Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Physiol ; 601(24): 5509-5525, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36988138

RESUMEN

Sleep disordered breathing (SDB) is a complex, sex specific and highly heterogeneous group of respiratory disorders. Nevertheless, sleep fragmentation and repeated fluctuations of arterial blood gases for several hours per night are at the core of the problem; together, they impose significant stress to the organism with deleterious consequences on physical and mental health. SDB increases the risk of obesity, diabetes, depression and anxiety disorders; however, the same health issues are risk factors for SDB. So, which came first, the chicken or the egg? What causes the appearance of the first significant apnoeic events during sleep? These are important questions because although moderate to severe SDB affects ∼500 million adults globally, we still have a poor understanding of the origins of the disease, and the main treatments (and animal models) focus on the symptoms rather than the cause. Because obesity, metabolic dysfunction and stress-related neurological disorders generally appear progressively, we discuss how the development of these diseases can lead to specific anatomical and non-anatomical traits of SDB in males and females while considering the impacts of sex steroids. In light of the growing evidence indicating that the carotid bodies are important sensors of key metabolic and endocrine signals associated with stress and dysmetabolism, we propose that these organs play a key role in the process.


Asunto(s)
Diabetes Mellitus , Síndromes de la Apnea del Sueño , Masculino , Femenino , Humanos , Factores de Riesgo , Obesidad/complicaciones , Sueño
2.
Exp Physiol ; 108(11): 1376-1385, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37642495

RESUMEN

Sleep-disordered breathing is a respiratory disorder commonly experienced by pregnant women. The recurrent hypoxaemic events associated with sleep-disordered breathing have deleterious consequences for the mother and fetus. Adult male (but not female) rats born to dams subjected to gestational intermittent hypoxia (GIH) have a higher resting blood pressure than control animals and show behavioural/neurodevelopmental disorders. The origin of this persistent, sex-specific effect of GIH in offspring is unknown, but disruption of the neuroendocrine stress pathways is a key mechanism by which gestational stress increases disease risk in progeny. Using FosB immunolabelling as a chronic marker of neuronal activation, we determined whether GIH augments basal expression of FosB in the perikaryas of cells in the paraventricular nucleus of the hypothalamus (PVN), a key structure in the regulation of the stress response and blood pressure. From gestational day 10, female rats were subjected to GIH for 8 h/day (light phase) until the day before delivery (gestational day 21); GIH consisted of 2 min hypoxic bouts (10.5% O2 ) alternating with normoxia. Control rats were exposed to intermittent normoxia over the same period (GNX). At adulthood (10-15 weeks), the brains of male and female rats were harvested for FosB immunohistochemistry. In males, GIH augmented PVN FosB labelling density by 30%. Conversely, PVN FosB density in GIH females was 28% lower than that of GNX females. We conclude that GIH has persistent and sex-specific impacts on the development of stress pathways, thereby offering a plausible mechanism by which GIH can disturb neural development and blood pressure homeostasis in adulthood. NEW FINDINGS: What is the central question of this study? In pregnant women, sleep apnoea increases the risk of disease for the offspring at various life stages. Given that gestational stress disrupts the programming of the stress pathways, we determined whether exposing female rats to gestational intermittent hypoxia (GIH) activates hypothalamic neurons regulating the stress response in adult rats. What is the main finding and its importance? Using FosB immunolabelling as a marker of marker of neuronal activation, we showed that GIH augmented basal activation of the paraventricular nucleus of the hypothalamus in males, but not females. Disruption of the stress pathways is a new hypothesis to explain the persistent and sex-specific impacts of GIH on offspring health.


Asunto(s)
Hipertensión , Síndromes de la Apnea del Sueño , Animales , Femenino , Humanos , Masculino , Embarazo , Ratas , Hipotálamo/metabolismo , Hipoxia , Núcleo Hipotalámico Paraventricular/metabolismo , Ratas Sprague-Dawley
3.
Adv Exp Med Biol ; 1427: 61-71, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37322336

RESUMEN

Sleep apnea (SA) is a major respiratory disorder with increased risk for hypertension and obesity; however, our understanding of the origins of this complex disorder remains limited. Because apneas lead to recurrent drops in O2 during sleep, intermittent hypoxia (IH) is the main animal model to explore the pathophysiology of SA. Here, we assessed the impacts of IH on metabolic function and related signals. Adult male rats were exposed to 1 week of moderate IH (FiO2 = 0.10-30 s, ten cycles/hour, 8 h/day). Using whole-body plethysmography, we measured respiratory variability and apnea index during sleep. Blood pressure and heart rate were measured by the tail-cuff method; blood samples were taken for multiplex assay. At rest, IH augmented arterial blood pressure, respiratory instability, but not apnea index. IH induced weight, fat, and fluid loss. IH also reduced food intake and plasma leptin, adrenocorticotropic hormone (ACTH), and testosterone levels but increased inflammatory cytokines. We conclude that IH does not replicate the metabolic clinical features of SA patient, thus raising our awareness of the limitations of the IH model. The fact that the risk for hypertension occurs before the appearance of apneas provides new insights into the progression of the disease.


Asunto(s)
Hipertensión , Síndromes de la Apnea del Sueño , Masculino , Ratas , Animales , Hipoxia , Hipertensión/etiología , Pérdida de Peso , Fenotipo
4.
Artículo en Inglés | MEDLINE | ID: mdl-37088410

RESUMEN

Tadpole development is influenced by environmental cues and hypoxia can favor the emergence of the neural networks driving air breathing. Exposing isolated brainstems from pre-metamorphic tadpoles to acute hypoxia (∼0% O2; 15 min) leads to a progressive increase in fictive air breaths (∼3 fold) in the hours that follow stimulation. Here, we first determined whether this effect persists over longer periods (<18 h); we then evaluated maturity of the motor output by comparing the breathing pattern of hypoxia-exposed brainstems to that of preparations from adult bullfrogs under basal conditions. Because progressive withdrawal of GABAB-mediated inhibition contributes to the developmental increase in fictive lung ventilation, we then hypothesised that hypoxia reduces respiratory sensitivity to baclofen (selective GABAB-agonist). Experiments were performed on isolated brainstem preparations from pre-metamorphic tadpoles (TK stages IV to XIV); respiratory-related neural activity was recorded from cranial nerves V/VII and X before and 18 h after exposure to hypoxia (0% O2 + 2% CO2; 25 min). Time-control experiments (no hypoxia) were performed. Exposing pre-metamorphic tadpoles to hypoxia did not affect gill burst frequency, but augmented the frequency of fictive lung bursts and the incidence of episodic breathing levels intermediate between pre-metamorphic and adult preparations. Addition of baclofen to the aCSF (0,2 µM - 20 min) reduced lung burst frequency, but the response of hypoxia-exposed brainstems was greater than controls. We conclude that acute hypoxia facilitates development and maturation of the motor command driving air breathing. We propose that a greater number of active rhythmogenic neurons expressing GABAb receptors contributes to this effect.


Asunto(s)
Baclofeno , Respiración , Animales , Baclofeno/farmacología , Larva/fisiología , Pulmón/fisiología , Branquias/fisiología , Hipoxia , Rana catesbeiana
5.
J Exp Biol ; 224(8)2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33914034

RESUMEN

In pre-metamorphic tadpoles, the neural network generating lung ventilation is present but actively inhibited; the mechanisms leading to the onset of air breathing are not well understood. Orexin (ORX) is a hypothalamic neuropeptide that regulates several homeostatic functions, including breathing. While ORX has limited effects on breathing at rest, it potentiates reflexive responses to respiratory stimuli mainly via ORX receptor 1 (OX1R). Here, we tested the hypothesis that OX1Rs facilitate the expression of the motor command associated with air breathing in pre-metamorphic bullfrog tadpoles (Lithobates catesbeianus). To do so, we used an isolated diencephalic brainstem preparation to determine the contributions of OX1Rs to respiratory motor output during baseline breathing, hypercapnia and hypoxia. A selective OX1R antagonist (SB-334867; 5-25 µmol l-1) or agonist (ORX-A; 200 nmol l-1 to 1 µmol l-1) was added to the superfusion media. Experiments were performed under basal conditions (media equilibrated with 98.2% O2 and 1.8% CO2), hypercapnia (5% CO2) or hypoxia (5-7% O2). Under resting conditions gill, but not lung, motor output was enhanced by the OX1R antagonist and ORX-A. Hypercapnia alone did not stimulate respiratory motor output, but its combination with SB-334867 increased lung burst frequency and amplitude, lung burst episodes, and the number of bursts per episode. Hypoxia alone increased lung burst frequency and its combination with SB-334867 enhanced this effect. Inactivation of OX1Rs during hypoxia also increased gill burst amplitude, but not frequency. In contrast with our initial hypothesis, we conclude that ORX neurons provide inhibitory modulation of the CO2 and O2 chemoreflexes in pre-metamorphic tadpoles.


Asunto(s)
Pulmón , Respiración , Animales , Larva , Orexinas , Rana catesbeiana
6.
Am J Physiol Regul Integr Comp Physiol ; 316(3): R281-R297, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30601705

RESUMEN

Amphibian respiratory development involves a dramatic metamorphic transition from gill to lung breathing and coordination of distinct motor outputs. To determine whether the emergence of adult respiratory motor patterns was associated with similarly dramatic changes in motoneuron (MN) properties, we characterized the intrinsic electrical properties of American bullfrog trigeminal MNs innervating respiratory muscles comprising the buccal pump. In premetamorphic tadpoles (TK stages IX-XVIII) and adult frogs, morphometric analyses and whole cell recordings were performed in trigeminal MNs identified by fluorescent retrograde labeling. Based on the amplitude of the depolarizing sag induced by hyperpolarizing voltage steps, two MN subtypes (I and II) were identified in tadpoles and adults. Compared with type II MNs, type I MNs had larger sag amplitudes (suggesting a larger hyperpolarization-activated inward current), greater input resistance, lower rheobase, hyperpolarized action potential threshold, steeper frequency-current relationships, and fast firing rates and received fewer excitatory postsynaptic currents. Postmetamorphosis, type I MNs exhibited similar sag, enhanced postinhibitory rebound, and increased action potential amplitude with a smaller-magnitude fast afterhyperpolarization. Compared with tadpoles, type II MNs from frogs received higher-frequency, larger-amplitude excitatory postsynaptic currents. Input resistance decreased and rheobase increased postmetamorphosis in all MNs, concurrent with increased soma area and hyperpolarized action potential threshold. We suggest that type I MNs are likely recruited in response to smaller, buccal-related synaptic inputs as well as larger lung-related inputs, whereas type II MNs are likely recruited in response to stronger synaptic inputs associated with larger buccal breaths, lung breaths, or nonrespiratory behaviors involving powerful muscle contractions.


Asunto(s)
Branquias/crecimiento & desarrollo , Branquias/fisiología , Pulmón/crecimiento & desarrollo , Pulmón/fisiología , Metamorfosis Biológica/fisiología , Neuronas Motoras/fisiología , Rana catesbeiana/fisiología , Músculos Respiratorios/inervación , Músculos Respiratorios/fisiología , Potenciales de Acción/fisiología , Animales , Mejilla/inervación , Mejilla/fisiología , Potenciales Postsinápticos Excitadores/fisiología , Transmisión Sináptica/fisiología , Nervio Trigémino/fisiología
7.
Exp Physiol ; 104(4): 463-468, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30729595

RESUMEN

NEW FINDINGS: What is the central question of the study? Progesterone is considered a respiratory stimulant drug, but its effect on medullary respiratory neurons are poorly documented. We investigated whether progesterone alters spontaneous activity of neurons in the nucleus of the solitary tract (NTS). What is the main finding and its importance? In NTS neurons, progesterone decreases the action potential firing frequency in response to current injections and the amplitude of excitatory postsynaptic currents. Based on the established neuroprotective effect of progesterone against excitotoxicity resulting from insults, this inhibitory effect is likely to reflect inhibition of ion fluxes. These results are important because they further our understanding of the mechanisms underlying the diversity of respiratory effects of progesterone. ABSTRACT: Progesterone is known to stimulate breathing, but its actions on the respiratory control system have received limited attention. We addressed this issue at the cellular level by testing the hypothesis that progesterone augments excitatory currents at the level of the nucleus tractus solitarii (NTS). Medullary slices from juvenile male rats (14-17 days of age) containing the commissural region of the NTS (NTScom) were incubated with progesterone (1 µm) or vehicle (0.004% DMSO) for 60 min. We performed whole-cell voltage-clamp recordings of spontaneous excitatory postsynaptic currents (EPSCs) in the NTScom and determined membrane properties by applying depolarizing current steps. In comparison to vehicle-treated cells, progesterone exposure attenuates the firing frequency response to current injection and reduces the EPSC amplitude without modifying the EPSC frequency or the basal membrane properties. These data do not support our hypothesis, because they indicate that incubation with progesterone attenuates intrinsic action potential generation and inhibits excitatory synaptic inputs in the NTS. Given that these results are more in line with the protective effect of progesterone against excitotoxicity resulting from various insults, we propose that progesterone acts via inhibition of ionic flux.


Asunto(s)
Neuronas/metabolismo , Progesterona/metabolismo , Núcleo Solitario/metabolismo , Potenciales de Acción/fisiología , Animales , Potenciales Postsinápticos Excitadores/fisiología , Ácido Glutámico/metabolismo , Masculino , Potenciales de la Membrana/fisiología , Técnicas de Placa-Clamp/métodos , Ratas , Ratas Sprague-Dawley , Transmisión Sináptica/fisiología
8.
Exp Physiol ; 103(9): 1192-1199, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29920821

RESUMEN

NEW FINDINGS: What is the central question of the study? Does neonatal stress, in the form of neonatal maternal separation, influence the maturation of microglial density, morphology and neuronal signalling in medullary regions regulating cardiorespiratory function in rat pups? What is the main finding and its importance? Using Iba-1 immunohistochemistry, we show that neonatal maternal separation augments microglial density and the proportion of cells with an amoeboid morphology in the medulla. Although the current understanding of the effect of early life stress on medullary development is relatively limited, these data show that within this area, microglia are affected by neonatal stress. Microglia could therefore be important effectors in cardiorespiratory disorders resulting from maternal separation. ABSTRACT: Neonatal stress has wide-ranging consequences for the developing brain, including the medullary cardiorespiratory network. In rat pups, the reflexive cardiorespiratory inhibition triggered by the presence of liquids near the larynx is augmented by neonatal maternal separation (NMS), especially in males. Sex-specific enhancement of synaptic connectivity by NMS might explain this cardiorespiratory dysfunction. Microglia influence the formation, maturation, activity and elimination of developing synapses, but their role in the wiring of medullary networks is unknown. Owing to their sensitivity to sex hormones and stress hormones, microglial dysfunction could contribute to the abnormal cardiorespiratory phenotype observed in NMS pups. Here, we first used ionized calcium-binding adapter molecule-1 (Iba-1) immunolabelling to compare the density and morphology of microglia in the medulla of male versus female rat pups (14-15 days old) that were either undisturbed or subjected to NMS (3 h day-1 ; postnatal days 3-12). Neonatal maternal separation augmented the density of Iba-1+ cells (caudal region of the NTS), increased the size of the soma and reduced the arborization area (especially in the dorsal motor nucleus of the vagus). Sex-based differences were not observed. Given that the actions of microglia are regulated by neuronal fractalkine (CX3 CL1 ), we then used western blot analysis to compare the expression of CX3 CL1 and its microglial receptor (CX3 CR1 ) in medullary homogenates from control and NMS pups. Although CX3 CR1 expression was 59% greater in males versus females, NMS had no effect on CX3 CL1 /CX3 CR1 signalling. Given that an amoeboid morphology reflects an immature phenotype in developing microglia, NMS could interfere with synaptic pruning via a different mechanism.


Asunto(s)
Animales Recién Nacidos , Bulbo Raquídeo/patología , Microglía/patología , Estrés Psicológico/patología , Animales , Ansiedad de Separación/patología , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/metabolismo , Proteínas de Unión al Calcio/metabolismo , Quimiocina CX3CL1/genética , Quimiocina CX3CL1/metabolismo , Femenino , Masculino , Privación Materna , Proteínas de Microfilamentos/metabolismo , Ratas , Ratas Sprague-Dawley , Caracteres Sexuales , Transducción de Señal , Sinapsis/patología
9.
Exp Physiol ; 103(10): 1403-1411, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29974527

RESUMEN

NEW FINDINGS: What is the central question of the study? Is there a sex-based difference in the incidence of apnoea of prematurity and the success or failure of caffeine therapy in preterm infants? What is the main finding and its importance? Our data show that females received fewer days of caffeine treatment than males. This was most noticeable in infants born between 260/7 and 276/7  weeks of gestational age. These results highlight the importance of considering sex in clinical and basic research investigating the pathophysiology of apnoea of prematurity. ABSTRACT: This retrospective cohort study assessed whether sex influences the occurrence of apnoea of prematurity (AOP) in preterm infants. The analysis included a cohort of 24,387 preterm infants born between the gestational ages (GA) of 240/7 and 336/7  weeks that were admitted to tertiary neonatal care units participating in the Canadian Neonatal Network from January 2011 to December 2015. Of those, 13,983 (57%) were diagnosed with AOP. More females were diagnosed with AOP than males, but the difference in the male/female ratio was marginal (P = 0.058). The majority (89%) of infants diagnosed with AOP received caffeine (89% of males; 89% of females). By using the discontinuation of caffeine therapy as a proxy for the resolution of significant AOP, data analysis showed that females born before 336/7 weeks of GA stopped caffeine treatment earlier than males whether the caffeine was discontinued before 34 or 37 weeks of GA. Consequently, females had fewer days of caffeine therapy than males, especially infants born between 260/7 and 276/7  weeks (P < 0.004), 280/7 and 296/7  weeks (P < 0.03), and 320/7 and 336/7  weeks of GA (P < 0.04). Similar trends were observed when the corrected GA at discontinuation of caffeine was used. Given that AOP is indicative of an immature respiratory system, our data suggest that the maturation of the respiratory system might occur more rapidly in females than males. We conclude that sex needs to be considered in future studies on AOP.


Asunto(s)
Recien Nacido Prematuro/fisiología , Síndromes de la Apnea del Sueño/fisiopatología , Cafeína/uso terapéutico , Canadá , Femenino , Edad Gestacional , Humanos , Recién Nacido , Masculino , Estudios Retrospectivos , Caracteres Sexuales , Síndromes de la Apnea del Sueño/tratamiento farmacológico
10.
Adv Exp Med Biol ; 1071: 75-82, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30357736

RESUMEN

During vertebrate development, the neural networks underlying air-breathing undergo changes in connectivity and functionality, allowing lung ventilation to emerge. Yet, the factors regulating development of these critical homeostatic networks remain unresolved. In amphibians, air-breathing occurs sporadically prior to metamorphosis. However, in tadpoles of Lithobates catesbeianus (American bullfrog), hypoxia stimulates gill and lung ventilation during early development. Because accelerated metamorphosis is a useful strategy to escape deterioration of the milieu, we hypothesized that central hypoxia would elicit long-term expression of the lung motor command for air breathing in pre-metamorphic tadpoles (TK stages VI-XIII). To do this, we recorded respiratory activity from cranial nerves V and VII in isolated brainstems before, during, and up to 2 h after exposure to 15 min of mild (PwO2 range: 114-152 Torr) or moderate (PwO2 range: 38-76 Torr) hypoxia. To test for stage-dependent effects, data were compared between early (VI-IX) and mid (X-XIII) stages. Early stages responded strongly during moderate hypoxia with increased lung burst frequency (167%). Mild and moderate hypoxia increased lung burst frequency during the 2 h re-oxygenation period in early stage brainstems (136%, 497%, respectively), but produced only marginal effects on mid stage brainstems (39%, 31%, respectively). In contrast, hypoxia was not an important factor controlling fictive buccal burst frequency, which drives continuous gill ventilation in tadpoles prior to metamorphosis (all stages showed <25% increase). These preliminary results suggest that central hypoxia elicits long-term increases in lung burst frequency in a severity- and stage-dependent manner.


Asunto(s)
Hipoxia , Larva/fisiología , Pulmón/fisiología , Rana catesbeiana/fisiología , Animales , Metamorfosis Biológica , Respiración
11.
J Physiol ; 595(5): 1637-1655, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27861919

RESUMEN

KEY POINTS: Incomplete development of the neural circuits that control breathing contributes to respiratory disorders in pre-term infants. Manifestations include respiratory instability, prolonged apnoeas and poor ventilatory responses to stimuli. Based on evidence suggesting that omega-3 polyunsaturated fatty acids (n-3 PUFA) improves brain development, we determined whether n-3 PUFA supplementation (via the maternal diet) improves respiratory function in 10-11-day-old rat pups. n-3 PUFA treatment prolonged apnoea duration but augmented the relative pulmonary surface area and the ventilatory response to hypoxia. During hypoxia, the drop in body temperature measured in treated pups was 1 °C less than in controls. n-3 PUFA treatment also reduced microglia cell density in the brainstem. Although heterogeneous, the results obtained in rat pups constitute a proof of concept that n-3 PUFA supplementation can have positive effects on neonatal respiration. This includes a more sustained hypoxic ventilatory response and a decreased respiratory inhibition during laryngeal chemoreflex. ABSTRACT: Most pre-term infants present respiratory instabilities and apnoeas as a result of incomplete development of the neural circuits that control breathing. Because omega-3 polyunsaturated fatty acids (n-3 PUFA) benefit brain development, we hypothesized that n-3 PUFA supplementation (via the maternal diet) improves respiratory function in rat pups. Pups received n-3 PUFA supplementation from an enriched diet (13 g kg-1 of n-3 PUFA) administered to the mother from birth until the experiments were performed (postnatal days 10-11). Controls received a standard diet (0.3 g kg-1 of n-3 PUFA). Breathing was measured in intact pups at rest and during hypoxia (FiO2  = 0.12; 20 min) using whole body plethysmography. The duration of apnoeas induced by stimulating the laryngeal chemoreflex (LCR) was measured under anaesthesia. Lung morphology was compared between groups. Maternal n-3 PUFA supplementation effectively raised n-3 PUFA levels above control levels both in the blood and brainstem of pups. In intact, resting pups, n-3 PUFA increased the frequency and duration of apnoeas, especially in females. During hypoxia, n-3 PUFA supplemented pups hyperventilated 23% more than controls; their anapyrexic response was 1 °C less than controls. In anaesthetized pups, n-3 PUFA shortened the duration of LCR-induced apnoeas by 32%. The relative pulmonary surface area of n-3 PUFA supplemented pups was 12% higher than controls. Although n-3 PUFA supplementation augments apnoeas, there is no clear evidence of deleterious consequences on these pups. Based on the improved lung architecture and responses to respiratory challenges, this neonatal treatment appears to be beneficial to the offspring. However, further experiments are necessary to establish its overall safety.


Asunto(s)
Suplementos Dietéticos , Ácidos Grasos Omega-3/farmacología , Respiración/efectos de los fármacos , Animales , Animales Recién Nacidos , Apnea/fisiopatología , Femenino , Hipoxia/fisiopatología , Laringe/fisiología , Masculino , Intercambio Materno-Fetal , Embarazo , Ratas Sprague-Dawley
12.
J Exp Biol ; 220(Pt 24): 4571-4577, 2017 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-29074702

RESUMEN

Fetal development of the respiratory tract and diaphragm requires strict coordination between genetically controlled signals and mechanical forces produced by the neural network that generates breathing. HOXA5, which is expressed in the mesenchyme of the trachea, lung and diaphragm, and in phrenic motor neurons, is a key transcription factor regulating lung development and function. Consequently, most Hoxa5-/- mutants die at birth from respiratory failure. However, the extensive effect of the null mutation makes it difficult to identify the origins of respiratory dysfunction in newborns. To address the physiological impact of Hoxa5 tissue-specific roles, we used conditional gene targeting with the Dermo1Cre and Olig2Cre mouse lines to produce specific Hoxa5 deletions in the mesenchyme and motor neurons, respectively. Hoxa5 expression in the mesenchyme is critical for trachea development, whereas its expression in phrenic motor neurons is essential for diaphragm formation. Breathing measurements in adult mice with whole-body plethysmography demonstrated that, at rest, only the motor neuron deletion affects respiration, resulting in higher breathing frequency and decreased tidal volume. But subsequent exposure to a moderate hypoxic challenge (FiO2 =0.12; 10 min) revealed that both mutant mice hyperventilate more than controls. Hoxa5flox/flox;Dermo1+/Cre mice showed augmented tidal volume while Hoxa5flox/flox;Olig2+/Cre mice had the largest increase in breathing frequency. No significant differences were observed between medulla-spinal cord preparations from E18.5 control and Hoxa5flox/flox;Olig2+/Cre mouse embryos that could support a role for Hoxa5 in fetal inspiratory motor command. According to our data, Hoxa5 expression in the mesenchyme and phrenic motor neurons controls distinct aspects of respiratory development.


Asunto(s)
Proteínas de Homeodominio/genética , Pulmón/embriología , Fosfoproteínas/genética , Insuficiencia Respiratoria/genética , Animales , Diafragma/fisiopatología , Eliminación de Gen , Marcación de Gen , Proteínas de Homeodominio/fisiología , Técnicas In Vitro , Pulmón/metabolismo , Pulmón/fisiopatología , Ratones , Ratones Noqueados , Mutación , Fosfoproteínas/fisiología , Pletismografía Total , Insuficiencia Respiratoria/fisiopatología , Tráquea/fisiopatología , Factores de Transcripción
13.
Am J Physiol Regul Integr Comp Physiol ; 308(7): R659-67, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25652536

RESUMEN

Human and animal studies on sleep-disordered breathing and respiratory regulation show that the effects of sex hormones are heterogeneous. Because neonatal stress results in sex-specific disruption of the respiratory control in adult rats, we postulate that it might affect respiratory control modulation induced by ovarian steroids in female rats. The hypoxic ventilatory response (HVR) of adult female rats exposed to neonatal maternal separation (NMS) is ∼30% smaller than controls (24), but consequences of NMS on respiratory control in aging female rats are unknown. To address this issue, whole body plethysmography was used to evaluate the impact of NMS on the HVR (12% O2, 20 min) of middle-aged (MA; ∼57 wk old) female rats. Pups subjected to NMS were placed in an incubator 3 h/day for 10 consecutive days (P3 to P12). Controls were undisturbed. To determine whether the effects were related to sexual hormone decline or aging per se, experiments were repeated on bilaterally ovariectomized (OVX) young (∼12 wk old) adult female rats. OVX and MA both reduced the HVR significantly in control rats but had little effect on the HVR of NMS females. OVX (but not aging) reduced the anapyrexic response in both control and NMS animals. These results show that hormonal decline decreases the HVR of control animals, while leaving that of NMS female animals unaffected. This suggests that neonatal stress alters the interaction between sex hormone regulation and the development of body temperature, hormonal, and ventilatory responses to hypoxia.


Asunto(s)
Envejecimiento , Ansiedad de Separación/complicaciones , Regulación de la Temperatura Corporal , Sistema Endocrino/fisiopatología , Hipoxia/complicaciones , Pulmón/fisiopatología , Ventilación Pulmonar , Estrés Psicológico/complicaciones , Factores de Edad , Animales , Animales Recién Nacidos , Ansiedad de Separación/sangre , Ansiedad de Separación/fisiopatología , Peso Corporal , Modelos Animales de Enfermedad , Sistema Endocrino/metabolismo , Estradiol/sangre , Femenino , Hipoxia/fisiopatología , Ovariectomía , Consumo de Oxígeno , Pletismografía Total , Progesterona/sangre , Ratas Sprague-Dawley , Frecuencia Respiratoria , Factores de Riesgo , Síndromes de la Apnea del Sueño/sangre , Síndromes de la Apnea del Sueño/etiología , Síndromes de la Apnea del Sueño/fisiopatología , Estrés Psicológico/sangre , Estrés Psicológico/fisiopatología
14.
J Neurosci ; 33(2): 563-73, 2013 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-23303936

RESUMEN

Recurrent apneas are important causes of hospitalization and morbidity in newborns. Gestational stress (GS) compromises fetal brain development. Maternal stress and anxiety during gestation are linked to respiratory disorders in newborns; however, the mechanisms remain unknown. Here, we tested the hypothesis that repeated activation of the neuroendocrine response to stress during gestation is sufficient to disrupt the development of respiratory control and augment the occurrence of apneas in newborn rats. Pregnant dams were displaced and exposed to predator odor from days 9 to 19 of gestation. Control dams were undisturbed. Experiments were performed on male and female rats aged between 0 and 4 d old. Apnea frequency decreased with age but was consistently higher in stressed pups than controls. At day 4, GS augmented the proportion of apneas with O(2) desaturations by 12%. During acute hypoxia (12% O(2)), the reflexive increase in breathing augmented with age; however, this response was lower in stressed pups. Instability of respiratory rhythm recorded from medullary preparations decreased with age but was higher in stressed pups than controls. GS reduced medullary serotonin (5-HT) levels in newborn pups by 32%. Bath application of 5-HT and injection of 8-OH-DPAT [(±)-8-hydroxy-2-di-(n-propylamino) tetralin hydrobromide; 5-HT(1A) agonist; in vivo] reduced respiratory instability and apneas; these effects were greater in stressed pups than controls. Sex-specific effects were observed. We conclude that activation of the stress response during gestation is sufficient to disrupt respiratory control development and promote pathological apneas in newborn rats. A deficit in medullary 5-HT contributes to these effects.


Asunto(s)
Animales Recién Nacidos/fisiología , Apnea/etiología , Preñez/fisiología , Fenómenos Fisiológicos Respiratorios , Estrés Psicológico/fisiopatología , 8-Hidroxi-2-(di-n-propilamino)tetralin/farmacología , Animales , Apnea/fisiopatología , Corticosterona/sangre , Femenino , Hipercapnia/fisiopatología , Hipoxia/fisiopatología , Masculino , Consumo de Oxígeno/fisiología , Pletismografía , Embarazo , Ratas , Mecánica Respiratoria/efectos de los fármacos , Mecánica Respiratoria/fisiología , Neuronas Serotoninérgicas/fisiología , Serotonina/fisiología , Agonistas de Receptores de Serotonina/farmacología , Caracteres Sexuales , Estrés Psicológico/psicología , Testosterona/sangre
15.
Exp Physiol ; 99(5): 824-34, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24610832

RESUMEN

Neonatal stress disrupts development of homeostatic systems. During adulthood, male rats subjected to neonatal maternal separation (NMS) are hypertensive and show a larger hypoxic ventilatory response (HVR), with greater respiratory instability during sleep. Neonatal stress also affects sex hormone secretion; hypoxia increases circulating testosterone of NMS (but not control) male rats. Given that these effects of NMS are not observed in females, we tested the hypothesis that testosterone elevation is necessary for the stress-related increase of the HVR in adult male rats. Pups subjected to NMS were placed in an incubator for 3 h per day from postnatal day 3 to 12. Control pups remained undisturbed. Rats were reared until adulthood, and the HVR was measured by plethysmography (fractional inspired O2 = 0.12, for 20 min). We used gonadectomy to evaluate the effects of reducing testosterone on the HVR. Gonadectomy had no effect on the HVR of control animals but reduced that of NMS animals below control levels. Immunohistochemistry was used to quantify androgen receptors in brainstem areas involved in the HVR. Androgen receptor expression was generally greater in NMS rats than in control rats; the most significant increase was noted in the caudal region of the nucleus tractus solitarii. We conclude that the abnormal regulation of testosterone is important in stress-related augmentation of the HVR. The greater number of androgen receptors within the brainstem may explain why NMS rats are more sensitive to testosterone withdrawal. Based on the similarities of the cardiorespiratory phenotype of NMS rats and patients suffering from sleep-disordered breathing, these results provide new insight into its pathophysiology, especially sex-based differences in its prevalence.


Asunto(s)
Hipoxia/fisiopatología , Ventilación Pulmonar/efectos de los fármacos , Estrés Psicológico/fisiopatología , Testosterona/farmacología , Animales , Animales Recién Nacidos , Femenino , Masculino , Privación Materna , Consumo de Oxígeno/efectos de los fármacos , Ventilación Pulmonar/fisiología , Ratas Sprague-Dawley , Receptores Androgénicos/biosíntesis , Mecánica Respiratoria/efectos de los fármacos , Mecánica Respiratoria/fisiología , Síndromes de la Apnea del Sueño/fisiopatología , Testosterona/sangre
16.
Exp Physiol ; 98(8): 1312-24, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23603375

RESUMEN

Neonatal stress disrupts the developmental trajectory of homeostatic systems. Adult (8- to 10-week-old) male rats exposed to maternal separation (a form of neonatal stress) display several traits reported in patients suffering from sleep-disordered breathing, including an augmented hypoxic chemoreflex. To understand the mechanisms behind this effect, we tested the hypothesis that neonatal stress augments glutamatergic neurotransmission in three regions involved in respiratory regulation, namely the nucleus of the solitary tract, the paraventricular nucleus of the hypothalamus and the phrenic motor nucleus. Maternal separation was performed for 3 h day(-1) from postnatal day 3 to 12. Control pups were undisturbed. Adult rats were instrumented for intracerebroventricular injection of the AMPA/kainate receptor antagonist CNQX (0-4.3 µm). Using plethysmography, ventilatory activity was measured at rest in awake animals during normoxia (fractional inspired O2 = 0.21) and during acute hypoxia (fractional inspired O2 = 0.12; 20 min). Following vehicle injection, the hypoxic ventilatory response of stressed rats was 35% greater than that of controls. Microinjection of CNQX attenuated the hypoxic ventilatory response, but the effect observed in stressed rats was greater than that in control animals. Autoradiography experiments showed that neonatal stress augments expression of AMPA receptors within the paraventricular nucleus of the hypothalamus and the phrenic motor nucleus. Quantification of brain-derived neurotrophic factor showed that neonatal stress augments brain-derived neurotrophic factor expression only within the paraventricular nucleus. We conclude that neonatal stress augments the hypoxic chemoreflex by increasing the efficacy of glutamatergic synaptic inputs projecting onto key respiratory structures, especially the paraventricular nucleus of the hypothalamus. These data provide new insight into the aetiology of sleep-disordered breathing.


Asunto(s)
Hipoxia/fisiopatología , Receptores AMPA/metabolismo , Reflejo/fisiología , Síndromes de la Apnea del Sueño/fisiopatología , Estrés Fisiológico/fisiología , Transmisión Sináptica/fisiología , Animales , Animales Recién Nacidos , Femenino , Hipotálamo/metabolismo , Hipotálamo/fisiopatología , Hipoxia/metabolismo , Masculino , Núcleo Hipotalámico Paraventricular/metabolismo , Núcleo Hipotalámico Paraventricular/fisiopatología , Nervio Frénico/metabolismo , Nervio Frénico/fisiopatología , Ratas , Ratas Sprague-Dawley , Respiración , Síndromes de la Apnea del Sueño/metabolismo , Núcleo Solitario/metabolismo , Núcleo Solitario/fisiopatología
17.
Front Physiol ; 14: 1183933, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37265841

RESUMEN

CO2 is a fundamental component of living matter. This chemical signal requires close monitoring to ensure proper match between metabolic production and elimination by lung ventilation. Besides ventilatory adjustments, CO2 can also trigger innate behavioral and physiological responses associated with fear and escape but the changes in brain CO2/pH required to induce ventilatory adjustments are generally lower than those evoking fear and escape. However, for patients suffering from panic disorder (PD), the thresholds for CO2-evoked hyperventilation, fear and escape are reduced and the magnitude of those reactions are excessive. To explain these clinical observations, Klein proposed the false suffocation alarm hypothesis which states that many spontaneous panics occur when the brain's suffocation monitor erroneously signals a lack of useful air, thereby maladaptively triggering an evolved suffocation alarm system. After 30 years of basic and clinical research, it is now well established that anomalies in respiratory control (including the CO2 sensing system) are key to PD. Here, we explore how a stress-related affective disorder such as PD can disrupt respiratory control. We discuss rodent models of PD as the concepts emerging from this research has influenced our comprehension of the CO2 chemosensitivity network, especially structure that are not located in the medulla, and how factors such as stress and biological sex modulate its functionality. Thus, elucidating why hormonal fluctuations can lead to excessive responsiveness to CO2 offers a unique opportunity to gain insights into the neuroendocrine mechanisms regulating this key aspect of respiratory control and the pathophysiology of respiratory manifestations of PD.

18.
Neuroscience ; 530: 95-107, 2023 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-37619768

RESUMEN

Pro-opiomelanocortin (POMC)-expressing neurons in the arcuate nucleus of the hypothalamus (ARC) are considered a major site of leptin action. Due to increasing evidence that POMC neurons are highly heterogeneous and indications that the conventional molecular tools to study their functions have important limitations, a reassessment of leptin's effects on definitive POMC neurons is needed. POMC neurons are also expressed in the retrochiasmatic area (RCA), where their function is poorly understood. Furthermore, the response of POMC neurons to leptin in females is largely unknown. Therefore, the present study aimed to determine the differences in leptin responsiveness of POMC neurons in the ARC and the RCA using a mouse model allowing adult-inducible fluorescent labeling. We performed whole-cell patch clamp electrophysiology on 154 POMC neurons from male and female mice. We confirmed and extended the model by which leptin depolarizes POMC neurons, in both the ARC and the RCA. Furthermore, we characterized the electrophysiological properties of an underappreciated subpopulation representing ∼10% of hypothalamic POMC neurons that are inhibited by leptin. We also provide evidence that sex does not appear to be a major determinant of basal properties and leptin responsiveness of POMC neurons, but that females are overall less responsive to leptin compared to males.

19.
J Exp Biol ; 215(Pt 7): 1144-50, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22399659

RESUMEN

The emergence of air breathing during amphibian metamorphosis requires significant changes to the brainstem circuits that generate and regulate breathing. However, the mechanisms controlling this developmental process are unknown. Because corticosterone plays an important role in the neuroendocrine regulation of amphibian metamorphosis, we tested the hypothesis that corticosterone augments fictive air breathing frequency in Xenopus laevis. To do so, we compared the fictive air breathing frequency produced by in vitro brainstem preparations from pre-metamorphic tadpoles and adult frogs before and after 1 h application of corticosterone (100 nmol l(-1)). Fictive breathing measurements related to gill and lung ventilation were recorded extracellularly from cranial nerve rootlets V and X. Corticosterone application had no immediate effect on respiratory-related motor output produced by brainstems from either developmental stage. One hour after corticosterone wash out, fictive lung ventilation frequency was increased whereas gill burst frequency was decreased. This effect was stage specific as it was significant only in preparations from tadpoles. GABA application (0.001-0.5 mmol l(-1)) augmented fictive air breathing in tadpole preparations. However, this effect of GABA was no longer observed following corticosterone treatment. An increase in circulating corticosterone is one of the endocrine processes that orchestrate central nervous system remodelling during metamorphosis. The age-specific effects of corticosterone application indicate that this hormone can act as an important regulator of respiratory control development in Xenopus tadpoles. Concurrent changes in GABAergic neurotransmission probably contribute to this maturation process, leading to the emergence of air breathing in this species.


Asunto(s)
Aire , Tronco Encefálico/efectos de los fármacos , Tronco Encefálico/fisiología , Corticosterona/farmacología , Respiración/efectos de los fármacos , Xenopus laevis/fisiología , Envejecimiento/efectos de los fármacos , Animales , Tronco Encefálico/crecimiento & desarrollo , Corticosterona/administración & dosificación , Técnicas In Vitro , Larva/efectos de los fármacos , Larva/fisiología , Pulmón/efectos de los fármacos , Pulmón/fisiología , Actividad Motora/efectos de los fármacos , Factores de Tiempo , Nervio Trigémino/efectos de los fármacos , Nervio Trigémino/fisiología , Nervio Vago/efectos de los fármacos , Nervio Vago/fisiología , Ácido gamma-Aminobutírico/farmacología
20.
Adv Exp Med Biol ; 758: 179-83, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23080160

RESUMEN

Baroreflex is involved in the regulation of arterial blood pressure (BP). An increase in BP activates vagal inhibitory pathways to decrease heart rate; a concomitant decrease in sympathetic discharge reduces vascular resistance. Both responses reduce BP towards normal value. Conversely, a decrease in BP produces opposite effects to increase heart rate and vascular resistance.


Asunto(s)
Barorreflejo/fisiología , Hipoxia/fisiopatología , Animales , Animales Recién Nacidos , Presión Sanguínea , Frecuencia Cardíaca , Masculino , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA