuntos
Potenciação de Longa Duração/fisiologia , Nervo Frênico/fisiologia , Biossíntese de Proteínas , Receptores de Serotonina/metabolismo , Medula Espinal/metabolismo , Animais , Cicloeximida/farmacologia , Emetina/farmacologia , Nervo Hipoglosso/fisiologia , Injeções Espinhais , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Metisergida/farmacologia , Neurônios Motores/fisiologia , Pescoço , Plasticidade Neuronal/fisiologia , Inibidores da Síntese de Proteínas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de Serotonina/efeitos dos fármacos , Respiração , Antagonistas da Serotonina/farmacologia , Medula Espinal/efeitos dos fármacos , Vagotomia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...