Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 423
Filtrar
1.
Respir Physiol Neurobiol ; 297: 103835, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34971764

RESUMEN

Allergic asthma affects both the respiratory function and central nervous system. Communication between the amygdala and respiratory control system is critical for regulating breathing function. To date, no study provides the effect of allergic inflammation on amygdala-respiration coupling. Here, we simultaneously recorded respiration and local field potentials of the amygdala during awake immobility in a rat model of allergic asthma. A decreased synchrony was found between amygdala and respiration in asthmatic rats. Allergen also reduced the modulatory effect of the respiration phase on amygdala power at delta, theta and gamma2 (80-120 Hz) frequencies. Moreover, in the animal model of allergic asthma, delta and theta oscillations strongly coordinate local gamma2 activity in the amygdala. These findings suggest that allergen can induce brain alterations and therefore shed light on future works to address how disruption of amygdala-respiration coupling contributes to respiratory dysfunction in allergic asthma.


Asunto(s)
Amígdala del Cerebelo/fisiopatología , Asma/fisiopatología , Ondas Encefálicas/fisiología , Hipersensibilidad/fisiopatología , Inflamación/fisiopatología , Centro Respiratorio/fisiopatología , Frecuencia Respiratoria/fisiología , Alérgenos/farmacología , Animales , Modelos Animales de Enfermedad , Ratas
2.
Respir Physiol Neurobiol ; 297: 103829, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34921999

RESUMEN

Perinatal inflammation triggers breathing disturbances early in life and affects the respiratory adaptations to challenging conditions, including the generation of amplitude long-term facilitation (LTF) by acute intermittent hypoxia (AIH). Some of these effects can be avoided by anti-inflammatory treatments like minocycline. Since little is known about the effects of perinatal inflammation on the inspiratory rhythm generator, located in the preBötzinger complex (preBötC), we tested the impact of acute lipopolysaccharide (LPS) systemic administration (sLPS), as well as gestational LPS (gLPS) and gestational chronic IH (gCIH), on respiratory rhythm generation and its long-term response to AIH in a brainstem slice preparation from neonatal mice. We also evaluated whether acute minocycline administration could influence these effects. We found that perinatal inflammation induced by sLPS or gLPS, as well as gCIH, modulate the frequency, signal-to-noise ratio and/or amplitude (and their regularity) of the respiratory rhythm recorded from the preBötC in the brainstem slice. Moreover, all these perinatal conditions inhibited frequency LTF and amplitude long-term depression (LTD); gCIH even induced frequency LTD of the respiratory rhythm after AIH. Some of these alterations were not observed in slices pre-treated in vitro with minocycline, when compared with slices obtained from naïve pups, suggesting that ongoing inflammatory conditions affect respiratory rhythm generation and its plasticity. Thus, it is likely that alterations in the inspiratory rhythm generator and its adaptive responses could contribute to the respiratory disturbances observed in neonates that suffered from perinatal inflammatory challenges.


Asunto(s)
Antiinflamatorios/farmacología , Generadores de Patrones Centrales/fisiopatología , Hipoxia/fisiopatología , Enfermedades del Recién Nacido/fisiopatología , Inflamación/tratamiento farmacológico , Inflamación/fisiopatología , Minociclina/farmacología , Plasticidad Neuronal/fisiología , Centro Respiratorio/fisiopatología , Frecuencia Respiratoria/fisiología , Animales , Animales Recién Nacidos , Antiinflamatorios/administración & dosificación , Modelos Animales de Enfermedad , Humanos , Recién Nacido , Enfermedades del Recién Nacido/tratamiento farmacológico , Inflamación/inducido químicamente , Minociclina/administración & dosificación
3.
Physiol Rep ; 9(21): e15109, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34755471

RESUMEN

Neural circuits at the brainstem involved in the central generation of the motor patterns of respiration and cardiorespiratory chemoreflexes organize as cell assemblies connected by chemical and electrical synapses. However, the role played by the electrical connectivity mainly mediated by connexin36 (Cx36), which expression reaches peak value during the postnatal period, is still unknown. To address this issue, we analyzed here the respiratory phenotype of a mouse strain devoid constitutively of Cx36 at P14. Male Cx36-knockout mice at rest showed respiratory instability of variable degree, including a periodic Cheyne-Stokes breathing. Moreover, mice lacking Cx36 exhibited exacerbated chemoreflexes to normoxic and hypoxic hypercapnia characterized by a stronger inspiratory/expiratory coupling due to an increased sensitivity to CO2 . Deletion of Cx36 also impaired the generation of the recurrent episodes of transient bradycardia (ETBs) evoked during hypercapnic chemoreflexes; these EBTs constituted a powerful mechanism of cardiorespiratory coupling capable of improving alveolar gaseous exchange under hypoxic hypercapnia conditions. Approximately half of the homo- and heterozygous Cx36KO, but none WT, mice succumbed by respiratory arrest when submitted to hypoxia-hypercapnia, the principal exogenous stressor causing sudden infant death syndrome (SIDS). The early suppression of EBTs, which worsened arterial O2  saturation, and the generation of a paroxysmal generalized clonic-tonic activity, which provoked the transition from eupneic to gasping respiration, were the critical events causing sudden death in the Cx36KO mice. These results indicate that Cx36 expression plays a pivotal role in respiratory control, cardiorespiratory coordination, and protection against SIDS at the postnatal period.


Asunto(s)
Conexinas/genética , Respiración , Muerte Súbita del Lactante/genética , Animales , Conexinas/metabolismo , Femenino , Eliminación de Gen , Humanos , Lactante , Masculino , Ratones , Ratones Endogámicos C57BL , Reflejo , Centro Respiratorio/metabolismo , Centro Respiratorio/fisiopatología , Proteína delta-6 de Union Comunicante
4.
Anesth Analg ; 132(5): 1244-1253, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33857966

RESUMEN

Opioids are commonly used for pain management, perioperative procedures, and addiction treatment. There is a current opioid epidemic in North America that is paralleled by a marked increase in related deaths. Since 2000, chronic opioid users have been recognized to have significant central sleep apnea (CSA). After heart failure-related Cheyne-Stokes breathing (CSB), opioid-induced CSA is now the second most commonly seen CSA. It occurs in around 24% of chronic opioid users, typically after opioids have been used for more than 2 months, and usually corresponds in magnitude to opioid dose/plasma concentration. Opioid-induced CSA events often mix with episodes of ataxic breathing. The pathophysiology of opioid-induced CSA is based on dysfunction in respiratory rhythm generation and ventilatory chemoreflexes. Opioids have a paradoxical effect on different brain regions, which result in irregular respiratory rhythm. Regarding ventilatory chemoreflexes, chronic opioid use induces hypoxia that appears to stimulate an augmented hypoxic ventilatory response (high loop gain) and cause a narrow CO2 reserve, a combination that promotes respiratory instability. To date, no direct evidence has shown any major clinical consequence from CSA in chronic opioid users. A line of evidence suggested increased morbidity and mortality in overall chronic opioid users. CSA in chronic opioid users is likely to be a compensatory mechanism to avoid opioid injury and is potentially beneficial. The current treatments of CSA in chronic opioid users mainly focus on continuous positive airway pressure (CPAP) and adaptive servo-ventilation (ASV) or adding oxygen. ASV is more effective in reducing CSA events than CPAP. However, a recent ASV trial suggested an increased all-cause and cardiovascular mortality with the removal of CSA/CSB in cardiac failure patients. A major reason could be counteracting of a compensatory mechanism. No similar trial has been conducted for chronic opioid-related CSA. Future studies should focus on (1) investigating the phenotypes and genotypes of opioid-induced CSA that may have different clinical outcomes; (2) determining if CSA in chronic opioid users is beneficial or detrimental; and (3) assessing clinical consequences on different treatment options on opioid-induced CSA.


Asunto(s)
Analgésicos Opioides/efectos adversos , Encéfalo/efectos de los fármacos , Pulmón/inervación , Trastornos Relacionados con Opioides/complicaciones , Respiración/efectos de los fármacos , Apnea Central del Sueño/inducido químicamente , Encéfalo/fisiopatología , Humanos , Trastornos Relacionados con Opioides/mortalidad , Trastornos Relacionados con Opioides/fisiopatología , Pronóstico , Centro Respiratorio/efectos de los fármacos , Centro Respiratorio/fisiopatología , Medición de Riesgo , Factores de Riesgo , Apnea Central del Sueño/mortalidad , Apnea Central del Sueño/fisiopatología
5.
J Neurosci ; 41(21): 4732-4747, 2021 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-33863785

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disorder anatomically characterized by a progressive loss of dopaminergic neurons in the substantia nigra compacta (SNpc). Much less known, yet clinically very important, are the detrimental effects on breathing associated with this disease. Consistent with the human pathophysiology, the 6-hydroxydopamine hydrochloride (6-OHDA) rodent model of PD shows reduced respiratory frequency (fR) and NK1r-immunoreactivity in the pre-Bötzinger complex (preBötC) and PHOX2B+ neurons in the retrotrapezoid nucleus (RTN). To unravel mechanisms that underlie bradypnea in PD, we employed a transgenic approach to label or stimulate specific neuron populations in various respiratory-related brainstem regions. PD mice were characterized by a pronounced decreased number of putatively rhythmically active excitatory neurons in the preBötC and adjacent ventral respiratory column (VRC). Specifically, the number of Dbx1 and Vglut2 neurons was reduced by 47.6% and 17.3%, respectively. By contrast, inhibitory Vgat+ neurons in the VRC, as well as neurons in other respiratory-related brainstem regions, showed relatively minimal or no signs of neuronal loss. Consistent with these anatomic observations, optogenetic experiments identified deficits in respiratory function that were specific to manipulations of excitatory (Dbx1/Vglut2) neurons in the preBötC. We conclude that the decreased number of this critical population of respiratory neurons is an important contributor to the development of irregularities in inspiratory rhythm generation in this mouse model of PD.SIGNIFICANCE STATEMENT We found a decreased number of a specific population of medullary neurons which contributes to breathing abnormalities in a mouse model of Parkinson's disease (PD).


Asunto(s)
Neuronas/patología , Trastornos Parkinsonianos/fisiopatología , Trastornos Respiratorios/fisiopatología , Centro Respiratorio/fisiopatología , Animales , Femenino , Inhalación/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Trastornos Parkinsonianos/complicaciones , Trastornos Parkinsonianos/patología , Trastornos Respiratorios/etiología , Trastornos Respiratorios/patología , Centro Respiratorio/patología
6.
J Med Virol ; 93(3): 1296-1303, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32964419

RESUMEN

The recent outbreak of the novel coronavirus, SARS-CoV-2, has emerged to be highly pathogenic in nature. Although lungs are considered as the primary infected organs by SARS-CoV-2, some of the other organs, including the brain, have also been found to be affected. Here, we have discussed how SARS-CoV-2 might infect the brain. The infection of the respiratory center in the brainstem could be hypothesized to be responsible for the respiratory failure in many COVID-19 patients. The virus might gain entry through the olfactory bulb and invade various parts of the brain, including the brainstem. Alternatively, the entry might also occur from peripheral circulation into the central nervous system by compromising the blood-brain barrier. Finally, yet another possible entry route could be its dispersal from the lungs into the vagus nerve via the pulmonary stretch receptors, eventually reaching the brainstem. Therefore, screening neurological symptoms in COVID-19 patients, especially toward the breakdown of the respiratory center in the brainstem, might help us better understand this disease.


Asunto(s)
Encéfalo/virología , COVID-19/fisiopatología , COVID-19/virología , Vías Nerviosas/virología , Centro Respiratorio/virología , SARS-CoV-2/patogenicidad , Animales , Encéfalo/patología , Encéfalo/fisiopatología , COVID-19/patología , Citocinas/metabolismo , Humanos , Inflamación , Vías Nerviosas/fisiopatología , Neuronas/virología , Centro Respiratorio/patología , Centro Respiratorio/fisiopatología , Insuficiencia Respiratoria , Tropismo Viral
7.
Sleep Breath ; 25(2): 563-570, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32870421

RESUMEN

PURPOSE: Obstructive sleep apnea (OSA) is a complex disease in which phenotypic analysis and understanding pathological mechanisms facilitate personalized treatment and outcomes. However, the pathophysiology responsible for this robust observation is incompletely understood. The objective of the present work was to review how respiratory center regulation varies during sleep and wakeness in patients with OSA. DATA SOURCES: We searched for relevant articles up to December 31, 2019 in PubMed database. METHODS: This review examines the current literature on the characteristics of respiratory center regulation during wakefulness and sleep in OSA, detection method, and phenotypic treatment for respiratory center regulation. RESULTS: Mechanisms for ventilatory control system instability leading to OSA include different sleep stages in chemoresponsiveness to hypoxia and hypercapnia and different chemosensitivity at different time. One can potentially stabilize the breathing center in sleep-related breathing disorders by identifying one or more of these pathophysiological mechanisms. CONCLUSIONS: Advancing mechanism research in OSA will guide symptom research and provide alternate and novel opportunities for effective treatment for patients with OSA.


Asunto(s)
Centro Respiratorio/fisiopatología , Apnea Obstructiva del Sueño/fisiopatología , Humanos
8.
Neurol India ; 68(5): 989-993, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33109839

RESUMEN

BACKGROUND: A terrible pandemic, Covid-19, has captivated scientists to investigate if SARS-CoV-2 virus infects the central nervous system (CNS). A crucial question is if acute respiratory distress syndrome (ARDS), the main cause of death in this pandemic, and often refractory to treatments, can be explained by respiratory center dysfunction. OBJECTIVE: To discuss that ARDS can be caused by SARS-CoV-2 infection of the respiratory center in the brainstem. MATERIALS AND METHODS: I reviewed literature about SARS-CoV-2 mechanisms to infect the respiratory center in the brainstem. RESULTS AND CONCLUSIONS: An increasing amount of reports demonstrates that neurotropism is a common feature of coronavirus, which have been found in the brains of patients and experimental models, where the brainstem was severely infested. Recent studies have provided tremendous indication of the incidence of acute respiratory failure due to SARS-CoV-2 infection of the brainstem. SARS-CoV-2 might infect the CNS through the olfactory bulb, spreading from the olfactory nerves to the rhinencephalon, and finally reaching the brainstem. Hence, the virus infection causes respiratory center dysfunctions, leading to ARDS in COVID-19 patients. I conclude that acute ARDS in Covid-19 can be caused by SARS-CoV-2 invasion of brainstem respiratory center, suggesting the needs of more specific and aggressive treatments, with the direct participation of neurologists and neurointensivists.


Asunto(s)
Infecciones por Coronavirus/fisiopatología , Neumonía Viral/fisiopatología , Centro Respiratorio/fisiopatología , Síndrome de Dificultad Respiratoria/fisiopatología , Betacoronavirus , Tronco Encefálico/fisiopatología , Tronco Encefálico/virología , COVID-19 , Humanos , Hipoxia/fisiopatología , Pandemias , Centro Respiratorio/virología , SARS-CoV-2 , Tropismo Viral
9.
ACS Chem Neurosci ; 11(16): 2416-2421, 2020 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-32600045

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been established as a cause of severe alveolar damage and pneumonia in patients with advanced Coronavirus disease (COVID-19). The consolidation of lung parenchyma precipitates the alterations in blood gases in COVID-19 patients that are known to complicate and cause hypoxemic respiratory failure. With SARS-CoV-2 damaging multiple organs in COVID-19, including the central nervous system that regulates the breathing process, it is a daunting task to compute the extent to which the failure of the central regulation of the breathing process contributes to the mortality of COVID-19 affected patients. Emerging data on COVID-19 cases from hospitals and autopsies in the last few months have helped in the understanding of the pathogenesis of respiratory failures in COVID-19. Recent reports have provided overwhelming evidence of the occurrence of acute respiratory failures in COVID-19 due to neurotropism of the brainstem by SARS-CoV-2. In this review, a cascade of events that may follow the alterations in blood gases and possible neurological damage to the respiratory regulation centers in the central nervous system (CNS) in COVID-19 are related to the basic mechanism of respiratory regulation in order to understand the acute respiratory failure reported in this disease. Though a complex metabolic and respiratory dysregulation also occurs with infections caused by SARS-CoV-1 and MERS that are known to contribute toward deaths of the patients in the past, we highlight here the role of systemic dysregulation and the CNS respiratory regulation mechanisms in the causation of mortalities seen in COVID-19. The invasion of the CNS by SARS-CoV-2, as shown recently in areas like the brainstem that control the normal breathing process with nuclei like the pre-Bötzinger complex (pre-BÖTC), may explain why some of the patients with COVID-19, who have been reported to have recovered from pneumonia, could not be weaned from invasive mechanical ventilation and the occurrences of acute respiratory arrests seen in COVID-19. This debate is important for many reasons, one of which is the fact that permanent damage to the medullary respiratory centers by SARS-CoV-2 would not benefit from mechanical ventilators, as is possibly occurring during the management of COVID-19 patients.


Asunto(s)
Infecciones por Coronavirus/fisiopatología , Hipoxia/fisiopatología , Neumonía Viral/fisiopatología , Centro Respiratorio/fisiopatología , Insuficiencia Respiratoria/fisiopatología , Betacoronavirus , Análisis de los Gases de la Sangre , COVID-19 , Infecciones por Coronavirus/metabolismo , Infecciones por Coronavirus/mortalidad , Humanos , Hipoxia/metabolismo , Pandemias , Neumonía Viral/metabolismo , Neumonía Viral/mortalidad , Centro Respiratorio/metabolismo , Centro Respiratorio/virología , Insuficiencia Respiratoria/metabolismo , Insuficiencia Respiratoria/mortalidad , SARS-CoV-2 , Tropismo Viral
10.
ACS Chem Neurosci ; 11(10): 1379-1381, 2020 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-32348111

RESUMEN

Following the identification of severe acute respiratory syndrome coronavirus (SARS-CoV) in 2002 and Middle East respiratory syndrome coronavirus (MERS-CoV) in 2012, we are now again facing a global highly pathogenic novel coronavirus (SARS-CoV-2) epidemic. Although the lungs are one of the most critically affected organs, several other organs, including the brain may also get infected. Here, we have highlighted that SARS-CoV-2 might infect the central nervous system (CNS) through the olfactory bulb. From the olfactory bulb, SARS-CoV-2 may target the deeper parts of the brain including the thalamus and brainstem by trans-synaptic transfer described for many other viral diseases. Following this, the virus might infect the respiratory center of brain, which could be accountable for the respiratory breakdown of COVID-19 patients. Therefore, it is important to screen the COVID-19 patients for neurological symptoms as well as possibility of the collapse of the respiratory center in the brainstem should be investigated in depth.


Asunto(s)
Betacoronavirus/patogenicidad , Infecciones por Coronavirus/fisiopatología , Infecciones por Coronavirus/virología , Neumonía Viral/fisiopatología , Neumonía Viral/virología , Trastornos Respiratorios/virología , Centro Respiratorio/virología , Animales , Betacoronavirus/aislamiento & purificación , COVID-19 , Infecciones por Coronavirus/epidemiología , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Pandemias , Neumonía Viral/epidemiología , Trastornos Respiratorios/fisiopatología , Centro Respiratorio/fisiopatología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/patogenicidad , SARS-CoV-2
11.
Biol Open ; 9(4)2020 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-32086251

RESUMEN

Respiration is controlled by central pattern generating circuits in the brain stem, whose activity can be modulated by inputs from other brain areas to adapt respiration to autonomic and behavioral demands. The cerebellum is known to be part of the neuronal circuitry activated during respiratory challenges, such as hunger for air, but has not been found to be involved in the control of spontaneous, unobstructed breathing (eupnea). Here we applied a measure of intrinsic rhythmicity, the CV2, which evaluates the similarity of subsequent intervals and is thus sensitive to changes in rhythmicity at the temporal resolution of individual respiratory intervals. The variability of intrinsic respiratory rhythmicity was reduced in a mouse model of cerebellar ataxia compared to their healthy littermates. Irrespective of that difference, the average respiratory rate and the average coefficient of variation (CV) were comparable between healthy and ataxic mice. We argue that these findings are consistent with a proposed role of the cerebellum in modulating the duration of individual respiratory intervals, which could serve the purpose of coordinating respiration with other rhythmic orofacial movements, such as fluid licking and swallowing.


Asunto(s)
Cerebelo/fisiopatología , Periodicidad , Respiración , Animales , Ataxia Cerebelosa/etiología , Ataxia Cerebelosa/metabolismo , Ataxia Cerebelosa/fisiopatología , Cerebelo/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Femenino , Genotipo , Masculino , Ratones , Ratones Transgénicos , Centro Respiratorio/fisiopatología , Mecánica Respiratoria
12.
J Am Heart Assoc ; 8(19): e012441, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31549565

RESUMEN

Background The potential alterations of respiratory pathophysiology after cardiopulmonary resuscitation (CPR) are relatively undefined. While untreated arrest is known to affect post-cardiopulmonary resuscitation circulation, whether it affects respiratory pathophysiology remains unclear. We aimed to investigate the post-cardiopulmonary resuscitation changes in respiratory mechanics and neural respiratory drive with varying delays (5 or 10 minutes) in the treatment of ventricular fibrillation (VF). Methods and Results Twenty-six male Yorkshire pigs were used. Anesthetized pigs weighing 38±5 kg were randomized into 3 groups (n=10 each in the VF5 and VF10 groups, with VF kept untreated for 5 and 10 minutes, respectively, and n=6 in the sham group without VF). Defibrillation was attempted after 6 minutes of cardiopulmonary resuscitation. Pulse-induced contour cardiac output, respiratory mechanics, diaphragmatic electromyogram, blood gas, lung imaging, and histopathology were evaluated for 12 hours. Significantly elevated mean root mean square of diaphragmatic electromyogram, transdiaphragmatic pressure, and minute ventilation were observed, but reduced minute ventilation/mean root mean square, dynamic pulmonary compliance, and Pao2 were noted in both VF groups. Despite recovery of spontaneous breathing, the abnormalities in respiratory mechanics and neural respiratory drive, Pao2, and extravascular lung water continued to last for >12 hours. The changes in imaging (P=0.027) and histopathology (P=0.012) were more severe in the VF10 group compared with the VF5 group. Conclusions There is an uncoupling between the respiratory center and ventilation after restoration of spontaneous circulation. Prolonged untreated arrest from cardiac arrest contributes to more serious alterations in lung pathophysiology.


Asunto(s)
Reanimación Cardiopulmonar , Paro Cardíaco/terapia , Pulmón/inervación , Centro Respiratorio/fisiopatología , Mecánica Respiratoria , Fibrilación Ventricular/terapia , Animales , Modelos Animales de Enfermedad , Paro Cardíaco/diagnóstico , Paro Cardíaco/fisiopatología , Hemodinámica , Pulmón/diagnóstico por imagen , Masculino , Edema Pulmonar/diagnóstico por imagen , Edema Pulmonar/fisiopatología , Recuperación de la Función , Sus scrofa , Factores de Tiempo , Fibrilación Ventricular/diagnóstico , Fibrilación Ventricular/fisiopatología
13.
Respir Physiol Neurobiol ; 265: 172-179, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30009993

RESUMEN

The hypothalamus is a higher center of the autonomic nervous system and maintains essential body homeostasis including respiration. The paraventricular nucleus, perifornical area, dorsomedial hypothalamus, and lateral and posterior hypothalamus are the primary nuclei of the hypothalamus critically involved in respiratory control. These hypothalamic nuclei are interconnected with respiratory nuclei located in the midbrain, pons, medulla and spinal cord. We provide an extensive review of the role of the above hypothalamic nuclei in the maintenance of basal ventilation, and modulation of respiration in hypoxic and hypercapnic conditions, during dynamic exercise, in awake and sleep states, and under stress. Dysfunction of the hypothalamus causes abnormal breathing and hypoventilation. However, the cellular and molecular mechanisms how the hypothalamus integrates and modulates autonomic and respiratory functions remain to be elucidated.


Asunto(s)
Ejercicio Físico/fisiología , Hipercapnia/fisiopatología , Hipotálamo/fisiología , Hipoxia/fisiopatología , Red Nerviosa/fisiología , Respiración , Centro Respiratorio/fisiología , Estrés Psicológico/fisiopatología , Animales , Humanos , Red Nerviosa/fisiopatología , Centro Respiratorio/fisiopatología
14.
Respir Physiol Neurobiol ; 265: 153-160, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-29933053

RESUMEN

Breathing at rest is typically characterized by three phases: active inspiration, post-inspiration (or stage 1 expiration), and passive expiration (or stage 2 expiration). Breathing during periods of increased respiratory demand, on the other hand, engages active expiration through recruitment of abdominal muscles in order to increase ventilation. It is currently hypothesized that different phases of the respiratory rhythm are driven by three coupled oscillators: the preBötzinger Complex, driving inspiration, the parafacial respiratory group (pFRG), driving active expiration and the post-inspiratory Complex, driving post-inspiration. In this paper we review advances in the understanding of the pFRG and its role in the generation of active expiration across different developmental stages and vigilance states. Recent experiments suggest that the abdominal recruitment varies across development depending on the vigilance state, possibly following the maturation of the network responsible for the generation of active expiration and neuromodulatory systems that influence its activity. The activity of the pFRG is tonically inhibited by GABAergic inputs and strongly recruited by cholinergic systems. However, the sources of these modulatory inputs and the physiological conditions under which these mechanisms are used to recruit active expiration and increase ventilation need further investigation. Some evidence suggests that active expiration during hypercapnia is evoked through disinhibition, while during hypoxia it is elicited through activation of catecholaminergic C1 neurons. Finally, a discussion of experiments indicating that the pFRG is anatomically and functionally distinct from the adjacent and partially overlapping chemosensitive neurons of the retrotrapezoid nucleus is also presented.


Asunto(s)
Músculos Abdominales/fisiología , Espiración/fisiología , Desarrollo Fetal/fisiología , Hipercapnia/fisiopatología , Hipoxia/fisiopatología , Centro Respiratorio/fisiología , Sueño REM/fisiología , Animales , Humanos , Centro Respiratorio/fisiopatología
15.
Hypertens Res ; 41(12): 1000-1012, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30242293

RESUMEN

The aim of this study is to evaluate the short-term and long-term effects elicited by carotid body removal (CBR) on ventilatory function and the development of hypertension in the offspring of malnourished rats. Wistar rats were fed a normo-protein (NP, 17% casein) or low-protein (LP, 8% casein) diet during pregnancy and lactation. At 29 days of age, the animals were submitted to CBR or a sham surgery, according to the following groups: NP-cbr, LP-cbr, NP-sham, or LP-sham. In the short-term, at 30 days of age, the respiratory frequency (RF) and immunoreactivity for Fos on the retrotrapezoid nucleus (RTN; brainstem site containing CO2 sensitive neurons) after exposure to CO2 were evaluated. In the long term, at 90 days of age, arterial pressure (AP), heart rate (HR), and cardiovascular variability were evaluated. In the short term, an increase in the baseline RF (~6%), response to CO2 (~8%), and Fos in the RTN (~27%) occurred in the LP-sham group compared with the NP-sham group. Interestingly, the CBR in the LP group normalized the RF in response to CO2 as well as RTN cell activation. In the long term, CBR reduced the mean AP by ~20 mmHg in malnourished rats. The normalization of the arterial pressure was associated with a decrease in the low-frequency (LF) oscillatory component of AP (~58%) and in the sympathetic tonus to the cardiovascular system (~29%). In conclusion, carotid body inputs in malnourished offspring may be responsible for the following: (i) enhanced respiratory frequency and CO2 chemosensitivity in early life and (ii) the production of autonomic imbalance and the development of hypertension.


Asunto(s)
Presión Arterial/fisiología , Cuerpo Carotídeo/cirugía , Dieta con Restricción de Proteínas , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Frecuencia Respiratoria/fisiología , Animales , Cuerpo Carotídeo/fisiopatología , Femenino , Frecuencia Cardíaca/fisiología , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Wistar , Centro Respiratorio/metabolismo , Centro Respiratorio/fisiopatología
16.
Exp Physiol ; 103(10): 1377-1389, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30070746

RESUMEN

NEW FINDINGS: What is the central question of this study? What is the relationship between neuroanatomical and functional respiratory changes in an experimental model of Parkinson's disease? What is the main finding and its importance? Sixty days after induction of Parkinson's disease in a rat model, there are decreases in baseline breathing and in the number of neurons, density of the neurokinin-1 receptor and density of astrocytes in the ventrolateral respiratory region. These results provide the first evidence that neuroanatomical changes occur before functional respiratory deficits in a Parkinson's disease model and that there is a positive correlation between those sets of changes. The neuroanatomical changes impair respiratory activity and are presumably a major cause of the respiratory problems observed in Parkinson's disease. ABSTRACT: We showed previously that 60 days after the induction of Parkinson's disease (PD) in a rat model, there are decreases in baseline breathing and in the number of phox2b-expressing neurons of the retrotrapezoid nucleus (RTN) and nucleus of the solitary tract (NTS), as well as a reduction in the density of the neurokinin-1 receptor (NK1r) in the pre-Bötzinger complex (preBötC) and rostral ventrolateral respiratory group (rVRG). Here, our aim was to evaluate the correlation between neuroanatomical and functional respiratory changes in an experimental model of PD. Male Wistar rats with bilateral injections of 6-hydroxydopamine (6-OHDA, 24 µg µl-1 ) or vehicle into the striatum had respiratory parameters assessed by whole-body plethysmography 1 day before and 30, 40 or 60 days after the ablation. From the 30th day after the ablation, we observed a reduction in the number of phox2b neurons in the RTN and NTS and a reduction in the density of astrocytes in the rVRG. At 40 days after the ablation, we observed decreases in the density of NK1r in the preBötC and rVRG and of astrocytes in the RTN region. At 60 days, we observed a reduction in the density of astrocytes in the NTS and preBötC regions. The functional data showed changes in the resting and hypercapnia-induced respiratory rates and tidal volume from days 40-60 after injury. Our data suggest that the neuroanatomical changes impair respiratory activity and are presumably a major cause of the respiratory problems observed in PD.


Asunto(s)
Neuronas/patología , Enfermedad de Parkinson/fisiopatología , Centro Respiratorio/fisiopatología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Cuerpo Estriado/fisiopatología , Proteínas de Homeodominio/metabolismo , Hipercapnia/metabolismo , Hipercapnia/fisiopatología , Masculino , Modelos Teóricos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oxidopamina/administración & dosificación , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Ratas , Ratas Wistar , Receptores de Neuroquinina-1/metabolismo , Respiración/efectos de los fármacos , Centro Respiratorio/efectos de los fármacos , Centro Respiratorio/metabolismo , Núcleo Solitario/efectos de los fármacos , Núcleo Solitario/metabolismo , Núcleo Solitario/fisiopatología , Factores de Transcripción/metabolismo
17.
Med Hypotheses ; 110: 101-104, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29317050

RESUMEN

Oxygen therapy, usually administered by a facemask or nasal cannulae, is the current default treatment of respiratory failure. Since respiration entails intake of oxygen and release of carbon dioxide from tissues as waste product, the notion of administering carbon dioxide in respiratory failure appears counter-intuitive. However, carbon dioxide stimulates the chemosensitive area of the medulla, known as the central respiratory chemoreceptor, which activates the respiratory groups of neurones in the brainstem and stimulates inspiration thereby initiating oxygen intake during normal breathing. This vital initiation of normal breathing is via a reduction in the pH of the cerebrospinal fluid and the medullary interstitial fluid. We hypothesise that in cases of type I respiratory failure in which the PaCO2 is low, administration of carbon dioxide by inhalation would stimulate the respiratory groups of brainstem neurones and facilitate breathing, which would be of therapeutic value. Preliminary clinical evidence in favour of this hypothesis is presented and we recommend that a formal randomised study be carried out.


Asunto(s)
Dióxido de Carbono/uso terapéutico , Hipocapnia/terapia , Insuficiencia Respiratoria/terapia , Administración por Inhalación , Adulto , Dióxido de Carbono/administración & dosificación , Dióxido de Carbono/sangre , Células Quimiorreceptoras/fisiología , Femenino , Humanos , Hipocapnia/complicaciones , Hipocapnia/fisiopatología , Modelos Biológicos , Terapia por Inhalación de Oxígeno , Centro Respiratorio/fisiopatología , Insuficiencia Respiratoria/complicaciones , Insuficiencia Respiratoria/fisiopatología
18.
Adv Exp Med Biol ; 1015: 217-237, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29080029

RESUMEN

Neural networks, including the respiratory network, can undergo a reconfiguration process by just changing the number, the connectivity or the activity of their elements. Those elements can be either brain regions or neurons, which constitute the building blocks of macrocircuits and microcircuits, respectively. The reconfiguration processes can also involve changes in the number of connections and/or the strength between the elements of the network. These changes allow neural networks to acquire different topologies to perform a variety of functions or change their responses as a consequence of physiological or pathological conditions. Thus, neural networks are not hardwired entities, but they constitute flexible circuits that can be constantly reconfigured in response to a variety of stimuli. Here, we are going to review several examples of these processes with special emphasis on the reconfiguration of the respiratory rhythm generator in response to different patterns of hypoxia, which can lead to changes in respiratory patterns or lasting changes in frequency and/or amplitude.


Asunto(s)
Hipoxia/fisiopatología , Modelos Neurológicos , Red Nerviosa/fisiopatología , Centro Respiratorio/fisiopatología , Mecánica Respiratoria/fisiología , Animales , Neuronas/fisiología
19.
Epilepsia ; 58(6): e87-e90, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28464295

RESUMEN

Seizure spread into autonomic and respiratory brainstem regions is thought to play an important role in sudden unexpected death in epilepsy (SUDEP). As the clinical dataset of cases of definite SUDEP available for study grows, evidence points to a sequence of events that includes postictal apnea, bradycardia, and asystole as critical events that can lead to death. One possible link between the precipitating seizure and the critical postictal sequence is seizure-driven laryngospasm sufficient to completely obstruct the airway for an extended period, but ictal laryngospasm is difficult to fully assess. Herein, we demonstrate in a rat model how the electrical artifacts of attempts to inspire during airway obstruction and features of the cardiac rhythm establish this link between ictal and postictal activity and can be used as practical biomarkers of obstructive apnea due to laryngospasm or other causes of airway obstruction.


Asunto(s)
Sistema Nervioso Autónomo/fisiopatología , Biomarcadores , Tronco Encefálico/fisiopatología , Muerte Súbita/etiología , Muerte Súbita/prevención & control , Modelos Animales de Enfermedad , Electrocardiografía , Electromiografía , Epilepsia/complicaciones , Epilepsia/fisiopatología , Laringismo/complicaciones , Laringismo/fisiopatología , Centro Respiratorio/fisiopatología , Procesamiento de Señales Asistido por Computador , Apnea Obstructiva del Sueño/complicaciones , Apnea Obstructiva del Sueño/fisiopatología , Animales , Masculino , Ratas Sprague-Dawley , Factores de Riesgo , Convulsiones/complicaciones , Convulsiones/fisiopatología , Trabajo Respiratorio
20.
Am J Physiol Regul Integr Comp Physiol ; 312(6): R864-R872, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28438764

RESUMEN

Despite recent advances in the knowledge of the neural control of cardiovascular function, the cause of sympathetic overactivity in neurogenic hypertension remains unknown. Studies from our laboratory point out that rats submitted to chronic intermittent hypoxia (CIH), an experimental model of neurogenic hypertension, present changes in the central respiratory network that impact the pattern of sympathetic discharge and the levels of arterial pressure. In addition to the fine coordination of respiratory muscle contraction and relaxation, which is essential for O2 and CO2 pulmonary exchanges, neurons of the respiratory network are connected precisely to the neurons controlling the sympathetic activity in the brain stem. This respiratory-sympathetic neuronal interaction provides adjustments in the sympathetic outflow to the heart and vasculature during each respiratory phase according to the metabolic demands. Herein, we report that CIH-induced sympathetic over activity and mild hypertension are associated with increased frequency discharge of ventral medullary presympathetic neurons. We also describe that their increased frequency discharge is dependent on synaptic inputs, mostly from neurons of the brain stem respiratory network, rather than changes in their intrinsic electrophysiological properties. In perspective, we are taking into consideration the possibility that changes in the central respiratory rhythm/pattern generator contribute to increased sympathetic outflow and the development of neurogenic hypertension. Our experimental evidence provides support for the hypothesis that changes in the coupling of respiratory and sympathetic networks might be one of the unrevealed secrets of neurogenic hypertension in rats.


Asunto(s)
Presión Arterial , Sistema Cardiovascular/inervación , Hipertensión/fisiopatología , Pulmón/inervación , Respiración , Centro Respiratorio/fisiopatología , Sistema Nervioso Simpático/fisiopatología , Animales , Modelos Animales de Enfermedad , Humanos , Hipertensión/etiología , Hipoxia/complicaciones , Hipoxia/fisiopatología , Modelos Cardiovasculares , Vías Nerviosas/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...