Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Children (Basel) ; 10(6)2023 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-37371150

RESUMEN

OBJECTIVE: To test the potential utility of applying machine learning methods to regional cerebral (rcSO2) and peripheral oxygen saturation (SpO2) signals to detect brain injury in extremely preterm infants. STUDY DESIGN: A subset of infants enrolled in the Management of Hypotension in Preterm infants (HIP) trial were analysed (n = 46). All eligible infants were <28 weeks' gestational age and had continuous rcSO2 measurements performed over the first 72 h and cranial ultrasounds performed during the first week after birth. SpO2 data were available for 32 infants. The rcSO2 and SpO2 signals were preprocessed, and prolonged relative desaturations (PRDs; data-driven desaturation in the 2-to-15-min range) were extracted. Numerous quantitative features were extracted from the biosignals before and after the exclusion of the PRDs within the signals. PRDs were also evaluated as a stand-alone feature. A machine learning model was used to detect brain injury (intraventricular haemorrhage-IVH grade II-IV) using a leave-one-out cross-validation approach. RESULTS: The area under the receiver operating characteristic curve (AUC) for the PRD rcSO2 was 0.846 (95% CI: 0.720-0.948), outperforming the rcSO2 threshold approach (AUC 0.593 95% CI 0.399-0.775). Neither the clinical model nor any of the SpO2 models were significantly associated with brain injury. CONCLUSION: There was a significant association between the data-driven definition of PRDs in rcSO2 and brain injury. Automated analysis of PRDs of the cerebral NIRS signal in extremely preterm infants may aid in better prediction of IVH compared with a threshold-based approach. Further investigation of the definition of the extracted PRDs and an understanding of the physiology underlying these events are required.

3.
Annu Int Conf IEEE Eng Med Biol Soc ; 2021: 1010-1013, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34891459

RESUMEN

Preterm infants are at high risk of developing brain injury in the first days of life as a consequence of poor cerebral oxygen delivery. Near-infrared spectroscopy (NIRS) is an established technology developed to monitor regional tissue oxygenation. Detailed waveform analysis of the cerebral NIRS signal could improve the clinical utility of this method in accurately predicting brain injury. Frequent transient cerebral oxygen desaturations are commonly observed in extremely preterm infants, yet their clinical significance remains unclear. The aim of this study was to examine and compare the performance of two distinct approaches in isolating and extracting transient deflections within NIRS signals. We optimized three different simultaneous low-pass filtering and total variation denoising (LPF-TVD) methods and compared their performance with a recently proposed method that uses singular-spectrum analysis and the discrete cosine transform (SSA-DCT). Parameters for the LPF-TVD methods were optimized over a grid search using synthetic NIRS-like signals. The SSA-DCT method was modified with a post-processing procedure to increase sparsity in the extracted components. Our analysis, using a synthetic NIRS-like dataset, showed that a LPF-TVD method outperformed the modified SSA-DCT method: median mean-squared error of 0.97 (95% CI: 0.86 to 1.07) was lower for the LPF-TVD method compared to the modified SSA-DCT method of 1.48 (95% CI: 1.33 to 1.63), P<0.001. The dual low-pass filter and total variation denoising methods are considerably more computational efficient, by 3 to 4 orders of magnitude, than the SSA-DCT method. More research is needed to examine the efficacy of these methods in extracting oxygen desaturation in real NIRS signals.Clinical relevance- Early and precise identification of abnormal brain oxygenation in premature infants would enable clinicians to employ therapeutic strategies that seek to prevent brain injury and long-term morbidity in this vulnerable population.


Asunto(s)
Lesiones Encefálicas , Espectroscopía Infrarroja Corta , Encéfalo/diagnóstico por imagen , Humanos , Recien Nacido Extremadamente Prematuro , Recién Nacido , Monitoreo Fisiológico
4.
Am J Physiol Regul Integr Comp Physiol ; 321(6): R879-R902, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34612068

RESUMEN

Toll-like receptors (TLRs) are crucial transmembrane receptors that form part of the innate immune response. They play a role in the recognition of various microorganisms and their elimination from the host. TLRs have been proposed as vital immunomodulators in the regulation of multiple neonatal stressors that extend beyond infection such as oxidative stress and pain. The immune system is immature at birth and takes some time to become fully established. As such, babies are especially vulnerable to sepsis at this early stage of life. Findings suggest a gestational age-dependent increase in TLR expression. TLRs engage with accessory and adaptor proteins to facilitate recognition of pathogens and their activation of the receptor. TLRs are generally upregulated during infection and promote the transcription and release of proinflammatory cytokines. Several studies report that TLRs are epigenetically modulated by chromatin changes and promoter methylation upon bacterial infection that have long-term influences on immune responses. TLR activation is reported to modulate cardiorespiratory responses during infection and may play a key role in driving homeostatic instability observed during sepsis. Although complex, TLR signaling and downstream pathways are potential therapeutic targets in the treatment of neonatal diseases. By reviewing the expression and function of key Toll-like receptors, we aim to provide an important framework to understand the functional role of these receptors in response to stress and infection in premature infants.


Asunto(s)
Antiinflamatorios/uso terapéutico , Sistema Inmunológico/efectos de los fármacos , Mediadores de Inflamación/antagonistas & inhibidores , Inflamación/tratamiento farmacológico , Sepsis Neonatal/tratamiento farmacológico , Receptores Toll-Like/efectos de los fármacos , Factores de Edad , Animales , Antiinflamatorios/efectos adversos , Desarrollo Infantil , Ensamble y Desensamble de Cromatina , Epigénesis Genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Inmunidad Innata/efectos de los fármacos , Recién Nacido , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Mediadores de Inflamación/metabolismo , Masculino , Terapia Molecular Dirigida , Sepsis Neonatal/genética , Sepsis Neonatal/inmunología , Sepsis Neonatal/metabolismo , Factores Sexuales , Transducción de Señal , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo
6.
Exp Physiol ; 105(1): 17-43, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31626357

RESUMEN

NEW FINDINGS: What is the topic of this review? We review the influence of prematurity on the cardiorespiratory system and examine the common sequel of alterations in oxygen tension, and immune activation in preterm infants. What advances does it highlight? The review highlights neonatal animal models of intermittent hypoxia, hyperoxia and infection that contribute to our understanding of the effect of stress on neurodevelopment and cardiorespiratory homeostasis. We also focus on some of the important physiological pathways that have a modulatory role on the cardiorespiratory system in early life. ABSTRACT: Preterm birth is one of the leading causes of neonatal mortality. Babies that survive early-life stress associated with immaturity have significant prevailing short- and long-term morbidities. Oxygen dysregulation in the first few days and weeks after birth is a primary concern as the cardiorespiratory system slowly adjusts to extrauterine life. Infants exposed to rapid alterations in oxygen tension, including exposures to hypoxia and hyperoxia, have altered redox balance and active immune signalling, leading to altered stress responses that impinge on neurodevelopment and cardiorespiratory homeostasis. In this review, we explore the clinical challenges posed by preterm birth, followed by an examination of the literature on animal models of oxygen dysregulation and immune activation in the context of early-life stress.


Asunto(s)
Sistema Cardiovascular/fisiopatología , Nacimiento Prematuro/fisiopatología , Sistema Respiratorio/fisiopatología , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Humanos , Hiperoxia/patología , Hipoxia/patología , Lactante , Recien Nacido Prematuro , Infecciones/patología , Oxígeno , Estrés Fisiológico
11.
J Physiol ; 595(21): 6653-6672, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28952155

RESUMEN

KEY POINTS: Respiratory failure is a leading cause of mortality in Duchenne muscular dystrophy (DMD), but little is known about the control of breathing in DMD and animal models. We show that young (8 weeks of age) mdx mice hypoventilate during basal breathing due to reduced tidal volume. Basal CO2 production is equivalent in wild-type and mdx mice. We show that carotid bodies from mdx mice have blunted responses to hyperoxia, revealing hypoactivity in normoxia. However, carotid body, ventilatory and metabolic responses to hypoxia are equivalent in wild-type and mdx mice. Our study revealed profound muscle weakness and muscle fibre remodelling in young mdx diaphragm, suggesting severe mechanical disadvantage in mdx mice at an early age. Our novel finding of potentiated neural motor drive to breathe in mdx mice during maximal chemoactivation suggests compensatory neuroplasticity enhancing respiratory motor output to the diaphragm and probably other accessory muscles. ABSTRACT: Patients with Duchenne muscular dystrophy (DMD) hypoventilate with consequential arterial blood gas derangement relevant to disease progression. Whereas deficits in DMD diaphragm are recognized, there is a paucity of knowledge in respect of the neural control of breathing in dystrophinopathies. We sought to perform an analysis of respiratory control in a model of DMD, the mdx mouse. In 8-week-old male wild-type and mdx mice, ventilation and metabolism, carotid body afferent activity, diaphragm muscle force-generating capacity, and muscle fibre size, distribution and centronucleation were determined. Diaphragm EMG activity and responsiveness to chemostimulation was determined. During normoxia, mdx mice hypoventilated, owing to a reduction in tidal volume. Basal CO2 production was not different between wild-type and mdx mice. Carotid sinus nerve responses to hyperoxia were blunted in mdx, suggesting hypoactivity. However, carotid body, ventilatory and metabolic responses to hypoxia were equivalent in wild-type and mdx mice. Diaphragm force was severely depressed in mdx mice, with evidence of fibre remodelling and damage. Diaphragm EMG responses to chemoactivation were enhanced in mdx mice. We conclude that there is evidence of chronic hypoventilation in young mdx mice. Diaphragm dysfunction confers mechanical deficiency in mdx resulting in impaired capacity to generate normal tidal volume at rest and decreased absolute ventilation during chemoactivation. Enhanced mdx diaphragm EMG responsiveness suggests compensatory neuroplasticity facilitating respiratory motor output, which may extend to accessory muscles of breathing. Our results may have relevance to emerging treatments for human DMD aiming to preserve ventilatory capacity.


Asunto(s)
Cuerpo Carotídeo/fisiopatología , Diafragma/fisiopatología , Distrofia Muscular de Duchenne/fisiopatología , Respiración , Animales , Dióxido de Carbono/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Oxígeno/metabolismo , Ventilación Pulmonar
12.
Can J Physiol Pharmacol ; 95(12): 1488-1492, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28881142

RESUMEN

Sudden infant death syndrome (SIDS) remains the leading cause of infant mortality beyond the neonatal period. An increase in body temperature as a result of high environmental temperature, overwrapping of infants, and (or) infection are associated with SIDS. Endotoxins such as lipopolysaccharide (LPS) and heat stress may perturb cardiorespiratory function and thermoregulation. Although LPS-mediated body temperature and cytokine responses are well documented in older animals, the capacity of LPS to induce fever and cytokine response in young rats remains unclear. Therefore, we sought to investigate the acute effects of LPS on body temperature and cytokine concentrations in rat pups. Postnatal day 7 rat pups were divided into 3 groups: Group 1, rats were administered LPS intraperitoneally (200 µg/kg); Group 2, rats received saline at volume equal to that administered in the LPS group; Group 3, rats received no treatment. Pups were placed in custom-made chambers maintained at ambient temperature of 33 °C. Body surface temperature was continuously monitored for 4 h. Thereafter, the rats were euthanized and serum was collected for cytokine analysis. We demonstrate that LPS treatment increased MIP-1α, IL-10, MCP-1, IP-10, fractalkine, and TNF-α with no concurrent rise in body surface temperature. Although neonatal rats produced an array of cytokines in response to LPS, there was no evidence of fever.


Asunto(s)
Citocinas/metabolismo , Lipopolisacáridos/farmacología , Animales , Temperatura Corporal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Ambiente Controlado , Estadios del Ciclo de Vida , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
13.
Am J Physiol Regul Integr Comp Physiol ; 311(6): R1113-R1124, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27733384

RESUMEN

Maternal cigarette smoke (CS) exposure exhibits a strong epidemiological association with Sudden Infant Death Syndrome, but other environmental stressors, including infection, hyperthermia, and hypoxia, have also been postulated as important risk factors. This study examines whether maternal CS exposure causes maladaptations within homeostatic control networks by influencing the response to lipopolysaccharide, heat stress, and/or hypoxia in neonatal rats. Pregnant dams were exposed to CS or parallel sham treatments daily for the length of gestation. Offspring were studied at postnatal days 6-8 at ambient temperatures (Ta) of 33°C or 38°C. Within each group, rats were allocated to control, saline, or LPS (200 µg/kg) treatments. Cardiorespiratory patterns were examined using head-out plethysmography and ECG surface electrodes during normoxia and hypoxia (10% O2). Serum cytokine concentrations were quantified from samples taken at the end of each experiment. Our results suggest maternal CS exposure does not alter minute ventilation (V̇e) or heart rate (HR) response to infection or high temperature, but independently increases apnea frequency. CS also primes the inflammatory system to elicit a stronger cytokine response to bacterial insult. High Ta independently depresses V̇e but augments the hypoxia-induced increase in V̇e Moreover, higher Ta increases HR during normoxia and hypoxia, and in the presence of an immune challenge, increases HR during normoxia, and reduces the increase normally associated with hypoxia. Thus, while most environmental risk factors increase the burden on the cardiorespiratory system in early life, hyperthermia and infection blunt the normal HR response to hypoxia, and gestational CS independently destabilizes breathing by increasing apneas.


Asunto(s)
Respuesta al Choque Térmico , Hipoxia/fisiopatología , Inflamación/fisiopatología , Exposición Materna/efectos adversos , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Contaminación por Humo de Tabaco/efectos adversos , Animales , Animales Recién Nacidos , Citocinas/sangre , Femenino , Frecuencia Cardíaca , Inflamación/sangre , Inflamación/inducido químicamente , Lipopolisacáridos , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/sangre , Efectos Tardíos de la Exposición Prenatal/etiología , Ventilación Pulmonar , Ratas Sprague-Dawley
14.
Front Physiol ; 7: 276, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27462274

RESUMEN

Alterations to the supply of oxygen during early life presents a profound stressor to physiological systems with aberrant remodeling that is often long-lasting. Chronic intermittent hypoxia (CIH) is a feature of apnea of prematurity, chronic lung disease, and sleep apnea. CIH affects respiratory control but there is a dearth of information concerning the effects of CIH on respiratory muscles, including the diaphragm-the major pump muscle of breathing. We investigated the effects of exposure to gestational CIH (gCIH) and postnatal CIH (pCIH) on diaphragm muscle function in male and female rats. CIH consisted of exposure in environmental chambers to 90 s of hypoxia reaching 5% O2 at nadir, once every 5 min, 8 h a day. Exposure to gCIH started within 24 h of identification of a copulation plug and continued until day 20 of gestation; animals were studied on postnatal day 22 or 42. For pCIH, pups were born in normoxia and within 24 h of delivery were exposed with dams to CIH for 3 weeks; animals were studied on postnatal day 22 or 42. Sham groups were exposed to normoxia in parallel. Following gas exposures, diaphragm muscle contractile, and endurance properties were examined ex vivo. Neither gCIH nor pCIH exposure had effects on diaphragm muscle force-generating capacity or endurance in either sex. Similarly, early life exposure to CIH did not affect muscle tolerance of severe hypoxic stress determined ex vivo. The findings contrast with our recent observation of upper airway dilator muscle weakness following exposure to pCIH. Thus, the present study suggests a relative resilience to hypoxic stress in diaphragm muscle. Co-ordinated activity of thoracic pump and upper airway dilator muscles is required for optimal control of upper airway caliber. A mismatch in the force-generating capacity of the complementary muscle groups could have adverse consequences for the control of airway patency and respiratory homeostasis.

15.
Front Physiol ; 7: 69, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26973537

RESUMEN

Intermittent hypoxia is a feature of apnea of prematurity (AOP), chronic lung disease, and sleep apnea. Despite the clinical relevance, the long-term effects of hypoxic exposure in early life on respiratory control are not well defined. We recently reported that exposure to chronic intermittent hypoxia (CIH) during postnatal development (pCIH) causes upper airway muscle weakness in both sexes, which persists for several weeks. We sought to examine if there are persistent sex-dependent effects of pCIH on respiratory muscle function into adulthood and/or increased susceptibility to re-exposure to CIH in adulthood in animals previously exposed to CIH during postnatal development. We hypothesized that pCIH would cause long-lasting muscle impairment and increased susceptibility to subsequent hypoxia. Within 24 h of delivery, pups and their respective dams were exposed to CIH: 90 s of hypoxia reaching 5% O2 at nadir; once every 5 min, 8 h per day for 3 weeks. Sham groups were exposed to normoxia in parallel. Three groups were studied: sham; pCIH; and pCIH combined with adult CIH (p+aCIH), where a subset of the pCIH-exposed pups were re-exposed to the same CIH paradigm beginning at 13 weeks. Following gas exposures, sternohyoid and diaphragm muscle isometric contractile and endurance properties were examined ex vivo. There was no apparent lasting effect of pCIH on respiratory muscle function in adults. However, in both males and females, re-exposure to CIH in adulthood in pCIH-exposed animals caused sternohyoid (but not diaphragm) weakness. Exposure to this paradigm of CIH in adulthood alone had no effect on muscle function. Persistent susceptibility in pCIH-exposed airway dilator muscle to subsequent hypoxic insult may have implications for the control of airway patency in adult humans exposed to intermittent hypoxic stress during early life.

16.
Physiol Rep ; 4(2)2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26811056

RESUMEN

Sudden infant death syndrome (SIDS) is one of the most common causes of postneonatal infant mortality in the developed world. An insufficient cardiorespiratory response to multiple environmental stressors (such as prone sleeping positioning, overwrapping, and infection), during a critical period of development in a vulnerable infant, may result in SIDS. However, the effect of multiple risk factors on cardiorespiratory responses has rarely been tested experimentally. Therefore, this study aimed to quantify the independent and possible interactive effects of infection, hyperthermia, and hypoxia on cardiorespiratory control in rats during the neonatal period. We hypothesized that lipopolysaccharide (LPS) administration will negatively impact cardiorespiratory responses to increased ambient temperature and hypoxia in neonatal rats. Sprague-Dawley neonatal rat pups were studied at postnatal day 6-8. Rats were examined at an ambient temperature of 33°C or 38°C. Within each group, rats were allocated to control, saline, or LPS (200 µg/kg) treatments. Cardiorespiratory and thermal responses were recorded and analyzed before, during, and after a hypoxic exposure (10% O2). Serum samples were taken at the end of each experiment to measure cytokine concentrations. LPS significantly increased cytokine concentrations (such as TNFα, IL-1ß, MCP-1, and IL-10) compared to control. Our results do not support a three-way interaction between experimental factors on cardiorespiratory control. However, independently, heat stress decreased minute ventilation during normoxia and increased the hypoxic ventilatory response. Furthermore, LPS decreased hypoxia-induced tachycardia. Herein, we provide an extensive serum cytokine profile under various experimental conditions and new evidence that neonatal cardiorespiratory responses are adversely affected by dual interactions of environmental stress factors.


Asunto(s)
Citocinas/biosíntesis , Fiebre/complicaciones , Hipoxia/complicaciones , Infecciones/complicaciones , Muerte Súbita del Lactante/etiología , Animales , Animales Recién Nacidos , Citocinas/análisis , Modelos Animales de Enfermedad , Fiebre/fisiopatología , Frecuencia Cardíaca/fisiología , Humanos , Hipoxia/fisiopatología , Recién Nacido , Infecciones/fisiopatología , Lipopolisacáridos/toxicidad , Reacción en Cadena de la Polimerasa Multiplex , Pletismografía , Ventilación Pulmonar/fisiología , Ratas , Ratas Sprague-Dawley
17.
Exp Physiol ; 100(8): 947-66, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26096367

RESUMEN

NEW FINDINGS: What is the central question of this study? Chronic intermittent hypoxia (CIH) is a dominant feature of respiratory control disorders, which are common. We sought to examine the effects of exposure to CIH during neonatal development on respiratory muscle form and function in male and female rats. What is the main finding and its importance? Exposure to CIH during neonatal development caused sternohyoid muscle weakness in both sexes; an effect that persisted into young adult life upon return to normoxia. Upper airway dilator muscle dysfunction in vivo could predispose to airway collapse, leading to impaired respiratory homeostasis. Chronic intermittent hypoxia (CIH) is a feature of sleep-disordered breathing, which is very common. Exposure to CIH is associated with aberrant plasticity in the respiratory control system including the final effector organs, the striated muscles of breathing. We reasoned that developmental age and sex are key factors determining the functional response of respiratory muscle to CIH. We tested the hypothesis that exposure to CIH causes persistent impairment of sternohyoid muscle function due to oxidative stress and that males are more susceptible to CIH-induced muscle impairment than females. Wistar rat litters (with respective dams) were exposed to intermittent hypoxia for 12 cycles per hour, 8 h per day for 3 weeks from the first day of life [postnatal day (P) 0]. Sham experiments were run in parallel. Half of each litter was studied on P22; the other half was returned to normoxia and studied on P42. Functional properties of the sternohyoid muscle were determined ex vivo. Exposure to CIH significantly decreased sternohyoid muscle force in both sexes; an effect that persisted into young adult life. Chronic intermittent hypoxia had no effect on sternohyoid muscle endurance. Chronic intermittent hypoxia did not affect sternohyoid myosin fibre type, succinate dehydrogenase or glycerol-3-phosphate dehydrogenase activities, or protein free thiol and carbonyl content. Muscles exposed to CIH had smaller cross-sectional areas, consistent with the observation of muscle weakness. In human infants with disordered breathing, CIH-induced upper airway dilator muscle weakness could increase the propensity for airway narrowing or collapse, which could serve to perpetuate impaired respiratory homeostasis.


Asunto(s)
Hipoxia/complicaciones , Hipoxia/patología , Debilidad Muscular/etiología , Debilidad Muscular/patología , Músculos Respiratorios/crecimiento & desarrollo , Músculos Respiratorios/patología , Factores de Edad , Animales , Animales Recién Nacidos , Enfermedad Crónica , Femenino , Masculino , Ratas , Ratas Wistar
18.
Front Physiol ; 6: 15, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25688214

RESUMEN

Chronic intermittent hypoxia (CIH) causes upper airway muscle dysfunction. We hypothesized that the superoxide generating NADPH oxidase (NOX) is upregulated in CIH-exposed muscle causing oxidative stress. Adult male Wistar rats were exposed to intermittent hypoxia (5% O2 at the nadir for 90 s followed by 210 s of normoxia), for 8 h per day for 14 days. The effect of CIH exposure on the expression of NOX subunits, total myosin and 4-hydroxynonenal (4-HNE) protein adducts in sternohyoid muscle was determined by western blotting and densitometry. Sternohyoid protein free thiol and carbonyl group contents were determined by 1D electrophoresis using specific fluorophore probes. Aconitase and glutathione reductase activities were measured as indices of oxidative stress. HIF-1α content and key oxidative and glycolytic enzyme activities were determined. Contractile properties of sternohyoid muscle were determined ex vivo in the absence and presence of apocynin (putative NOX inhibitor). We observed an increase in NOX 2 and p47 phox expression in CIH-exposed sternohyoid muscle with decreased aconitase and glutathione reductase activities. There was no evidence, however, of increased lipid peroxidation or protein oxidation in CIH-exposed muscle. CIH exposure did not affect sternohyoid HIF-1α content or aldolase, lactate dehydrogenase, or glyceraldehyde-3-phosphate dehydrogenase activities. Citrate synthase activity was also unaffected by CIH exposure. Apocynin significantly increased sternohyoid force and power. We conclude that CIH exposure upregulates NOX expression in rat sternohyoid muscle with concomitant modest oxidative stress but it does not result in a HIF-1α-dependent increase in glycolytic enzyme activity. Constitutive NOX activity decreases sternohyoid force and power. Our results implicate NOX-dependent reactive oxygen species in CIH-induced upper airway muscle dysfunction which likely relates to redox modulation of key regulatory proteins in excitation-contraction coupling.

19.
Prog Brain Res ; 212: 237-51, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25194201

RESUMEN

Nitric oxide (NO) is an important modulator of striated muscle function. Nitric oxide synthase (NOS) expression and activity is altered by hypoxia and NO is implicated in respiratory muscle remodeling following chronic sustained hypoxia. We sought to determine if NO is implicated in upper airway dilator muscle adaptation to chronic intermittent hypoxia (CIH). Thirty-two adult male Wistar rats (284±13, mean±SD) were exposed to alternating bouts of hypoxia (90 s; 5% O2 at the nadir) and normoxia (210 s; 21% O2) for 12 cycles per hour, 8h/day for 3 weeks. Sham animals were exposed to normoxia in parallel. Half of the animals in both groups received the nNOS inhibitor-L-NNA (2mM) in the drinking water throughout the study (N=8 for all groups). Sternohyoid (pharyngeal dilator) muscle contractile and endurance properties were determined ex vivo. Sternohyoid muscle myosin heavy chain (MHC) isoform composition and cross-sectional area was determined by fluorescence microscopy. Chronic nNOS blockade did not alter sternohyoid muscle peak force or force-frequency relationship in sham or CIH-treated animals. In contrast, chronic nNOS blockade significantly decreased sternohyoid muscle endurance with equivalent effects in sham and CIH-treated rats. Our results suggest that NO is an important modulator of sternohyoid muscle endurance. However, our data provide no evidence to suggest that NO is implicated in upper airway muscle adaptation to CIH.


Asunto(s)
Adaptación Fisiológica/fisiología , Hipoxia/fisiopatología , Óxido Nítrico Sintasa de Tipo I/antagonistas & inhibidores , Músculos Faríngeos/fisiología , Animales , Enfermedad Crónica , Masculino , Contracción Muscular/fisiología , Cadenas Pesadas de Miosina/metabolismo , Ratas , Ratas Wistar
20.
Respir Physiol Neurobiol ; 201: 75-83, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25017785

RESUMEN

In unrestrained whole body plethysmography, tidal volume is commonly determined using the barometric method, which assumes that temperature and humidity changes (the 'barometric component') are solely responsible for breathing-related chamber pressure fluctuations. However, in small animals chamber pressure is also influenced by a 'mechanical component' dependent on airway resistance and airflow. We devised a novel 'mechanical lung' capable of simulating neonatal mouse breathing in the absence of temperature or humidity changes. Using this device, we confirm that the chamber pressure fluctuations produced by breathing of neonatal mice are dominated by the mechanical component, precluding direct quantitative assessment of tidal volume. Recognizing the importance of airway resistance to the chamber pressure signal and the ability of our device to simulate neonatal breathing at different frequencies and tidal volumes, we invented a novel in vivo, non-invasive method for conscious airway resistance and ventilation estimation (CARVE) in neonatal rodents. This technique will allow evaluation of developmental, pathological and pharmaceutical effects on airway resistance.


Asunto(s)
Resistencia de las Vías Respiratorias/fisiología , Estado de Conciencia/fisiología , Mediciones del Volumen Pulmonar/métodos , Pletismografía , Mecánica Respiratoria , Animales , Animales Recién Nacidos , Temperatura Corporal , Simulación por Computador , Ratones , Modelos Biológicos , Estimulación Física , Volumen de Ventilación Pulmonar , Ventiladores Mecánicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA