Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Anesthesiology ; 132(5): 1197-1211, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32294065

RESUMO

BACKGROUND: Opioids can induce significant respiratory depression when administered as analgesics for the treatment of acute, postoperative, and chronic pain. There are currently no pharmacologic means of reversing opioid-induced respiratory depression without interfering with analgesia. Further, there is a growing epidemic of opioid overdose that could benefit from therapeutic advancements. The aim of this study was to test the ability of two partial agonists of α4ß2 nicotinic acetylcholine receptors, varenicline (used clinically for smoking cessation) and ABT 594 (tebanicline, developed as an analgesic), to reduce respiratory depression induced by fentanyl, remifentanil, morphine, and a combination of fentanyl and diazepam. METHODS: Whole body plethysmographic recordings, nociception testing, and righting reflex testing were used to examine ventilation, analgesia, and sedation in adult male Sprague-Dawley rats. RESULTS: Pre-, co-, or postadministration of varenicline or ABT 594 did not alter baseline breathing but markedly reduced opioid-induced respiratory depression. Varenicline had no effect on fentanyl-induced analgesia and ABT 594 potentiated fentanyl-induced analgesia. Specifically, 10-min administration of fentanyl induced a decrease in respiratory rate to 43 ± 32% of control in vehicle group, which was alleviated by preadministration of varenicline (85 ± 14% of control, n = 8, P < 0.001) or ABT 594 (81 ± 36% of control, n = 8, P = 0.001). ABT 594 or varenicline with a low dose of naloxone (1 µg/kg), but not varenicline alone, partially reversed fentanyl-induced lethal apnea, but neither compound provided the very rapid and complete reversal of apnea achieved with high doses of naloxone (0.03 to 1 mg/kg). Administration of varenicline (n = 4, P = 0.034) or ABT 594 (n = 4, P = 0.034) prevented lethal apneas induced by the combination of fentanyl and diazepam. CONCLUSIONS: Activation of α4ß2 nicotinic acetylcholine receptors by varenicline and ABT 594 counters opioid-induced respiratory depression without interfering with analgesia.


Assuntos
Analgésicos Opioides/toxicidade , Azetidinas/administração & dosagem , Agonistas Nicotínicos/administração & dosagem , Piridinas/administração & dosagem , Receptores Nicotínicos/fisiologia , Insuficiência Respiratória/prevenção & controle , Vareniclina/administração & dosagem , Animais , Agonismo Parcial de Drogas , Masculino , Ratos , Ratos Sprague-Dawley , Insuficiência Respiratória/induzido quimicamente , Insuficiência Respiratória/fisiopatologia
2.
Am J Physiol Regul Integr Comp Physiol ; 317(1): R190-R202, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31091151

RESUMO

Proinflammatory cytokines like interleukin-1ß (IL-1ß) affect the control of breathing. Our aim is to determine the effect of the anti-inflammatory cytokine IL-10 οn the control of breathing. IL-10 knockout mice (IL-10-/-, n = 10) and wild-type mice (IL-10+/+, n = 10) were exposed to the following test gases: hyperoxic hypercapnia 7% CO2-93% O2, normoxic hypercapnia 7% CO2-21% O2, hypoxic hypercapnia 7% CO2-10% O2, and hypoxic normocapnia 3% CO2-10% O2. The ventilatory function was assessed using whole body plethysmography. Recombinant mouse IL-10 (rIL-10; 10 µg/kg) was administered intraperitoneally to wild-type mice (n = 10) 30 min before the onset of gas challenge. IL-10 was administered in neonatal medullary slices (10-30 ng/ml, n = 8). We found that IL-10-/- mice exhibited consistently increased frequency and reduced tidal volume compared with IL-10+/+ mice during room air breathing and in all test gases (by 23.62 to 33.2%, P < 0.05 and -36.23 to -41.69%, P < 0.05, respectively). In all inspired gases, the minute ventilation of IL-10-/- mice was lower than IL-10+/+ (by -15.67 to -22.74%, P < 0.05). The rapid shallow breathing index was higher in IL-10-/- mice compared with IL-10+/+ mice in all inspired gases (by 50.25 to 57.5%, P < 0.05). The intraperitoneal injection of rIL-10 caused reduction of the respiratory rate and augmentation of the tidal volume in room air and also in all inspired gases (by -12.22 to -29.53 and 32.18 to 45.11%, P < 0.05, respectively). IL-10 administration in neonatal rat (n = 8) in vitro rhythmically active medullary slice preparations did not affect either rhythmicity or peak amplitude of hypoglossal nerve discharge. In conclusion, IL-10 may induce a slower and deeper pattern of breathing.


Assuntos
Dióxido de Carbono/farmacologia , Interleucina-10/metabolismo , Oxigênio/farmacologia , Fenômenos Fisiológicos Respiratórios/efeitos dos fármacos , Animais , Encéfalo/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Interleucina-10/genética , Interleucina-10/farmacologia , Masculino , Bulbo/efeitos dos fármacos , Bulbo/fisiologia , Camundongos , Camundongos Knockout
3.
Anesthesiology ; 130(6): 1017-1031, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31008764

RESUMO

BACKGROUND: Opioid analgesics are widely used for treatment of acute, postoperative, and chronic pain. However, activation of opioid receptors can result in severe respiratory depression. There is an unmet clinical need to develop a pharmacologic therapy to counter opioid-induced respiratory depression without interfering with analgesia. Further, additional advances to confront accidental lethal overdose with the use of fentanyl and other opioids are needed. Here, the authors test the hypothesis that activation of nicotinic receptors expressed within respiratory rhythm-generating networks would counter opioid-induced respiratory depression without compromising analgesia. METHODS: Respiratory neural discharge was measured using in vitro brainstem-spinal cord and medullary slice rat preparations. In vivo, plethysmographic recording, nociception testing, and righting reflexes were used to examine respiratory ventilation, analgesia, and sedation, respectively. RESULTS: The administration of nicotine, selective α4ß2 nicotinic receptor agonist A85380, but not α7 nicotinic receptor agonist PNU282987, reversed opioid-induced respiratory depression in neonatal pups in vitro and in vivo. In adult rats in vivo, administration of A85380 (0.03 mg/kg), but not PNU282987, provides a rapid and robust reversal of fentanyl-induced decrease in respiratory rate (93.4 ± 33.7% of control 3 min after A85380 vs. 31 ± 20.5% of control after vehicle, n = 8 each, P < 0.001), without marked side effects. The coadministration of A85380 (0.06 mg/kg) with fentanyl or remifentanil markedly reduced respiratory depression and apneas, and enhanced the fentanyl-induced analgesia, as evidenced by increased paw withdrawal latency in Hargreaves plantar test (14.4 ± 2.8 s vs. vehicle: 11.3 ± 2.4 s, n = 8 each, P = 0.013) and decreased formalin-induced nocifensive duration (2.5 ± 2.4 min vs. vehicle: 5.4 ± 2.7 min, n = 8 each, P = 0.029). CONCLUSIONS: The novel strategy of targeting α4ß2 nicotinic acetylcholine receptors has the potential for advancing pain control and reducing opioid-induced respiratory depression and overdose.


Assuntos
Analgésicos Opioides/efeitos adversos , Fentanila/efeitos adversos , Receptores Nicotínicos/metabolismo , Insuficiência Respiratória/tratamento farmacológico , Insuficiência Respiratória/metabolismo , Animais , Animais Recém-Nascidos , Azetidinas/farmacologia , Feminino , Masculino , Agonistas Nicotínicos/farmacologia , Agonistas Nicotínicos/uso terapêutico , Gravidez , Ratos , Ratos Sprague-Dawley , Insuficiência Respiratória/induzido quimicamente
4.
Hum Mol Genet ; 25(17): 3798-3809, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27436578

RESUMO

Prader-Willi syndrome is characterized by severe hypotonia in infancy, with decreased lean mass and increased fat mass in childhood followed by severe hyperphagia and consequent obesity. Scoliosis and other orthopaedic manifestations of hypotonia are common in children with Prader-Willi syndrome and cause significant morbidity. The relationships among hypotonia, reduced muscle mass and scoliosis have been difficult to establish. Inactivating mutations in one Prader-Willi syndrome candidate gene, MAGEL2, cause a Prader-Willi-like syndrome called Schaaf-Yang syndrome, highlighting the importance of loss of MAGEL2 in Prader-Willi syndrome phenotypes. Gene-targeted mice lacking Magel2 have excess fat and decreased muscle, recapitulating altered body composition in Prader-Willi syndrome. We now demonstrate that Magel2 is expressed in the developing musculoskeletal system, and that loss of Magel2 causes muscle-related phenotypes in mice consistent with atrophy caused by altered autophagy. Magel2-null mice serve as a preclinical model for therapies targeting muscle structure and function in children lacking MAGEL2 diagnosed with Prader-Willi or Schaaf-Yang syndrome.


Assuntos
Antígenos de Neoplasias/genética , Músculo Esquelético/patologia , Síndrome de Prader-Willi/patologia , Proteínas/genética , Animais , Antígenos de Neoplasias/metabolismo , Autofagia , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Knockout , Músculo Esquelético/embriologia , Músculo Esquelético/metabolismo , Síndrome de Prader-Willi/genética , Proteínas/metabolismo
5.
Am J Respir Cell Mol Biol ; 56(2): 252-260, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27710012

RESUMO

Inhalation of capsaicin-based sprays can cause central respiratory depression and lethal apneas. There are contradictory reports regarding the sites of capsaicin action. Furthermore, an understanding of the neurochemical mechanisms underlying capsaicin-induced apneas and the development of pharmacological interventions is lacking. The main objectives of this study were to perform a systematic study of the mechanisms of action of capsaicin-induced apneas and to provide insights relevant to pharmacological intervention. In vitro and in vivo rat and transient receptor potential vanilloid superfamily member 1 (TRPV1)-null mouse models were used to measure respiratory parameters and seizure-like activity in the presence of capsaicin and compounds that modulate glutamatergic neurotransmission. Administration of capsaicin to in vitro and in vivo rat and wild-type mouse models induced dose-dependent apneas and the production of seizure-like activity. No significant changes were observed in TRPV1-null mice or rat medullary slice preparations. The capsaicin-induced effects were inhibited by the TRPV1 antagonist capsazepine, amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA) receptor antagonists CNQX, NBQX, perampanel, and riluzole, a drug that inhibits glutamate release and increases glutamate uptake. The capsaicin-induced effects on breathing and seizure-like activity were accentuated by positive allosteric modulators of the AMPA receptors, CX717 and cyclothiazide. To summarize, capsaicin-induced apneas and seizure-like behaviors are mediated via TRPV1 activation acting at lung afferents, spinal cord-ascending tracts, and medullary structures (including nucleus tractus solitarius). AMPA receptor-mediated conductances play an important role in capsaicin-induced apneas and seizure-like activity. A pharmaceutical strategy targeted at reducing AMPA receptor-mediated glutamatergic signaling may reduce capsaicin-induced deleterious effects.


Assuntos
Apneia/induzido quimicamente , Apneia/patologia , Animais , Animais Recém-Nascidos , Apneia/metabolismo , Tronco Encefálico/efeitos dos fármacos , Capsaicina/análogos & derivados , Capsaicina/farmacologia , Camundongos Endogâmicos C57BL , Pletismografia , Ratos Sprague-Dawley , Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transdução de Sinais/efeitos dos fármacos , Medula Espinal/efeitos dos fármacos , Canais de Cátion TRPV/antagonistas & inibidores , Canais de Cátion TRPV/metabolismo
6.
Anesthesiology ; 124(3): 641-50, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26675532

RESUMO

BACKGROUND: Drugs acting on µ-opioid receptors (MORs) are widely used as analgesics but present side effects including life-threatening respiratory depression. MORs are G-protein-coupled receptors inhibiting neuronal activity through calcium channels, adenylyl cyclase, and/or G-protein-gated inwardly rectifying potassium (GIRK) channels. The pathways underlying MOR-dependent inhibition of rhythmic breathing are unknown. METHODS: By using a combination of genetic, pharmacological, and physiological tools in rodents in vivo, the authors aimed to identify the role of GIRK channels in MOR-mediated inhibition of respiratory circuits. RESULTS: GIRK channels were expressed in the ventrolateral medulla, a neuronal population regulating rhythmic breathing, and GIRK channel activation with flupirtine reduced respiratory rate in rats (percentage of baseline rate in mean ± SD: 79.4 ± 7.4%, n = 7), wild-type mice (82.6 ± 3.8%, n = 3), but not in mice lacking the GIRK2 subunit, an integral subunit of neuronal GIRK channels (GIRK2, 101.0 ± 1.9%, n = 3). Application of the MOR agonist [D-Ala, N-MePhe, Gly-ol]-enkephalin (DAMGO) to the ventrolateral medulla depressed respiratory rate, an effect partially reversed by the GIRK channel blocker Tertiapin-Q (baseline: 42.1 ± 7.4 breath/min, DAMGO: 26.1 ± 13.4 breath/min, Tertiapin-Q + DAMGO: 33.9 ± 9.8 breath/min, n = 4). Importantly, DAMGO applied to the ventrolateral medulla failed to reduce rhythmic breathing in GIRK2 mice (percentage of baseline rate: 103.2 ± 12.1%, n = 4), whereas it considerably reduced rate in wild-type mice (62.5 ± 17.7% of baseline, n = 4). Respiratory rate depression by systemic injection of the opioid analgesic fentanyl was markedly reduced in GIRK2 (percentage of baseline: 12.8 ± 15.8%, n = 5) compared with wild-type mice (72.9 ± 27.3%). CONCLUSIONS: Overall, these results identify that GIRK channels contribute to respiratory inhibition by MOR, an essential step toward understanding respiratory depression by opioids.


Assuntos
Analgésicos Opioides/toxicidade , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/fisiologia , Insuficiência Respiratória/induzido quimicamente , Insuficiência Respiratória/metabolismo , Animais , Venenos de Abelha/farmacologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/toxicidade , Feminino , Canais de Potássio Corretores do Fluxo de Internalização Acoplados a Proteínas G/antagonistas & inibidores , Masculino , Camundongos , Camundongos Knockout , Ratos , Ratos Wistar , Receptores Opioides mu/agonistas , Receptores Opioides mu/fisiologia
7.
Am J Respir Crit Care Med ; 191(6): 704-10, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25594679

RESUMO

RATIONALE: Apnea of prematurity, which is prevalent among infants born at less than 34 weeks gestation, is treated with caffeine, theophylline, or aminophylline. However, not all newborns respond adequately to, or tolerate, methylxanthine administration, and thus alternative pharmacological therapies are required. OBJECTIVES: Rodent models are used to test the hypothesis that the ampakine CX1739, a positive allosteric modulator of amino-3-hydroxy-5-methyl-4-isoxazolepropionate receptors, strengthens perinatal respiratory drive and reduces apneas. We also provide a systematic study of the effects of caffeine for comparison. METHODS: Respiratory neural activity was recorded from brainstem-spinal cord in vitro perinatal rat preparations, and [Formula: see text]e was recorded in newborn rat pups using whole-body plethysmography under normoxic and hypoxic conditions. MEASUREMENTS AND MAIN RESULTS: Using in vitro brainstem-spinal cord preparations, we found that CX1739 (10-100 µM) dose-dependently increases the frequency of respiratory activity generated by fetal and newborn rat preparations under normoxic and hypoxic conditions. Plethysmographic recordings in vivo from Postnatal Day 0 rats demonstrated that CX1739 (10 mg/kg) increases the frequency and regularity of ventilation, reduces apneas, and protects against hypoxia-induced respiratory depression. CONCLUSIONS: The net effect of ampakine enhancement of respiratory drive in perinatal rodents is a marked increase in ventilation and the regularity of respiratory patterns in perinatal rat preparations. Importantly, from the perspective of clinical applications, CX1739 readily crosses the blood-brain barrier, is metabolically stable, and has passed through phase I and II clinical trials in adults.


Assuntos
Analgésicos Opioides/farmacologia , Apneia/tratamento farmacológico , Respiração/efeitos dos fármacos , Síndrome do Desconforto Respiratório do Recém-Nascido/tratamento farmacológico , Animais , Animais Recém-Nascidos , Tronco Encefálico/efeitos dos fármacos , Cafeína/farmacologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feto/efeitos dos fármacos , Técnicas In Vitro , Pletismografia , Ratos , Medula Espinal/efeitos dos fármacos
8.
Vet Anaesth Analg ; 43(5): 528-38, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27531058

RESUMO

OBJECTIVES: To determine whether CX1942 reverses respiratory depression in etorphine-immobilized goats, and to compare its effects with those of doxapram hydrochloride. STUDY DESIGN: A prospective, crossover experimental trial conducted at 1753 m.a.s.l. ANIMALS: Eight adult female Boer goats (Capra hircus) with a mean ± standard deviation mass of 27.1 ± 1.6 kg. METHODS: Following immobilization with 0.1 mg kg(-1) etorphine, goats received one of doxapram, CX1942 or sterile water intravenously, in random order in three trials. Respiratory rate, ventilation and tidal volume were measured continuously. Arterial blood samples for the determination of PaO2 , PaCO2 , pH and SaO2 were taken 2 minutes before and then at 5 minute intervals after drug administration for 25 minutes. RESULTS: Doxapram corrected etorphine-induced respiratory depression but also led to arousal and hyperventilation at 2 minutes after its administration, as indicated by the low PaCO2 (27.8 ± 4.5 mmHg) and ventilation of 5.32 ± 5.24 L minute(-1) above pre-immobilization values. CX1942 improved respiratory parameters and corrected etorphine's hypoxaemic effects more gradually than did doxapram, with a more sustained improvement in PaO2 and SaO2 in comparison with the control trial. CONCLUSIONS: CX1942 attenuated opioid-induced respiratory depression and corrected the hypoxaemic effects of etorphine in immobilized goats. CLINICAL RELEVANCE: Ampakines potentially offer advantages over doxapram, a conventional treatment, in reversing etorphine-induced respiratory depression without causing unwanted side effects, particularly arousal, in immobilized animals.


Assuntos
Analgésicos Opioides/farmacologia , Etorfina/farmacologia , Hipóxia/induzido quimicamente , Receptores de AMPA/agonistas , Insuficiência Respiratória/tratamento farmacológico , Animais , Doxapram/farmacologia , Feminino , Cabras , Hipóxia/tratamento farmacológico , Imobilização , Naltrexona/administração & dosagem , Antagonistas de Entorpecentes/farmacologia , Insuficiência Respiratória/induzido quimicamente , Medicamentos para o Sistema Respiratório/farmacologia
9.
Am J Respir Cell Mol Biol ; 53(3): 326-35, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25569118

RESUMO

Pompe disease results from a mutation in the acid α-glucosidase gene leading to lysosomal glycogen accumulation. Respiratory insufficiency is common, and the current U.S. Food and Drug Administration-approved treatment, enzyme replacement, has limited effectiveness. Ampakines are drugs that enhance α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor responses and can increase respiratory motor drive. Recent work indicates that respiratory motor drive can be blunted in Pompe disease, and thus pharmacologic stimulation of breathing may be beneficial. Using a murine Pompe model with the most severe clinical genotype (the Gaa(-/-) mouse), our primary objective was to test the hypothesis that ampakines can stimulate respiratory motor output and increase ventilation. Our second objective was to confirm that neuropathology was present in Pompe mouse medullary respiratory control neurons. The impact of ampakine CX717 on breathing was determined via phrenic and hypoglossal nerve recordings in anesthetized mice and whole-body plethysmography in unanesthetized mice. The medulla was examined using standard histological methods coupled with immunochemical markers of respiratory control neurons. Ampakine CX717 robustly increased phrenic and hypoglossal inspiratory bursting and reduced respiratory cycle variability in anesthetized Pompe mice, and it increased inspiratory tidal volume in unanesthetized Pompe mice. CX717 did not significantly alter these variables in wild-type mice. Medullary respiratory neurons showed extensive histopathology in Pompe mice. Ampakines stimulate respiratory neuromotor output and ventilation in Pompe mice, and therefore they have potential as an adjunctive therapy in Pompe disease.


Assuntos
Doença de Depósito de Glicogênio Tipo II/tratamento farmacológico , Isoxazóis/farmacologia , Respiração/efeitos dos fármacos , Medicamentos para o Sistema Respiratório/farmacologia , Animais , Tronco Encefálico/patologia , Avaliação Pré-Clínica de Medicamentos , Doença de Depósito de Glicogênio Tipo II/fisiopatologia , Isoxazóis/uso terapêutico , Camundongos da Linhagem 129 , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Nervo Frênico/efeitos dos fármacos , Nervo Frênico/fisiopatologia , Medicamentos para o Sistema Respiratório/uso terapêutico
10.
Anesthesiology ; 122(2): 424-34, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25313880

RESUMO

BACKGROUND: There is an unmet clinical need to develop a pharmacological therapy to counter opioid-induced respiratory depression without interfering with analgesia or behavior. Several studies have demonstrated that 5-HT1A receptor agonists alleviate opioid-induced respiratory depression in rodent models. However, there are conflicting reports regarding their effects on analgesia due in part to varied agonist receptor selectivity and presence of anesthesia. Therefore the authors performed a study in rats with befiradol (F13640 and NLX-112), a highly selective 5-HT1A receptor agonist without anesthesia. METHODS: Respiratory neural discharge was measured using in vitro preparations. Plethysmographic recording, nociception testing, and righting reflex were used to examine respiratory ventilation, analgesia, and sedation, respectively. RESULTS: Befiradol (0.2 mg/kg, n = 6) reduced fentanyl-induced respiratory depression (53.7 ± 5.7% of control minute ventilation 4 min after befiradol vs. saline 18.7 ± 2.2% of control, n = 9; P < 0.001), duration of analgesia (90.4 ± 11.6 min vs. saline 130.5 ± 7.8 min; P = 0.011), duration of sedation (39.8 ± 4 min vs. saline 58 ± 4.4 min; P = 0.013); and induced baseline hyperventilation, hyperalgesia, and "behavioral syndrome" in nonsedated rats. Further, the befiradol-induced alleviation of opioid-induced respiratory depression involves sites or mechanisms not functioning in vitro brainstem-spinal cord and medullary slice preparations. CONCLUSIONS: The reversal of opioid-induced respiratory depression and sedation by befiradol in adult rats was robust, whereas involved mechanisms are unclear. However, there were adverse concomitant decreases in fentanyl-induced analgesia and altered baseline ventilation, nociception, and behavior.


Assuntos
Analgesia , Anestésicos Intravenosos/farmacologia , Sedação Consciente , Fentanila/antagonistas & inibidores , Piperidinas/farmacologia , Piridinas/farmacologia , Receptor 5-HT1A de Serotonina/efeitos dos fármacos , Insuficiência Respiratória/prevenção & controle , Agonistas do Receptor 5-HT1 de Serotonina/farmacologia , Envelhecimento/fisiologia , Anestésicos Intravenosos/toxicidade , Animais , Nível de Alerta , Comportamento Animal/efeitos dos fármacos , Tronco Encefálico/efeitos dos fármacos , Fentanila/farmacologia , Fentanila/toxicidade , Técnicas In Vitro , Bulbo/efeitos dos fármacos , Medição da Dor/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Insuficiência Respiratória/induzido quimicamente , Medula Espinal/efeitos dos fármacos
11.
J Neurosci ; 32(48): 17230-40, 2012 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-23197715

RESUMO

Rett syndrome (RTT) is a severe neurological disorder that is associated with mutations in the methyl-CpG binding protein 2 (MECP2) gene. RTT patients suffer from mental retardation and behavioral disorders, including heightened anxiety and state-dependent breathing irregularities, such as hyperventilation and apnea. Many symptoms are recapitulated by the Mecp2-null male mice (Mecp2(-/y)). To characterize developmental progression of the respiratory phenotype and explore underlying mechanisms, we examined Mecp2(-/y) and wild-type (WT) mice from presymptomatic periods to end-stage disease. We monitored breathing patterns of unrestrained mice during wake-sleep states and while altering stress levels using movement restraint or threatening odorant (trimethylthiazoline). Respiratory motor patterns generated by in situ working heart-brainstem preparations (WHBPs) were measured to assess function of brainstem respiratory networks isolated from suprapontine structures. Data revealed two general stages of respiratory dysfunction in Mecp2(-/y) mice. At the early stage, respiratory abnormalities were limited to wakefulness, correlated with markers of stress (increased fecal deposition and blood corticosterone levels), and alleviated by antalarmin (corticotropin releasing hormone receptor 1 antagonist). Furthermore, the respiratory rhythm generated by WHBPs was similar in WT and Mecp2(-/y) mice. During the later stage, respiratory abnormalities were evident during wakefulness and sleep. Also, WHBPs from Mecp2(-/y) showed central apneas. We conclude that, at early disease stages, stress-related modulation from suprapontine structures is a significant factor in the Mecp2(-/y) respiratory phenotype and that anxiolytics may be effective. At later stages, abnormalities of brainstem respiratory networks are a significant cause of irregular breathing patterns and central apneas.


Assuntos
Ansiedade/fisiopatologia , Proteína 2 de Ligação a Metil-CpG/metabolismo , Transtornos Respiratórios/psicologia , Respiração , Síndrome de Rett/fisiopatologia , Animais , Ansiedade/genética , Ansiedade/psicologia , Modelos Animais de Doenças , Masculino , Proteína 2 de Ligação a Metil-CpG/genética , Camundongos , Transtornos Respiratórios/genética , Transtornos Respiratórios/fisiopatologia , Síndrome de Rett/genética , Síndrome de Rett/psicologia , Vigília/fisiologia
12.
J Neurosci ; 32(33): 11259-70, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22895710

RESUMO

Respiratory activity is most fragile during sleep, in particular during paradoxical [or rapid eye movement (REM)] sleep and sleep state transitions. Rats are commonly used to study respiratory neuromodulation, but rodent sleep is characterized by a highly fragmented sleep pattern, thus making it very challenging to examine different sleep states and potential pharmacological manipulations within them. Sleep-like brain-state alternations occur in rats under urethane anesthesia and may be an effective and efficient model for sleep itself. The present study assessed state-dependent changes in breathing and respiratory muscle modulation under urethane anesthesia to determine their similarity to those occurring during natural sleep. Rats were anesthetized with urethane and respiratory airflow, as well as electromyographic activity in respiratory muscles were recorded in combination with local field potentials in neocortex and hippocampus to determine how breathing pattern and muscle activity are modulated with brain state. Measurements were made in normoxic, hypoxic, and hypercapnic conditions. Results were compared with recordings made from rats during natural sleep. Brain-state alternations under urethane anesthesia were closely correlated with changes in breathing rate and variability and with modulation of respiratory muscle tone. These changes closely mimicked those observed in natural sleep. Of great interest was that, during both REM and REM-like states, genioglossus muscle activity was strongly depressed and abdominal muscle activity showed potent expiratory modulation. We demonstrate that, in urethane-anesthetized rats, respiratory airflow and muscle activity are closely correlated with brain-state transitions and parallel those shown in natural sleep, providing a useful model to systematically study sleep-related changes in respiratory control.


Assuntos
Anestésicos Intravenosos/farmacologia , Ondas Encefálicas/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Respiração/efeitos dos fármacos , Fases do Sono/efeitos dos fármacos , Uretana/farmacologia , Músculos Abdominais/efeitos dos fármacos , Músculos Abdominais/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Encéfalo/fisiologia , Ondas Encefálicas/fisiologia , Eletroencefalografia , Eletromiografia , Hipercapnia/induzido quimicamente , Hipercapnia/fisiopatologia , Hipóxia/fisiopatologia , Masculino , Ratos , Ratos Sprague-Dawley , Análise Espectral , Volume de Ventilação Pulmonar/efeitos dos fármacos , Vigília/efeitos dos fármacos , Vigília/fisiologia
13.
Anesthesiology ; 118(6): 1437-45, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23542802

RESUMO

BACKGROUND: Propofol (2,6-diisopropylphenol) is used for the induction and maintenance of anesthesia in human and veterinary medicine. Propofol's disadvantages include the induction of respiratory depression and apnea. Here, the authors report a clinically feasible pharmacological solution for reducing propofol-induced respiratory depression via a mechanism that does not interfere with anesthesia. Specifically, they test the hypothesis that the AMPAKINE CX717, which has been proven metabolically stable and safe for human use, can prevent and rescue from propofol-induced severe apnea. METHODS: The actions of propofol and the AMPAKINE CX717 were measured via (1) ventral root recordings from newborn rat brainstem-spinal cord preparations, (2) phrenic nerve recordings from an adult mouse in situ working heart-brainstem preparation, and (3) plethysmographic recordings from unrestrained newborn and adult rats. RESULTS: In vitro, respiratory depression caused by propofol (2 µM, n = 11, mean ± SEM, 41 ± 5% of control frequency, 63 ± 5% of control duration) was alleviated by CX717 (n = 4, 50-150 µM). In situ, a decrease in respiratory frequency (44 ± 9% of control), phrenic burst duration (66 ± 7% of control), and amplitude (78 ± 5% of control) caused by propofol (2 µM, n = 5) was alleviated by coadministration of CX717 (50 µM, n = 5). In vivo, pre- or coadministration of CX717 (20-25mg/kg) with propofol markedly reduced propofol-induced respiratory depression (n = 7; 20mg/kg) and propofol-induced lethal apnea (n = 6; 30 mg/kg). CONCLUSIONS: Administration of CX717 before or in conjunction with propofol provides an increased safety margin against profound apnea and death.


Assuntos
Anestésicos Intravenosos/farmacologia , Apneia/complicações , Apneia/prevenção & controle , Isoxazóis/farmacologia , Propofol/farmacologia , Insuficiência Respiratória/prevenção & controle , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Quimioterapia Combinada/métodos , Masculino , Camundongos , Pletismografia/métodos , Ratos , Ratos Sprague-Dawley , Insuficiência Respiratória/complicações
14.
J Neurosci ; 31(4): 1292-301, 2011 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-21273414

RESUMO

The analgesic properties of the opium poppy Papever somniferum were first mentioned by Hippocrates around 400 BC, and opioid analgesics remain the mainstay of pain management today. These drugs can cause the serious side-effect of respiratory depression that can be lethal with overdose, however the critical brain sites and neurochemical identity of the neurons mediating this depression are unknown. By locally manipulating neurotransmission in the adult rat, we identify the critical site of the medulla, the preBötzinger complex, that mediates opioid-induced respiratory depression in vivo. Here we show that opioids at the preBötzinger complex cause respiratory depression or fatal apnea, with anesthesia and deep-sleep being particularly vulnerable states for opioid-induced respiratory depression. Importantly, we establish that the preBötzinger complex is fully responsible for respiratory rate suppression following systemic administration of opioid analgesics. The site in the medulla most sensitive to opioids corresponds to a region expressing neurokinin-1 receptors, and we show in rhythmically active brainstem section in vitro that neurokinin-1 receptor-expressing preBötzinger complex neurons are selectively inhibited by opioids. In summary, neurokinin-1 receptor-expressing preBötzinger complex neurons constitute the critical site mediating opioid-induced respiratory rate depression, and the key therapeutic target for its prevention or reversal.


Assuntos
Analgésicos Opioides/efeitos adversos , Bulbo/fisiologia , Neurônios/fisiologia , Receptores da Neurocinina-1/biossíntese , Respiração/efeitos dos fármacos , Anestesia , Animais , Apneia/induzido quimicamente , Tronco Encefálico/efeitos dos fármacos , Tronco Encefálico/metabolismo , Depressão Química , Técnicas In Vitro , Masculino , Periodicidade , Ratos , Ratos Sprague-Dawley , Receptores Opioides mu/agonistas , Taxa Respiratória/efeitos dos fármacos , Sono , Transmissão Sináptica
15.
Proc Natl Acad Sci U S A ; 106(51): 21966-71, 2009 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-20007372

RESUMO

Rett syndrome (RTT) is characterized by specific motor, cognitive, and behavioral deficits. Because several of these abnormalities occur in other disease states associated with alterations in aminergic neurotransmitters, we investigated the contribution of such alterations to RTT pathogenesis. We found that both individuals with RTT and Mecp2-null mice have lower-than-normal levels of aminergic metabolites and content. Deleting Mecp2 from either TH-positive dopaminergic and noradrenergic neurons or PET1-positive serotonergic neurons in mice decreased corresponding neurotransmitter concentration and specific phenotypes, likely through MeCP2 regulation of rate-limiting enzymes involved in aminergic neurotransmitter production. These data support a cell-autonomous, MeCP2-dependent mechanism for the regulation of aminergic neurotransmitter synthesis contributing to unique behavioral phenotypes.


Assuntos
Aminas/metabolismo , Ácido Homovanílico/metabolismo , Ácido Hidroxi-Indolacético/metabolismo , Transtornos Mentais/metabolismo , Proteína 2 de Ligação a Metil-CpG/metabolismo , Neurônios/metabolismo , Animais , Proteína 2 de Ligação a Metil-CpG/genética , Camundongos , Camundongos Knockout , Neurônios/enzimologia , Fenótipo , Triptofano Hidroxilase/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
16.
J Neurotrauma ; 38(24): 3467-3482, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34806433

RESUMO

Respiratory compromise after cervical spinal cord injury (SCI) is a leading cause of mortality and morbidity. Most SCIs are incomplete, and spinal respiratory motoneurons as well as proprio- and bulbospinal synaptic pathways provide a neurological substrate to enhance respiratory output. Ampakines are allosteric modulators of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, which are prevalent on respiratory neurons. We hypothesized that low dose ampakine treatment could safely and effectively increase diaphragm electromyography (EMG) activity that has been impaired as a result of acute- or sub-acute cervical SCI. Diaphragm EMG was recorded using chronic indwelling electrodes in unanesthetized, freely moving rats. A spinal hemi-lesion was induced at C2 (C2Hx), and rats were studied at 4 and 14 days post-injury during room air breathing and acute respiratory challenge accomplished by inspiring a 10% O2, 7% CO2 gas mixture. Once a stable baseline recording was established, one of two different ampakines (CX717 or CX1739, 5 mg/kg, intravenous) or a vehicle (2-hydroxypropyl-beta-cyclodextrin [HPCD]) was delivered. At 4 days post-injury, both ampakines increased diaphragm EMG output ipsilateral to C2Hx during both baseline breathing and acute respiratory challenge. Only CX1739 treatment also led to a sustained (15 min) increase in ipsilateral EMG output. At 14 days post-injury, both ampakines produced sustained increases in ipsilateral diaphragm EMG output and enabled increased output during the respiratory challenge. We conclude that low dose ampakine treatment can increase diaphragm EMG activity after cervical SCI, and therefore may provide a pharmacological strategy that could be useful in the context of respiratory rehabilitation.


Assuntos
Medula Cervical/lesões , Diafragma/efeitos dos fármacos , Diafragma/fisiopatologia , Isoxazóis/uso terapêutico , Traumatismos da Medula Espinal/complicações , Animais , Vértebras Cervicais , Eletromiografia , Feminino , Masculino , Ratos , Ratos Sprague-Dawley , Receptores de AMPA/efeitos dos fármacos , Traumatismos da Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/terapia
17.
J Neurosci ; 29(47): 14713-25, 2009 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-19940166

RESUMO

ATP released during hypoxia from the ventrolateral medulla activates purinergic receptors (P2Rs) to attenuate the secondary hypoxic depression of breathing by a mechanism that likely involves a P2Y(1)R-mediated excitation of preBötzinger complex (preBötC) inspiratory rhythm-generating networks. In this study, we used rhythmically active in vitro preparations from embryonic and postnatal rats and ATP microinjection into the rostral ventral respiratory group (rVRG)/preBötC to reveal that these networks are sensitive to ATP when rhythm emerges at embryonic day 17 (E17). The peak frequency elicited by ATP at E19 and postnatally was the same ( approximately 45 bursts/min), but relative sensitivity was threefold greater at E19, reflecting a lower baseline frequency (5.6 +/- 0.9 vs 19.0 +/- 1.3 bursts/min). Combining microinjection techniques with ATP biosensors revealed that ATP concentration in the rVRG/preBötC falls rapidly as a result of active processes and closely correlates with inspiratory frequency. A phosphate assay established that preBötC-containing tissue punches degrade ATP at rates that increase perinatally. Thus, the agonist profile [ATP/ADP/adenosine (ADO)] produced after ATP release in the rVRG/preBötC will change perinatally. Electrophysiology further established that the ATP metabolite ADP is excitatory and that, in fetal but not postnatal animals, ADO at A(1) receptors exerts a tonic depressive action on rhythm, whereas A(1) antagonists extend the excitatory action of ATP on inspiratory rhythm. These data demonstrate that ATP is a potent excitatory modulator of the rVRG/preBötC inspiratory network from the time it becomes active and that ATP actions are determined by a dynamic interaction between the actions of ATP at P2 receptors, ectonucleotidases that degrade ATP, and ATP metabolites on P2Y and P1 receptors.


Assuntos
Trifosfato de Adenosina/metabolismo , Centro Respiratório/crescimento & desenvolvimento , Centro Respiratório/metabolismo , Rombencéfalo/crescimento & desenvolvimento , Rombencéfalo/metabolismo , 5'-Nucleotidase/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Adenosina/metabolismo , Agonistas do Receptor A1 de Adenosina , Antagonistas do Receptor A1 de Adenosina , Difosfato de Adenosina/metabolismo , Difosfato de Adenosina/farmacologia , Trifosfato de Adenosina/farmacologia , Animais , Animais Recém-Nascidos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Inibição Neural/efeitos dos fármacos , Inibição Neural/fisiologia , Técnicas de Cultura de Órgãos , Periodicidade , Ratos , Ratos Sprague-Dawley , Receptor A1 de Adenosina/metabolismo , Receptores Purinérgicos P2/efeitos dos fármacos , Receptores Purinérgicos P2/metabolismo , Respiração , Centro Respiratório/efeitos dos fármacos , Rombencéfalo/efeitos dos fármacos
18.
Am J Respir Cell Mol Biol ; 42(3): 276-85, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19448158

RESUMO

Congenital diaphragmatic hernia (CDH) is a frequently occurring source of severe neonatal respiratory distress. It has been hypothesized that abnormal retinoid signaling contributes to the etiology of this developmental anomaly. Here, we use rodent models toward specifically understanding the role of retinoid signaling in the developing diaphragm and how its perturbation is a common mechanism in drug-induced CDH. This includes monitoring of retinoic acid (RA) response element (RARE) activation with RARE-lacZ mice, RA supplementation studies, systematic analyses of the expression profile of key elements in the RA signaling pathway within the developing diaphragm, and the in utero delivery of a RA receptor (RAR) antagonist. These data demonstrate the timing of RARE perturbation by CDH-inducing teratogens and the efficacy of RA supplementation. Furthermore, a detailed profile of retinoid binding proteins, synthetic enzymes, and retinoid receptors within primordial diaphragm cells was obtained. The expression profile of RAR-alpha was particularly striking in regard to its overlap with the regions of primordial diaphragm affected in multiple CDH models. Blocking of RAR signaling with the pan-RAR antagonist BMS493 induced a very high degree of CDH, with a marked left-right sidedness that depended on the timing of drug delivery. Collectively, these data demonstrate that retinoid signaling is essential for normal diaphragm development, providing further support to the hypothesis that abnormalities related to the retinoid signaling pathway cause diaphragmatic defects. This study also yielded a novel experimental model that should prove particularly useful for further studies of CDH.


Assuntos
Hérnia Diafragmática/etiologia , Hérnias Diafragmáticas Congênitas , Retinoides/metabolismo , Transdução de Sinais , Animais , Diafragma/efeitos dos fármacos , Diafragma/embriologia , Diafragma/enzimologia , Diafragma/patologia , Suplementos Nutricionais , Ativação Enzimática/efeitos dos fármacos , Hérnia Diafragmática/metabolismo , Hérnia Diafragmática/prevenção & controle , Camundongos , Ratos , Receptores do Ácido Retinoico/antagonistas & inibidores , Receptores do Ácido Retinoico/metabolismo , Elementos de Resposta/genética , Retinal Desidrogenase/metabolismo , Transdução de Sinais/efeitos dos fármacos , Estilbenos/farmacologia , Teratogênicos , Tretinoína/administração & dosagem , Tretinoína/farmacologia , beta-Galactosidase/metabolismo
19.
Hum Mol Genet ; 17(12): 1718-27, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18321864

RESUMO

Rett Syndrome, an X-linked dominant neurodevelopmental disorder characterized by regression of language and hand use, is primarily caused by mutations in methyl-CpG-binding protein 2 (MECP2). Loss of function mutations in MECP2 are also found in other neurodevelopmental disorders such as autism, Angelman-like syndrome and non-specific mental retardation. Furthermore, duplication of the MECP2 genomic region results in mental retardation with speech and social problems. The common features of human neurodevelopmental disorders caused by the loss or increase of MeCP2 function suggest that even modest alterations of MeCP2 protein levels result in neurodevelopmental problems. To determine whether a small reduction in MeCP2 level has phenotypic consequences, we characterized a conditional mouse allele of Mecp2 that expresses 50% of the wild-type level of MeCP2. Upon careful behavioral analysis, mice that harbor this allele display a spectrum of abnormalities such as learning and motor deficits, decreased anxiety, altered social behavior and nest building, decreased pain recognition and disrupted breathing patterns. These results indicate that precise control of MeCP2 is critical for normal behavior and predict that human neurodevelopmental disorders will result from a subtle reduction in MeCP2 expression.


Assuntos
Proteína 2 de Ligação a Metil-CpG/genética , Proteína 2 de Ligação a Metil-CpG/metabolismo , Síndrome de Rett/genética , Síndrome de Rett/fisiopatologia , Tonsila do Cerebelo/metabolismo , Animais , Ansiedade/genética , Ansiedade/fisiopatologia , Peso Corporal , Encéfalo/metabolismo , Cruzamentos Genéticos , Feminino , Imunofluorescência , Hipocampo/metabolismo , Humanos , Aprendizagem , Masculino , Camundongos , Comportamento de Nidação , Dor/fisiopatologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Desempenho Psicomotor , Reflexo de Sobressalto , Síndrome de Rett/psicologia , Teste de Desempenho do Rota-Rod , Comportamento Social
20.
Birth Defects Res A Clin Mol Teratol ; 88(1): 15-24, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19711422

RESUMO

Congenital diaphragmatic hernia (CDH) is a frequently occurring cause of neonatal respiratory distress and is associated with high mortality and long-term morbidity. Evidence from animal models suggests that CDH has its origins in the malformation of the pleuroperitoneal fold (PPF), a key structure in embryonic diaphragm formation. The aims of this study were to characterize the embryogenesis of the PPF in rats and humans, and to determine the potential mechanism that leads to abnormal PPF development in the nitrofen model of CDH. Analysis of rat embryos, and archived human embryo sections, allowed the timeframe of PPF formation to be determined for both species, thus delineating a critical period of diaphragm development in relation to CDH. Experiments on nitrofen-exposed NIH 3T3 cells in vitro led us to hypothesize that nitrofen might cause diaphragmatic hernia in vivo by two possible mechanisms: through decreased cell proliferation or by inducing apoptosis. Data from nitrofen-exposed rat embryos indicates that the primary mechanism of nitrofen teratogenesis in the PPF is through decreased cell proliferation. This study provides novel insight into the embryogenesis of the PPF in rats and humans, and it indicates that impaired cell proliferation might contribute to abnormal diaphragm development in the nitrofen model of CDH.


Assuntos
Anormalidades Induzidas por Medicamentos , Diafragma/anormalidades , Embrião de Mamíferos/anormalidades , Hérnia Diafragmática/embriologia , Praguicidas/toxicidade , Éteres Fenílicos/toxicidade , Animais , Apoptose/efeitos dos fármacos , Bromodesoxiuridina/metabolismo , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Diafragma/efeitos dos fármacos , Modelos Animais de Doenças , Embrião de Mamíferos/efeitos dos fármacos , Feminino , Hérnia Diafragmática/induzido quimicamente , Hérnias Diafragmáticas Congênitas , Humanos , Técnicas Imunoenzimáticas , Marcação In Situ das Extremidades Cortadas , Exposição Materna , Camundongos , Células NIH 3T3/efeitos dos fármacos , Células NIH 3T3/patologia , Células NIH 3T3/fisiologia , Peritônio/anormalidades , Peritônio/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa