Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 165
Filtrar
1.
Am J Physiol Regul Integr Comp Physiol ; 314(1): R135-R144, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29021191

RESUMEN

Moderate acute intermittent hypoxia (mAIH) elicits a form of respiratory motor plasticity known as phrenic long-term facilitation (pLTF). Preconditioning with modest protocols of chronic intermittent hypoxia enhances pLTF, demonstrating pLTF metaplasticity. Since "low-dose" protocols of repetitive acute intermittent hypoxia (rAIH) show promise as a therapeutic modality to restore respiratory (and nonrespiratory) motor function in clinical disorders with compromised breathing, we tested 1) whether preconditioning with a mild rAIH protocol enhances pLTF and hypoglossal (XII) LTF and 2) whether the enhancement is regulated by glycolytic flux. In anesthetized, paralyzed, and ventilated adult male Lewis rats, mAIH (three 5-min episodes of 10% O2) elicited pLTF (pLTF at 60 min post-mAIH: 49 ± 5% baseline). rAIH preconditioning (ten 5-min episodes of 11% O2/day with 5-min normoxic intervals, 3 times per week, for 4 wk) significantly enhanced pLTF (100 ± 16% baseline). XII LTF was unaffected by rAIH. When glycolytic flux was inhibited by 2-deoxy-d-glucose (2-DG) administered via drinking water (~80 mg·kg-1·day-1), pLTF returned to normal levels (58 ± 8% baseline); 2-DG had no effect on pLTF in normoxia-pretreated rats (59 ± 7% baseline). In ventral cervical (C4/5) spinal homogenates, rAIH increased inducible nitric oxide synthase mRNA vs. normoxic controls, an effect blocked by 2-DG. However, there were no detectable effects of rAIH or 2-DG on several molecules associated with phrenic motor plasticity, including serotonin 2A, serotonin 7, brain-derived neurotrophic factor, tropomyosin receptor kinase B, or VEGF mRNA. We conclude that modest, but prolonged, rAIH elicits pLTF metaplasticity and that a drug known to inhibit glycolytic flux (2-DG) blocks pLTF enhancement.


Asunto(s)
Antimetabolitos/farmacología , Desoxiglucosa/farmacología , Glucólisis/efectos de los fármacos , Hipoxia/fisiopatología , Potenciación a Largo Plazo/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Nervio Frénico/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Regulación Enzimológica de la Expresión Génica , Hipoxia/metabolismo , Masculino , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Nervio Frénico/fisiopatología , Ratas Endogámicas Lew , Factores de Tiempo
2.
Respir Physiol Neurobiol ; 256: 50-57, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-28549897

RESUMEN

Daily acute intermittent hypoxia (dAIH) elicits respiratory plasticity, enhancing respiratory motor output and restoring breathing capacity after incomplete cervical spinal injuries (cSCI). We hypothesized that dAIH-induced functional recovery of breathing capacity would occur after both acute (2 weeks) and chronic (8 weeks) cSCI, but through distinct cellular mechanisms. Specifically, we hypothesized that dAIH-induced breathing recovery would occur through serotonin-independent mechanisms 2wks post C2 cervical hemisection (C2Hs), versus serotonin-dependent mechanisms 8wks post C2Hs. In two independent studies, dAIH or sham (normoxia) was initiated 1 week (Study 1) or 7 weeks (Study 2) post-C2Hs to test our hypothesis. Rats were pre-treated with intra-peritoneal vehicle or methysergide, a broad-spectrum serotonin receptor antagonist, to determine the role of serotonin signaling in dAIH-induced functional recovery. Our data support the hypothesis that dAIH-induced recovery of breathing capacity transitions from a serotonin-independent mechanism with acute C2Hs to a serotonin-dependent mechanism with chronic C2Hs. An understanding of shifting mechanisms giving rise to dAIH-induced respiratory motor plasticity is vital for clinical translation of dAIH as a therapeutic modality.


Asunto(s)
Hipoxia , Recuperación de la Función/fisiología , Trastornos Respiratorios/etiología , Trastornos Respiratorios/terapia , Traumatismos de la Médula Espinal/complicaciones , Animales , Modelos Animales de Enfermedad , Hipoxia/fisiopatología , Masculino , Metisergida/farmacología , Pletismografía , Ratas , Ratas Endogámicas Lew , Ratas Sprague-Dawley , Recuperación de la Función/efectos de los fármacos , Antagonistas de la Serotonina/farmacología , Factores de Tiempo , Vagotomía
3.
Neuropharmacology ; 113(Pt A): 82-88, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27663700

RESUMEN

Spinal metabotropic serotonin receptors encode transient experiences into long-lasting changes in motor behavior (i.e. motor plasticity). While interactions between serotonin receptor subtypes are known to regulate plasticity, the significance of molecular divergence in downstream G protein coupled receptor signaling is not well understood. Here we tested the hypothesis that distinct cAMP dependent signaling pathways differentially regulate serotonin-induced phrenic motor facilitation (pMF); a well-studied model of spinal motor plasticity. Specifically, we studied the capacity of cAMP-dependent protein kinase A (PKA) and exchange protein activated by cAMP (EPAC) to regulate 5-HT2A receptor-induced pMF within adult male rats. Although spinal PKA, EPAC and 5-HT2A each elicit pMF when activated alone, concurrent PKA and 5-HT2A activation interact via mutual inhibition thereby blocking pMF expression. Conversely, concurrent EPAC and 5-HT2A activation enhance pMF expression reflecting additive contributions from both mechanisms. Thus, we demonstrate that distinct downstream cAMP signaling pathways enable differential regulation of 5-HT2A-induced pMF. Conditional activation of independent signaling mechanisms may explain experience amendable changes in plasticity expression (i.e. metaplasticity), an emerging concept thought to enable flexible motor control within the adult central nervous system.


Asunto(s)
AMP Cíclico/metabolismo , Neuronas Motoras/metabolismo , Plasticidad Neuronal , Nervio Frénico/metabolismo , Receptor de Serotonina 5-HT2A/metabolismo , Serotonina/metabolismo , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Factores de Intercambio de Guanina Nucleótido/antagonistas & inhibidores , Factores de Intercambio de Guanina Nucleótido/metabolismo , Masculino , Neuronas Motoras/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Nervio Frénico/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Agonistas del Receptor de Serotonina 5-HT2/administración & dosificación , Transducción de Señal
4.
Exp Neurol ; 287(Pt 2): 93-101, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27079999

RESUMEN

Daily acute intermittent hypoxia (dAIH) improves breathing capacity after C2 spinal hemisection (C2HS) in rats. Since C2HS disrupts spinal serotonergic innervation below the injury, adenosine-dependent mechanisms underlie dAIH-induced functional recovery 2weeks post-injury. We hypothesized that dAIH-induced functional recovery converts from an adenosine-dependent to a serotonin-dependent, adenosine-constrained mechanism with chronic injury. Eight weeks post-C2HS, rats began dAIH (10, 5-min episodes, 10.5% O2; 5-min intervals; 7days) followed by AIH 3× per week (3×wAIH) for 8 additional weeks with/without systemic A2A receptor inhibition (KW6002) on each AIH exposure day. Tidal volume (VT) and bilateral diaphragm (Dia) and T2 external intercostal motor activity were assessed in unanesthetized rats breathing air and during maximum chemoreflex stimulation (MCS: 7% CO2, 10.5% O2). Nine weeks post-C2HS, dAIH increased VT versus time controls (p<0.05), an effect enhanced by KW6002 (p<0.05). dAIH increased bilateral Dia activity (p<0.05), and KW6002 enhanced this effect in contralateral (p<0.05) and ipsilateral Dia activity (p<0.001), but not T2 inspiratory activity. Functional benefits of combined AIH plus systemic A2A receptor inhibition were maintained for 4weeks. Thus, in rats with chronic injuries: 1) dAIH improves VT and bilateral diaphragm activity; 2) VT recovery is enhanced by A2A receptor inhibition; and 3) functional recovery with A2A receptor inhibition and AIH "reminders" last 4weeks. Combined dAIH and A2A receptor inhibition may be a simple, safe, and effective strategy to accelerate/enhance functional recovery of breathing capacity in patients with respiratory impairment from chronic spinal injury.


Asunto(s)
Vértebras Cervicales , Hipoxia , Ventilación Voluntaria Máxima/fisiología , Receptores de Adenosina A2/metabolismo , Recuperación de la Función/fisiología , Trastornos Respiratorios/etiología , Trastornos Respiratorios/terapia , Traumatismos de la Médula Espinal/complicaciones , Antagonistas del Receptor de Adenosina A2/farmacología , Antagonistas del Receptor de Adenosina A2/uso terapéutico , Animales , Diafragma/efectos de los fármacos , Modelos Animales de Enfermedad , Lateralidad Funcional/efectos de los fármacos , Lateralidad Funcional/fisiología , Hipercapnia/fisiopatología , Masculino , Ventilación Voluntaria Máxima/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Contracción Muscular/efectos de los fármacos , Purinas/farmacología , Purinas/uso terapéutico , Ratas , Ratas Sprague-Dawley , Recuperación de la Función/efectos de los fármacos , Factores de Tiempo , Capacidad Vital/efectos de los fármacos , Capacidad Vital/fisiología
5.
Neuroscience ; 322: 479-88, 2016 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-26944605

RESUMEN

Repetitive acute intermittent hypoxia (rAIH) increases growth/trophic factor expression in respiratory motor neurons, thereby eliciting spinal respiratory motor plasticity and/or neuroprotection. Here we demonstrate that rAIH effects are not unique to respiratory motor neurons, but are also expressed in non-respiratory, spinal alpha motor neurons and upper motor neurons of the motor cortex. In specific, we used immunohistochemistry and immunofluorescence to assess growth/trophic factor protein expression in spinal sections from rats exposed to AIH three times per week for 10weeks (3×wAIH). 3×wAIH increased brain-derived neurotrophic factor (BDNF), its high-affinity receptor, tropomyosin receptor kinase B (TrkB), and phosphorylated TrkB (pTrkB) immunoreactivity in putative alpha motor neurons of spinal cervical 7 (C7) and lumbar 3 (L3) segments, as well as in upper motor neurons of the primary motor cortex (M1). 3×wAIH also increased immunoreactivity of vascular endothelial growth factor A (VEGFA), the high-affinity VEGFA receptor (VEGFR-2) and an important VEGF gene regulator, hypoxia-inducible factor-1α (HIF-1α). Thus, rAIH effects on growth/trophic factors are characteristic of non-respiratory as well as respiratory motor neurons. rAIH may be a useful tool in the treatment of disorders causing paralysis, such as spinal injury and motor neuron disease, as a pretreatment to enhance motor neuron survival during disease, or as preconditioning for cell-transplant therapies.


Asunto(s)
Hipoxia/metabolismo , Neuronas Motoras/metabolismo , Médula Espinal/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Vértebras Cervicales , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inmunohistoquímica , Vértebras Lumbares , Masculino , Corteza Motora/metabolismo , Corteza Motora/patología , Neuronas Motoras/patología , Fosforilación , Distribución Aleatoria , Ratas Sprague-Dawley , Receptor trkB/metabolismo , Médula Espinal/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo
6.
J Neurophysiol ; 114(3): 2015-22, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26269554

RESUMEN

Spinal serotonin type 7 (5-HT7) receptors elicit complex effects on motor activity. Whereas 5-HT7 receptor activation gives rise to long-lasting phrenic motor facilitation (pMF), it also constrains 5-HT2 receptor-induced pMF via "cross-talk inhibition." We hypothesized that divergent cAMP-dependent signaling pathways give rise to these distinct 5-HT7 receptor actions. Specifically, we hypothesized that protein kinase A (PKA) mediates cross-talk inhibition of 5-HT2 receptor-induced pMF whereas 5-HT7 receptor-induced pMF results from exchange protein activated by cAMP (EPAC) signaling. Anesthetized, paralyzed, and ventilated rats receiving intrathecal (C4) 5-HT7 receptor agonist (AS-19) injections expressed pMF for >90 min, an effect abolished by pretreatment with a selective EPAC inhibitor (ESI-05) but not a selective PKA inhibitor (KT-5720). Furthermore, intrathecal injections of a selective EPAC activator (8-pCPT-2'-Me-cAMP) were sufficient to elicit pMF. Finally, spinal mammalian target of rapamycin complex-1 (mTORC1) inhibition via intrathecal rapamycin abolished 5-HT7 receptor- and EPAC-induced pMF, demonstrating that spinal 5-HT7 receptors elicit pMF by an EPAC-mTORC1 signaling pathway. Thus 5-HT7 receptors elicit and constrain spinal phrenic motor plasticity via distinct signaling mechanisms that diverge at cAMP (EPAC vs. PKA). Selective manipulation of these molecules may enable refined regulation of serotonin-dependent spinal motor plasticity for therapeutic advantage.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/metabolismo , Complejos Multiproteicos/metabolismo , Nervio Frénico/metabolismo , Receptores de Serotonina/metabolismo , Médula Espinal/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Neuronas Motoras/metabolismo , Neuronas Motoras/fisiología , Plasticidad Neuronal , Nervio Frénico/fisiología , Ratas , Ratas Endogámicas Lew , Transducción de Señal , Médula Espinal/fisiología
7.
Exp Neurol ; 266: 1-10, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25687551

RESUMEN

A major cause of mortality after spinal cord injury is respiratory failure. In normal rats, acute intermittent hypoxia (AIH) induces respiratory motor plasticity, expressed as diaphragm (Dia) and second external intercostal (T2 EIC) long-term facilitation (LTF). Dia (not T2 EIC) LTF is enhanced by systemic adenosine 2A (A2A) receptor inhibition in normal rats. We investigated the respective contributions of Dia and T2 EIC to daily AIH-induced functional recovery of breathing capacity with/without A2A receptor antagonist (KW6002, i.p.) following C2 hemisection (C2HS). Rats received daily AIH (dAIH: 10, 5-min episodes, 10.5% O2; 5-min normoxic intervals; 7 successive days beginning 7days post-C2HS) or daily normoxia (dNx) with/without KW6002, followed by weekly (reminder) presentations for 8weeks. Ventilation and EMGs from bilateral diaphragm and T2 EIC muscles were measured with room air breathing (21% O2) and maximum chemoreceptor stimulation ( MCS: 7% CO2, 10.5% O2). dAIH increased tidal volume (VT) in C2HS rats breathing room air (dAIH+vehicle: 0.47±0.02, dNx+vehicle: 0.40±0.01ml/100g; p<0.05) and MCS (dAIH+vehicle: 0.83±0.01, dNx+vehicle: 0.73±0.01ml/100g; p<0.001); KW6002 had no significant effect. dAIH enhanced contralateral (uninjured) diaphragm EMG activity, an effect attenuated by KW6002, during room air breathing and MCS (p<0.05). Although dAIH enhanced contralateral T2 EIC EMG activity during room air breathing, KW6002 had no effect. dAIH had no statistically significant effects on diaphragm or T2 EIC EMG activity ipsilateral to injury. Thus, two weeks post-C2HS: 1) dAIH enhances breathing capacity by effects on contralateral diaphragm and T2 EIC activity; and 2) dAIH-induced recovery is A2A dependent in diaphragm, but not T2 EIC. Daily AIH may be a useful in promoting functional recovery of breathing capacity after cervical spinal injury, but A2A receptor antagonists (e.g. caffeine) may undermine its effectiveness shortly after injury.


Asunto(s)
Vértebras Cervicales , Diafragma/fisiopatología , Hipoxia , Músculos Intercostales/fisiopatología , Traumatismos de la Médula Espinal/fisiopatología , Traumatismos de la Médula Espinal/terapia , Antagonistas del Receptor de Adenosina A2/uso terapéutico , Animales , Aseo Animal , Masculino , Desempeño Psicomotor , Ratas , Ratas Sprague-Dawley , Receptor de Adenosina A2A/efectos de los fármacos , Recuperación de la Función , Mecánica Respiratoria/fisiología , Traumatismos de la Médula Espinal/psicología , Volumen de Ventilación Pulmonar
8.
J Appl Physiol (1985) ; 117(2): 180-8, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24833779

RESUMEN

Although rats are a frequent model for studies of plasticity in respiratory motor control, the relative capacity of rat accessory respiratory muscles to express plasticity is not well known, particularly in unanesthetized animals. Here, we characterized external intercostal (T2, T4, T5, T6, T7, T8, T9 EIC) and abdominal muscle (external oblique and rectus abdominis) electromyogram (EMG) activity in unanesthetized rats via radiotelemetry during normoxia (Nx: 21% O2) and following acute intermittent hypoxia (AIH: 10 × 5-min, 10.5% O2; 5-min intervals). Diaphragm and T2-T5 EIC EMG activity, and ventilation were also assessed during maximal chemoreceptor stimulation ( MCS: 7% CO2, 10.5% O2) and sustained hypoxia (SH: 10.5% O2). In Nx, T2 EIC exhibits prominent inspiratory activity, whereas T4, T5, T6, and T7 EIC inspiratory activity decreases in a caudal direction. T8 and T9 EIC and abdominal muscles show only tonic or sporadic activity, without consistent respiratory activity. MCS increases diaphragm and T2 EIC EMG amplitude and tidal volume more than SH (0.94 ± 0.10 vs. 0.68 ± 0.05 ml/100 g; P < 0.001). Following AIH, T2 EIC EMG amplitude remained above baseline for more than 60 min post-AIH (i.e., EIC long-term facilitation, LTF), and was greater than diaphragm LTF (41.5 ± 1.3% vs. 19.1 ± 2.0% baseline; P < 0.001). We conclude that 1) diaphragm and rostral T2-T5 EIC muscles exhibit inspiratory activity during Nx; 2) MCS elicits greater ventilatory, diaphragm, and rostral T2-T5 EIC muscle activity vs. SH; and 3) AIH induces greater rostral EIC LTF than diaphragm LTF.


Asunto(s)
Músculos Abdominales/fisiología , Diafragma/fisiología , Músculos Intercostales/fisiología , Músculos Respiratorios/fisiología , Animales , Células Quimiorreceptoras/fisiología , Electromiografía/métodos , Hipoxia/fisiopatología , Masculino , Nervio Frénico/fisiología , Ratas , Ratas Sprague-Dawley , Respiración , Mecánica Respiratoria/fisiología , Volumen de Ventilación Pulmonar/fisiología
9.
Respir Physiol Neurobiol ; 197: 19-28, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24681328

RESUMEN

In rats over-expressing SOD1G93A, ventilation is preserved despite significant loss of respiratory motor neurons. Thus, unknown forms of compensatory respiratory plasticity may offset respiratory motor neuron cell death. Although mechanisms of such compensation are unknown, other models of respiratory motor plasticity may provide a conceptual guide. Multiple cellular mechanisms give rise to phrenic motor facilitation; one mechanism requires spinal serotonin receptor and NADPH oxidase activity whereas another requires spinal adenosine receptor activation. Here, we studied whether these mechanisms contribute to compensatory respiratory plasticity in SOD1G93A rats. Using plethysmography, we assessed ventilation in end-stage SOD1G93A rats after: (1) serotonin depletion with parachlorophenylalanine (PCPA), (2) serotonin (methysergide) and A2A (MSX-3) receptor inhibition, (3) NADPH oxidase inhibition (apocynin), and (4) combined treatments. The ability to increase ventilation was not decreased by individual or combined treatments; thus, these mechanisms do not maintain breathing capacity at end-stage motor neuron disease. Possible mechanisms giving rise to enhanced breathing capacity with combined treatment in end-stage SOD1G93A rats are discussed.


Asunto(s)
Adenosina/fisiología , Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Esclerosis Amiotrófica Lateral/fisiopatología , Ventilación Pulmonar/efectos de los fármacos , Ventilación Pulmonar/fisiología , Respiración , Serotonina/fisiología , Acetofenonas/farmacología , Antagonistas del Receptor de Adenosina A2/farmacología , Esclerosis Amiotrófica Lateral/patología , Animales , Inhibidores Enzimáticos/farmacología , Humanos , Masculino , Metisergida/farmacología , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/patología , Neuronas Motoras/fisiología , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/metabolismo , Nervio Frénico/efectos de los fármacos , Nervio Frénico/patología , Nervio Frénico/fisiopatología , Pletismografía , Ratas Sprague-Dawley , Ratas Transgénicas , Respiración/efectos de los fármacos , Antagonistas de la Serotonina/farmacología , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1 , Volumen de Ventilación Pulmonar/efectos de los fármacos , Volumen de Ventilación Pulmonar/fisiología , Xantinas/farmacología
10.
Neuroscience ; 269: 67-78, 2014 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-24680940

RESUMEN

Acute intermittent hypoxia (AIH) induces phrenic long-term facilitation (pLTF) by a mechanism that requires spinal serotonin (5-HT) receptor activation and NADPH oxidase (NOX) activity. Here, we investigated whether: (1) spinal nitric oxide synthase (NOS) activity is necessary for AIH-induced pLTF; (2) episodic exogenous nitric oxide (NO) is sufficient to elicit phrenic motor facilitation (pMF) without AIH (i.e. pharmacologically); and (3) NO-induced pMF requires spinal 5-HT2B receptor and NOX activation. In anesthetized, mechanically ventilated adult male rats, AIH (3 × 5-min episodes; 10% O2; 5 min) elicited a progressive increase in the amplitude of integrated phrenic nerve bursts (i.e. pLTF), which lasted 60 min post-AIH (45.1 ± 8.6% baseline). Pre-treatment with intrathecal (i.t.) injections of a neuronal NOS inhibitor (nNOS-inhibitor-1) near the phrenic motor nucleus attenuated pLTF (14.7 ± 2.5%), whereas an inducible NOS (iNOS) inhibitor (1400 W) had no effect (56.3 ± 8.0%). Episodic i.t. injections (3 × 5µl volume; 5 min) of a NO donor (sodium nitroprusside; SNP) elicited pMF similar in time-course and magnitude (40.4 ± 6.0%, 60 min post-injection) to AIH-induced pLTF. SNP-induced pMF was blocked by a 5-HT2B receptor antagonist (SB206553), a superoxide dismutase mimetic (MnTMPyP), and two NOX inhibitors (apocynin and DPI). Neither pLTF nor pMF was affected by pre-treatment with a protein kinase G (PKG) inhibitor (KT-5823). Thus, spinal nNOS activity is necessary for AIH-induced pLTF, and episodic spinal NO is sufficient to elicit pMF by a mechanism that requires 5-HT2B receptor activation and NOX-derived ROS formation, which indicates AIH (and NO) elicits spinal respiratory plasticity by a nitrergic-serotonergic mechanism.


Asunto(s)
NADPH Oxidasas/metabolismo , Plasticidad Neuronal/fisiología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Nervio Frénico/fisiología , Receptor de Serotonina 5-HT2B/metabolismo , Médula Espinal/fisiología , Enfermedad Aguda , Animales , Proteínas Quinasas Dependientes de GMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Inhibidores Enzimáticos/farmacología , Hipoxia/fisiopatología , Masculino , Plasticidad Neuronal/efectos de los fármacos , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa de Tipo I/antagonistas & inhibidores , Nervio Frénico/efectos de los fármacos , Ratas , Especies Reactivas de Oxígeno/metabolismo , Antagonistas del Receptor de Serotonina 5-HT2/farmacología , Médula Espinal/efectos de los fármacos , Superóxido Dismutasa/metabolismo
11.
J Appl Physiol (1985) ; 116(11): 1345-52, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24526581

RESUMEN

Acute intermittent hypoxia (AIH; three 5-min hypoxic episodes) causes a form of phrenic motor facilitation (pMF) known as phrenic long-term facilitation (pLTF); pLTF is initiated by spinal activation of Gq protein-coupled 5-HT2 receptors. Because α1 adrenergic receptors are expressed in the phrenic motor nucleus and are also Gq protein-coupled, we hypothesized that α1 receptors are sufficient, but not necessary for AIH-induced pLTF. In anesthetized, paralyzed, and ventilated rats, episodic spinal application of the α1 receptor agonist phenylephrine (PE) elicited dose-dependent pMF (10 and 100 µM, P < 0.05; but not 1 µM). PE-induced pMF was blocked by the α1 receptor antagonist prazosin (1 mM; -20 ± 20% at 60 min, -5 ± 21% at 90 min; n = 6). Although α1 receptor activation is sufficient to induce pMF, it was not necessary for AIH-induced pLTF because intrathecal prazosin (1 mM) did not alter AIH-induced pLTF (56 ± 9% at 60 min, 78 ± 12% at 90 min; n = 9). Intravenous (iv) prazosin (150 µg/kg) appeared to reduce pLTF (21 ± 9% at 60 min, 26 ± 8% at 90 min), but this effect was not significant. Hypoglossal long-term facilitation was unaffected by intrathecal prazosin, but was blocked by iv prazosin (-4 ± 14% at 60 min, -13 ± 18% at 90 min), suggesting different LTF mechanisms in different motor neuron pools. In conclusion, Gq protein-coupled α1 adrenergic receptors evoke pMF, but they are not necessary for AIH-induced pLTF.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 1/administración & dosificación , Antagonistas de Receptores Adrenérgicos alfa 1/administración & dosificación , Hipoxia/metabolismo , Potenciación a Largo Plazo/efectos de los fármacos , Neuronas Motoras/efectos de los fármacos , Nervio Frénico/fisiopatología , Receptores Adrenérgicos alfa 1/metabolismo , Animales , Médula Cervical/efectos de los fármacos , Médula Cervical/fisiopatología , Relación Dosis-Respuesta a Droga , Masculino , Plasticidad Neuronal/efectos de los fármacos , Nervio Frénico/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
12.
Physiology (Bethesda) ; 29(1): 39-48, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24382870

RESUMEN

Intermittent hypoxia (IH) is most often thought of for its role in morbidity associated with sleep-disordered breathing, including central nervous system pathology. However, recent evidence suggests that the nervous system fights back in an attempt to minimize pathology by increasing the expression of growth/trophic factors that confer neuroprotection and neuroplasticity. For example, even modest ("low dose") IH elicits respiratory motor plasticity, increasing the strength of respiratory contractions and breathing. These low IH doses upregulate hypoxia-sensitive growth/trophic factors within respiratory motoneurons but do not elicit detectable pathologies such as hippocampal cell death, neuroinflammation, or systemic hypertension. Recent advances have been made toward understanding cellular mechanisms giving rise to IH-induced respiratory plasticity, and attempts have been made to harness the benefits of low-dose IH to treat respiratory insufficiency after cervical spinal injury. Our recent realization that IH also upregulates growth/trophic factors in nonrespiratory motoneurons and improves limb (or leg) function after incomplete chronic spinal injuries suggests that IH-induced plasticity is a general feature of motor systems. Collectively, available evidence suggests that low-dose IH may represent a safe and effective treatment to restore lost motor function in diverse clinical disorders that impair motor function.


Asunto(s)
Hipocampo/fisiopatología , Hipoxia/metabolismo , Neuronas Motoras/fisiología , Plasticidad Neuronal/fisiología , Fenómenos Fisiológicos Respiratorios , Animales , Hipocampo/citología , Humanos , Hipoxia/fisiopatología , Síndromes de la Apnea del Sueño/fisiopatología
13.
Neuroscience ; 250: 632-43, 2013 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-23850591

RESUMEN

Phrenic long-term facilitation (pLTF) is a form of serotonin-dependent respiratory plasticity induced by acute intermittent hypoxia (AIH). pLTF requires spinal Gq protein-coupled serotonin-2 receptor (5-HT2) activation, new synthesis of brain-derived neurotrophic factor (BDNF) and activation of its high-affinity receptor, TrkB. Intrathecal injections of selective agonists for Gs protein-coupled receptors (adenosine 2A and serotonin-7; 5-HT7) also induce long-lasting phrenic motor facilitation via TrkB "trans-activation." Since serotonin released near phrenic motor neurons may activate multiple serotonin receptor subtypes, we tested the hypothesis that 5-HT7 receptor activation contributes to AIH-induced pLTF. A selective 5-HT7 receptor antagonist (SB-269970, 5mM, 12 µl) was administered intrathecally at C4 to anesthetized, vagotomized and ventilated rats prior to AIH (3, 5-min episodes, 11% O2). Contrary to predictions, pLTF was greater in SB-269970 treated versus control rats (80 ± 11% versus 45 ± 6% 60 min post-AIH; p<0.05). Hypoglossal LTF was unaffected by spinal 5-HT7 receptor inhibition, suggesting that drug effects were localized to the spinal cord. Since 5-HT7 receptors are coupled to protein kinase A (PKA), we tested the hypothesis that PKA inhibits AIH-induced pLTF. Similar to 5-HT7 receptor inhibition, spinal PKA inhibition (KT-5720, 100 µM, 15 µl) enhanced pLTF (99 ± 15% 60 min post-AIH; p<0.05). Conversely, PKA activation (8-br-cAMP, 100 µM, 15 µl) blunted pLTF versus control rats (16 ± 5% versus 45 ± 6% 60 min post-AIH; p<0.05). These findings suggest a novel mechanism whereby spinal Gs protein-coupled 5-HT7 receptors constrain AIH-induced pLTF via PKA activity.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Hipoxia/metabolismo , Potenciación a Largo Plazo/fisiología , Nervio Frénico/fisiología , Receptores de Serotonina/metabolismo , Médula Espinal/metabolismo , Animales , Presión Arterial/efectos de los fármacos , Presión Arterial/fisiología , Análisis de los Gases de la Sangre , Carbazoles/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Interpretación Estadística de Datos , Inhibidores Enzimáticos/farmacología , Nervio Hipogloso/fisiología , Hipoxia/enzimología , Inyecciones Espinales , Masculino , NADPH Oxidasas/metabolismo , Fenoles/farmacología , Pirroles/farmacología , Ratas , Ratas Sprague-Dawley , Receptor Cross-Talk/fisiología , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Serotonina/efectos de los fármacos , Antagonistas de la Serotonina/farmacología , Médula Espinal/efectos de los fármacos , Médula Espinal/enzimología , Sulfonamidas/farmacología
14.
Respir Physiol Neurobiol ; 189(2): 429-37, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23692930

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal, progressive neurodegenerative disease. ALS selectively causes degeneration in upper and lower (spinal) motor neurons, leading to muscle weakness, paralysis and death by ventilatory failure. Although ventilatory failure is generally the cause of death in ALS, little is known concerning the impact of this disorder on respiratory motor neurons, the consequences of respiratory motor neuron cell death, or the ability of the respiratory control system to "fight back" via mechanisms of compensatory respiratory plasticity. Here we review known effects of ALS on breathing, including possible effects on rhythm generation, respiratory motor neurons, and their target organs: the respiratory muscles. We consider evidence for spontaneous compensatory plasticity, preserving breathing well into disease progression despite dramatic loss of spinal respiratory motor neurons. Finally, we review current and potential therapeutic approaches directed toward preserving the capacity to breathe in ALS patients.


Asunto(s)
Esclerosis Amiotrófica Lateral/fisiopatología , Esclerosis Amiotrófica Lateral/terapia , Ventilación no Invasiva/métodos , Ventilación Pulmonar/fisiología , Centro Respiratorio/fisiología , Esclerosis Amiotrófica Lateral/diagnóstico , Animales , Humanos , Ventilación con Presión Positiva Intermitente/métodos , Red Nerviosa/fisiología
15.
J Appl Physiol (1985) ; 114(7): 879-87, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23329821

RESUMEN

Although systemic inflammation occurs in most pathological conditions that challenge the neural control of breathing, little is known concerning the impact of inflammation on respiratory motor plasticity. Here, we tested the hypothesis that low-grade systemic inflammation induced by lipopolysaccharide (LPS, 100 µg/kg ip; 3 and 24 h postinjection) elicits spinal inflammatory gene expression and attenuates a form of spinal, respiratory motor plasticity: phrenic long-term facilitation (pLTF) induced by acute intermittent hypoxia (AIH; 3, 5 min hypoxic episodes, 5 min intervals). pLTF was abolished 3 h (vehicle control: 67.1 ± 27.9% baseline; LPS: 3.7 ± 4.2%) and 24 h post-LPS injection (vehicle: 58.3 ± 17.1% baseline; LPS: 3.5 ± 4.3%). Pretreatment with the nonsteroidal anti-inflammatory drug ketoprofen (12.5 mg/kg ip) restored pLTF 24 h post-LPS (55.1 ± 12.3%). LPS increased inflammatory gene expression in the spleen and cervical spinal cord (homogenates and isolated microglia) 3 h postinjection; however, all molecules assessed had returned to baseline by 24 h postinjection. At 3 h post-LPS, cervical spinal iNOS and COX-2 mRNA were differentially increased in microglia and homogenates, suggesting differential contributions from spinal cells. Thus LPS-induced systemic inflammation impairs AIH-induced pLTF, even after measured inflammatory genes returned to normal. Since ketoprofen restores pLTF even without detectable inflammatory gene expression, "downstream" inflammatory molecules most likely impair pLTF. These findings have important implications for many disease states where acute systemic inflammation may undermine the capacity for compensatory respiratory plasticity.


Asunto(s)
Citocinas/inmunología , Hipoxia/inmunología , Lipopolisacáridos , Potenciación a Largo Plazo/inmunología , Mielitis/inmunología , Nervio Frénico/inmunología , Enfermedad Aguda , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Hipoxia/inducido químicamente , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Mielitis/inducido químicamente , Nervio Frénico/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Síndrome de Respuesta Inflamatoria Sistémica/inducido químicamente , Síndrome de Respuesta Inflamatoria Sistémica/inmunología
16.
Neuroscience ; 229: 77-87, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23159317

RESUMEN

Although respiratory complications are a major cause of morbidity/mortality in many neural injuries or diseases, little is known concerning mechanisms whereby deficient myelin impairs breathing, or how patients compensate for such changes. Here, we tested the hypothesis that respiratory and forelimb motor functions are impaired in a rat model of focal dorsolateral spinal demyelination (ethidium bromide, EB). Ventilation, phrenic nerve activity and horizontal ladder walking were performed 7-14 days post-C2 injection of EB or vehicle (SHAM). EB caused dorsolateral demyelination at C2-C3 followed by significant spontaneous remyelination at 14 days post-EB. Although ventilation did not differ between groups, ipsilateral integrated phrenic nerve burst amplitude was significantly reduced versus SHAM during chemoreceptor activation at 7 days post-EB but recovered by 14 days. The ratio of ipsi- to contralateral phrenic nerve amplitude correlated with cross-sectional lesion area. This ratio was significantly reduced 7 days post-EB versus SHAM during baseline conditions, and versus SHAM and 14-day groups during chemoreceptor activation. Limb function ipsilateral to EB was impaired 7 days post-EB and partially recovered by 14 days post-EB. EB provides a reversible model of focal, spinal demyelination, and may be a useful model to study mechanisms of functional impairment and recovery via motor plasticity, or the efficacy of new therapeutic interventions to reduce severity or duration of disease.


Asunto(s)
Enfermedades Desmielinizantes/fisiopatología , Miembro Anterior/fisiología , Actividad Motora/fisiología , Nervio Frénico/fisiopatología , Respiración , Médula Espinal/fisiopatología , Animales , Vértebras Cervicales , Enfermedades Desmielinizantes/inducido químicamente , Diafragma/inervación , Diafragma/fisiopatología , Etidio , Neuronas Motoras/fisiología , Ratas , Ratas Sprague-Dawley , Caminata/fisiología
17.
J Appl Physiol (1985) ; 113(8): 1184-93, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22961271

RESUMEN

Acute intermittent hypoxia (AIH) elicits a form of spinal respiratory plasticity known as phrenic long-term facilitation (pLTF). pLTF requires spinal serotonin receptor-2 activation, the synthesis of new brain-derived neurotrophic factor (BDNF), and the activation of its high-affinity receptor tyrosine kinase, TrkB. Spinal adenosine 2A receptor activation elicits a distinct pathway to phrenic motor facilitation (pMF); this BDNF synthesis-independent pathway instead requires new synthesis of an immature TrkB isoform. Since hypoxia increases extracellular adenosine levels, we tested the hypothesis that new synthesis of TrkB and BDNF contribute to AIH-induced pLTF. Furthermore, given that signaling mechanisms "downstream" from TrkB are unknown in either mechanism, we tested the hypothesis that pLTF requires MEK/ERK and/or phosphatidylinositol 3-kinase (PI3K)/Akt activation. In anesthetized Sprague-Dawley rats, an intrathecal catheter at cervical level 4 was used to deliver drugs near the phrenic motor nucleus. Since pLTF was blocked by spinal injections of small interfering RNAs targeting BDNF mRNA but not TrkB mRNA, only new BDNF synthesis is required for AIH-induced pLTF. Pretreatment with a MEK inhibitor (U0126) blocked pLTF, whereas a PI3K inhibitor (PI-828) had no effect. Thus, AIH-induced pLTF requires MEK/ERK (not PI3K/AKT) signaling pathways. When U0126 was injected post-AIH, pLTF development was halted but not reversed, suggesting that ERK is critical for the development but not maintenance of pLTF. Thus, there are clear mechanistic distinctions between AIH-induced pLTF (i.e., BDNF synthesis and MEK/ERK dependent) versus adenosine 2A receptor-induced pMF (i.e., TrkB synthesis and PI3K/Akt dependent).


Asunto(s)
Hipoxia/metabolismo , Potenciación a Largo Plazo/fisiología , Sistema de Señalización de MAP Quinasas/genética , Nervio Frénico/fisiología , Receptor trkB/biosíntesis , Animales , Análisis de los Gases de la Sangre/métodos , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Butadienos/farmacología , Hipoxia/enzimología , Hipoxia/genética , Hipoxia/fisiopatología , Inyecciones Espinales/métodos , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/genética , Plasticidad Neuronal/fisiología , Nitrilos/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Nervio Frénico/efectos de los fármacos , Nervio Frénico/metabolismo , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Receptor trkB/genética , Receptor trkB/metabolismo , Receptores de Adenosina A2/genética , Receptores de Adenosina A2/metabolismo , Sistema Respiratorio/efectos de los fármacos , Sistema Respiratorio/metabolismo , Sistema Respiratorio/fisiopatología , Transducción de Señal , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Médula Espinal/fisiología
18.
Exp Neurol ; 234(2): 513-20, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22329943

RESUMEN

Atypical protein kinase C (aPKC) isoforms are expressed in phrenic motor neurons, a group of motor neurons critical for breathing. Following C2 cervical hemisection (C2HS), spontaneous plasticity occurs in crossed-spinal synaptic pathways to phrenic motor neurons, at least partially restoring inspiratory phrenic activity below the injury. Since aPKCs are necessary for synaptic plasticity in other systems, we tested the hypothesis that C2HS increases aPKC expression and activity in spinal regions associated with the phrenic motor nucleus. C2 laminectomy (sham) or C2HS was performed on adult, male Lewis rats. Ventral spinal segments C3-5 were harvested 1, 3 or 28 days post-surgery, and prepared for aPKC enzyme activity assays and immunoblots. Ventral cervical aPKC activity was elevated 1 and 28, but not 3, days post-C2HS (1 day: 63% vs sham ipsilateral to injury; p<0.05; 28 day: 426% vs sham; p<0.05; no difference in ipsilateral vs contralateral response). Total PKCζ/ι protein expression was unchanged by C2HS, but total and phosphorylated PKMζ (constitutively active PKCζ isoform) increased ipsilateral to injury 28 days post-C2HS (p<0.05). Ipsilateral aPKC activity and expression were strongly correlated (r(2)=0.675, p<0.001). In a distinct group of rats, immunohistochemistry confirmed that aPKCs are expressed in neurons 28 days post-C2HS, including large, presumptive phrenic motor neurons; aPKCs were not detected in adjacent microglia (OX-42 positive cells) or astrocytes (GFAP positive cells). Changes in aPKC expression in the phrenic motor nucleus following C2HS suggests that aPKCs may contribute to functional recovery following cervical spinal injury.


Asunto(s)
Neuronas Motoras/metabolismo , Nervio Frénico/metabolismo , Proteína Quinasa C/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Médula Espinal/metabolismo , Animales , Vértebras Cervicales , Masculino , Nervio Frénico/fisiopatología , Ratas , Ratas Endogámicas Lew , Médula Espinal/fisiopatología , Traumatismos de la Médula Espinal/fisiopatología
19.
Respir Physiol Neurobiol ; 178(3): 482-9, 2011 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-21729770

RESUMEN

Many lung and central nervous system disorders require robust and appropriate physiological responses to assure adequate breathing. Factors undermining the efficacy of ventilatory control will diminish the ability to compensate for pathology, threatening life itself. Although most of these same disorders are associated with systemic and/or neuroinflammation, and inflammation affects neural function, we are only beginning to understand interactions between inflammation and any aspect of ventilatory control (e.g. sensory receptors, rhythm generation, chemoreflexes, plasticity). Here we review available evidence, and present limited new data suggesting that systemic (or neural) inflammation impairs two key elements of ventilatory control: chemoreflexes and respiratory motor (versus sensory) plasticity. Achieving an understanding of mechanisms whereby inflammation undermines ventilatory control is fundamental since inflammation may diminish the capacity for natural, compensatory responses during pathological states, and the ability to harness respiratory plasticity as a therapeutic strategy in the treatment of devastating breathing disorders, such as during cervical spinal injury or motor neuron disease.


Asunto(s)
Plasticidad Neuronal , Reflejo , Mecánica Respiratoria , Síndrome de Respuesta Inflamatoria Sistémica/patología , Síndrome de Respuesta Inflamatoria Sistémica/fisiopatología , Animales , Humanos , Neuronas Motoras/patología , Neuronas Motoras/fisiología , Plasticidad Neuronal/fisiología , Reflejo/fisiología , Mecánica Respiratoria/fisiología
20.
Exp Neurol ; 231(1): 97-103, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21683697

RESUMEN

Respiratory failure is the leading cause of death after cervical spinal injury. We hypothesized that incomplete cervical spinal injuries would alter respiratory pattern and initiate plasticity in the neural control of breathing. Further, we hypothesized that the severity of cervical spinal contusion would correlate with changes in breathing pattern. Fourteen days after C4-C5 contusions, respiratory frequency and tidal volume were measured in unanesthetized Sprague Dawley rats in a whole body plethysmograph. Phrenic motor output was monitored in the same rats which were anesthetized, vagotomized, paralyzed and ventilated to eliminate and/or control sensory feedback that could alter breathing patterns. The extent of spinal injury was approximated histologically by measurements of the injury-induced cyst area in transverse sections; cysts ranged from 2 to 28% of spinal cross-sectional area, and had a unilateral bias. In unanesthetized rats, the severity of spinal injury correlated negatively with tidal volume (R(2)=0.85; p<0.001) and positively with breathing frequency (R(2)=0.65; p<0.05). Thus, the severity of C4-C5 spinal contusion dictates post-injury breathing pattern. In anesthetized rats, phrenic burst amplitude was decreased on the side of injury, and burst frequency correlated negatively with contusion size (R(2)=0.51; p<0.05). A strong correlation between unanesthetized breathing pattern and the pattern of phrenic bursts in anesthetized, vagotomized and ventilated rats suggests that changes in respiratory motor output after spinal injury reflect, at least in part, intrinsic neural mechanisms of CNS plasticity initiated by injury.


Asunto(s)
Neuronas Motoras/patología , Neuronas Motoras/fisiología , Nervio Frénico/fisiopatología , Parálisis Respiratoria/fisiopatología , Traumatismos de la Médula Espinal/fisiopatología , Médula Espinal/fisiopatología , Animales , Diafragma/inervación , Diafragma/fisiopatología , Modelos Animales de Enfermedad , Masculino , Ratas , Ratas Sprague-Dawley , Recuperación de la Función/fisiología , Centro Respiratorio/fisiopatología , Mecánica Respiratoria/fisiología , Parálisis Respiratoria/etiología , Índice de Severidad de la Enfermedad , Traumatismos de la Médula Espinal/complicaciones , Volumen de Ventilación Pulmonar/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA