Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Ther Adv Rare Dis ; 4: 26330040231181406, 2023.
Article in English | MEDLINE | ID: mdl-37621556

ABSTRACT

Background: Due to racial, cultural, and linguistic marginalization, some populations experience disproportionate barriers to genetic testing in both clinical and research settings. It is difficult to track such disparities due to non-inclusive self-reported race and ethnicity categories within the electronic health record (EHR). Inclusion and access for all populations is critical to achieve health equity and to capture the full spectrum of rare genetic disease. Objective: We aimed to create revised race and ethnicity categories. Additionally, we identified racial and ethnic under-representation amongst three cohorts: (1) the general Boston Children's Hospital patient population (general BCH), (2) the BCH patient population that underwent clinical genomic testing (clinical sequencing), and (3) Children's Rare Disease Cohort (CRDC) research initiative participants. Design and Methods: Race and ethnicity data were collected from the EHRs of the general BCH, clinical sequencing, and CRDC cohorts. We constructed a single comprehensive set of race and ethnicity categories. EHR-based race and ethnicity variables were mapped within each cohort to the revised categories. Then, the numbers of patients within each revised race and ethnicity category were compared across cohorts. Results: There was a significantly lower percentage of Black or African American/African, non-Hispanic/non-Latine individuals in the CRDC cohort compared with the general BCH cohort, but there was no statistically significant difference between the CRDC and the clinical sequencing cohorts. There was a significantly lower percentage of multi-racial, Hispanic/Latine individuals in the CRDC cohort than the clinical sequencing cohort. White, non-Hispanic/non-Latine individuals were over-represented in the CRDC compared to the two other groups. Conclusion: We highlight underrepresentation of certain racial and ethnic populations in sequencing cohorts compared to the general hospital population. We propose a range of measures to address these disparities, to strive for equitable future precision medicine-based clinical care and for the benefit of the whole rare disease community.


Racial and ethnic representation amongst general clinics, clinics that provide genetic testing, and genomic-based research at Boston Children's Hospital Background: Individuals who identify as belonging to a race or ethnicity that has been historically excluded from mainstream cultural, political, and economic activities ('historically marginalized') experience barriers to clinical care. These barriers are further complicated for families touched by rare genetic conditions. Obstacles can present as accessibility issues (transportation, financial, linguistic), low-quality medical care, or inadequate inclusion in research. It is important to have representation within rare disease research so that the full scope of these conditions is understood, leading to better patient care for all, and for health equity. Objective: We aimed to (1) to create new and inclusive race and ethnicity categories for the electronic health record (EHR) and (2) identify differences in racial and ethnic representation amongst patients generally seen at Boston Children's Hospital (general BCH), those who received genetic testing in a clinic at Boston Children's Hospital (clinical sequencing), and participants who enrolled in the CRDC research project at Boston Children's Hospital (CRDC). Design and Methods: We combined race and ethnicity categories to make more inclusive options than existing EHR categories. Differences in race and ethnicity representation were observed when looking at the three different patient groups (general BCH, clinical sequencing, and CRDC). Results: We observed a lower percentage of individuals who self-identify as Black or African American/African, non-Hispanic/non-Latine in the genetic testing groups (both research and clinical) than in the general BCH group. Individuals who self-identify as multi-racial, Hispanic/Latine are also under-represented in the CRDC research compared to the two other groups. The highest population percentage seen in all groups was that of patients who identify as White, non-Hispanic/non-Latine. This group was over-represented in the research CRDC group compared to the two others. Conclusion: Our study found that patients who are historically marginalized are underrepresented in clinical genetic testing and genomic research studies compared to their White counterparts. In order to benefit all patients with rare genetic conditions, these differences must be addressed by improving access to specialty physicians/researchers and incorporating inclusive language in the EHR, clinics, and research protocols.

2.
Mol Ther Methods Clin Dev ; 27: 32-46, 2022 Dec 08.
Article in English | MEDLINE | ID: mdl-36156879

ABSTRACT

Interest in gene-based therapies for neurodevelopmental disorders is increasing exponentially, driven by the rise in recognition of underlying genetic etiology, progress in genomic technology, and recent proof of concept in several disorders. The current prioritization of one genetic disorder over another for development of therapies is driven by competing interests of pharmaceutical companies, advocacy groups, and academic scientists. Although these are all valid perspectives, a consolidated framework will facilitate more efficient and rational gene therapy development. Here we outline features of Mendelian neurodevelopmental disorders that warrant consideration when determining suitability for gene therapy. These features fit into four broad domains: genetics, preclinical validation, clinical considerations, and ethics. We propose a simple mnemonic, GENE TARGET, to remember these features and illustrate how they could be scored using a preliminary scoring rubric. In this suggested rubric, for a given disorder, scores for each feature may be added up to a composite GENE TARGET suitability (GTS) score. In addition to proposing a systematic method to evaluate and compare disorders, our framework helps identify gaps in the translational pipeline for a given disorder, which can inform prioritization of future research efforts.

3.
J Headache Pain ; 21(1): 86, 2020 Jul 06.
Article in English | MEDLINE | ID: mdl-32631251

ABSTRACT

BACKGROUND: Migraine is a common headache disorder, with cortical spreading depolarization (CSD) considered as the underlying electrophysiological event. CSD is a slowly propagating wave of neuronal and glial depolarization. Sleep disorders are well known risk factors for migraine chronification, and changes in wake-sleep pattern such as sleep deprivation are common migraine triggers. The underlying mechanisms are unknown. As a step towards developing an animal model to study this, we test whether sleep deprivation, a modifiable migraine trigger, enhances CSD susceptibility in rodent models. METHODS: Acute sleep deprivation was achieved using the "gentle handling method", chosen to minimize stress and avoid confounding bias. Sleep deprivation was started with onset of light (diurnal lighting conditions), and assessment of CSD was performed at the end of a 6 h or 12 h sleep deprivation period. The effect of chronic sleep deprivation on CSD was assessed 6 weeks or 12 weeks after lesioning of the hypothalamic ventrolateral preoptic nucleus. All experiments were done in a blinded fashion with respect to sleep status. During 60 min of continuous topical KCl application, we assessed the total number of CSDs, the direct current shift amplitude and duration of the first CSD, the average and cumulative duration of all CSDs, propagation speed, and electrical CSD threshold. RESULTS: Acute sleep deprivation of 6 h (n = 17) or 12 h (n = 11) duration significantly increased CSD frequency compared to controls (17 ± 4 and 18 ± 2, respectively, vs. 14 ± 2 CSDs/hour in controls; p = 0.003 for both), whereas other electrophysiological properties of CSD were unchanged. Acute total sleep deprivation over 12 h but not over 6 h reduced the electrical threshold of CSD compared to controls (p = 0.037 and p = 0.095, respectively). Chronic partial sleep deprivation in contrast did not affect CSD susceptibility in rats. CONCLUSIONS: Acute but not chronic sleep deprivation enhances CSD susceptibility in rodents, possibly underlying its negative impact as a migraine trigger and exacerbating factor. Our findings underscore the importance of CSD as a therapeutic target in migraine and suggest that headache management should identify and treat associated sleep disorders.


Subject(s)
Migraine without Aura/physiopathology , Sleep Deprivation/physiopathology , Animals , Cortical Spreading Depression/physiology , Disease Models, Animal , Male , Rats , Rats, Sprague-Dawley
4.
Curr Biol ; 30(5): R219-R221, 2020 03 09.
Article in English | MEDLINE | ID: mdl-32155424

ABSTRACT

Acute activation of genetically defined sleep-promoting neurons in the preoptic hypothalamus does not facilitate anesthesia. This finding supports the notion that a mechanistic overlap between sleep and anesthesia does not explain the anesthetized brain.


Subject(s)
Anesthesia , Preoptic Area , Hypothalamus , Neurons , Sleep
5.
J Neurosci ; 39(40): 7910-7919, 2019 10 02.
Article in English | MEDLINE | ID: mdl-31420456

ABSTRACT

Proper function of pharyngeal dilator muscles, including the genioglossus muscle of the tongue, is required to maintain upper airway patency. During sleep, the activity of these muscles is suppressed, and as a result individuals with obstructive sleep apnea experience repeated episodes of upper airway closure when they are asleep, in particular during rapid-eye-movement (REM) sleep. Blocking cholinergic transmission in the hypoglossal motor nucleus (MoXII) restores REM sleep genioglossus activity, highlighting the importance of cholinergic transmission in the inhibition of hypoglossal motor neurons (HMNs) during REM sleep. Glutamatergic afferent input from neurons in the parahypoglossal (PH) region to the HMNs is critical for MoXII respiratory motor output. We hypothesized that state-dependent cholinergic regulation may be mediated by this pathway. Here we studied the effects of cholinergic transmission in HMNs in adult male and female mice using patch-clamp recordings in brain slices. Using channelrhodopsin-2-assisted circuit mapping, we first demonstrated that PH glutamatergic neurons directly and robustly activate HMNs (PHGlut → HMNs). We then show that carbachol consistently depresses this input and that this effect is presynaptic. Additionally, carbachol directly affects HMNs by a variable combination of muscarinic-mediated excitatory and inhibitory responses. Altogether, our results suggest that cholinergic signaling impairs upper airway dilator muscle activity by suppressing glutamatergic input from PH premotoneurons to HMNs and by directly inhibiting HMNs. Our findings highlight the complexity of cholinergic control of HMNs at both the presynaptic and postsynaptic levels and provide a possible mechanism for REM sleep suppression of upper airway muscle activity.SIGNIFICANCE STATEMENT Individuals with obstructive sleep apnea can breathe adequately when awake but experience repeated episodes of upper airway closure when asleep, in particular during REM sleep. Similar to skeletal postural muscles, pharyngeal dilator muscles responsible for maintaining an open upper airway become hypotonic during REM sleep. Unlike spinal motoneurons controlling postural muscles that are inhibited by glycinergic transmission during REM sleep, hypoglossal motoneurons that control the upper airway muscles are inhibited in REM sleep by the combination of monoaminergic disfacilitation and cholinergic inhibition. In this study, we demonstrated how cholinergic signaling inhibits hypoglossal motoneurons through presynaptic and postsynaptic muscarinic receptors. Our results provide a potential mechanism for upper airway hypotonia during REM sleep.


Subject(s)
Hypoglossal Nerve/physiopathology , Motor Neurons , Muscle Hypotonia/physiopathology , Parasympathetic Nervous System/physiopathology , Respiratory Muscles/physiopathology , Sleep, REM , Animals , Carbachol/pharmacology , Channelrhodopsins , Female , Glutamates/physiology , Hypoglossal Nerve/drug effects , Male , Mice , Muscarinic Agonists/pharmacology , Neurons, Afferent/drug effects , Neurons, Afferent/physiology , Optogenetics , Parasympathetic Nervous System/drug effects , Patch-Clamp Techniques , Synapses/drug effects
6.
J Comp Neurol ; 525(8): 1844-1860, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28032634

ABSTRACT

The Kölliker-Fuse nucleus (KF) is known primarily for its respiratory function as the "pneumotaxic center" or "pontine respiratory group." Considered part of the parabrachial (PB) complex, KF contains glutamatergic neurons that project to respiratory-related targets in the medulla and spinal cord (Yokota, Oka, Tsumori, Nakamura, & Yasui, 2007). Here we describe an unexpected population of neurons in the caudal KF and adjacent lateral crescent subnucleus (PBlc), which are γ-aminobutyric acid (GABA)ergic and have an entirely different pattern of projections than glutamatergic KF neurons. First, immunofluorescence, in situ hybridization, and Cre-reporter labeling revealed that many of these GABAergic neurons express FoxP2 in both rats and mice. Next, using Cre-dependent axonal tracing in Vgat-IRES-Cre and Vglut2-IRES-Cre mice, we identified different projection patterns from GABAergic and glutamatergic neurons in this region. GABAergic neurons in KF and PBlc project heavily and almost exclusively to trigeminal sensory nuclei, with minimal projections to cardiorespiratory nuclei in the brainstem, and none to the spinal cord. In contrast, glutamatergic KF neurons project heavily to the autonomic, respiratory, and motor regions of the medulla and spinal cord previously identified as efferent targets mediating KF cardiorespiratory effects. These findings identify a novel, GABAergic subpopulation of KF/PB neurons with a distinct efferent projection pattern targeting the brainstem trigeminal sensory system. Rather than regulating breathing, we propose that these neurons influence vibrissal sensorimotor function.


Subject(s)
GABAergic Neurons/cytology , Kolliker-Fuse Nucleus/cytology , Neural Pathways/cytology , Neurons/cytology , Animals , Female , Fluorescent Antibody Technique , Glutamic Acid , Image Processing, Computer-Assisted , Immunohistochemistry , In Situ Hybridization , Male , Mice , Mice, Transgenic , Rats , Rats, Wistar
7.
Anesthesiology ; 125(2): 333-45, 2016 08.
Article in English | MEDLINE | ID: mdl-27341276

ABSTRACT

BACKGROUND: Calabadion 2 is a new drug-encapsulating agent. In this study, the authors aim to assess its utility as an agent to reverse general anesthesia with etomidate and ketamine and facilitate recovery. METHODS: To evaluate the effect of calabadion 2 on anesthesia recovery, the authors studied the response of rats to calabadion 2 after continuous and bolus intravenous etomidate or ketamine and bolus intramuscular ketamine administration. The authors measured electroencephalographic predictors of depth of anesthesia (burst suppression ratio and total electroencephalographic power), functional mobility impairment, blood pressure, and toxicity. RESULTS: Calabadion 2 dose-dependently reverses the effects of ketamine and etomidate on electroencephalographic predictors of depth of anesthesia, as well as drug-induced hypotension, and shortens the time to recovery of righting reflex and functional mobility. Calabadion 2 displayed low cytotoxicity in MTS-3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium-based cell viability and adenylate kinase release cell necrosis assays, did not inhibit the human ether-à-go-go-related channel, and was not mutagenic (Ames test). On the basis of maximum tolerable dose and acceleration of righting reflex recovery, the authors calculated the therapeutic index of calabadion 2 in recovery as 16:1 (95% CI, 10 to 26:1) for the reversal of ketamine and 3:1 (95% CI, 2 to 5:1) for the reversal of etomidate. CONCLUSIONS: Calabadion 2 reverses etomidate and ketamine anesthesia in rats by chemical encapsulation at nontoxic concentrations.


Subject(s)
Anesthesia, General/methods , Heterocyclic Compounds, 4 or More Rings/pharmacology , Sulfonic Acids/pharmacology , Anesthetics, Dissociative/toxicity , Anesthetics, Intravenous/toxicity , Animals , Blood Pressure/drug effects , Cell Survival/drug effects , Electroencephalography/drug effects , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Etomidate/antagonists & inhibitors , Etomidate/toxicity , Ketamine/antagonists & inhibitors , Ketamine/toxicity , Male , Mutagens/toxicity , Necrosis/prevention & control , Postural Balance/drug effects , Rats , Rats, Sprague-Dawley , Reflex/drug effects
8.
Front Neurol ; 6: 140, 2015.
Article in English | MEDLINE | ID: mdl-26157418

ABSTRACT

The mesencephalic (or midbrain) locomotor region (MLR) was first described in 1966 by Shik and colleagues, who demonstrated that electrical stimulation of this region induced locomotion in decerebrate (intercollicular transection) cats. The pedunculopontine tegmental nucleus (PPT) cholinergic neurons and midbrain extrapyramidal area (MEA) have been suggested to form the neuroanatomical basis for the MLR, but direct evidence for the role of these structures in locomotor behavior has been lacking. Here, we tested the hypothesis that the MLR is composed of non-cholinergic spinally projecting cells in the lateral pontine tegmentum. Our results showed that putative MLR neurons medial to the PPT and MEA in rats were non-cholinergic, glutamatergic, and express the orexin (hypocretin) type 2 receptors. Fos mapping correlated with motor behaviors revealed that the dorsal and ventral MLR are activated, respectively, in association with locomotion and an erect posture. Consistent with these findings, chemical stimulation of the dorsal MLR produced locomotion, whereas stimulation of the ventral MLR caused standing. Lesions of the MLR (dorsal and ventral regions together) resulted in cataplexy and episodic immobility of gait. Finally, trans-neuronal tracing with pseudorabies virus demonstrated disynaptic input to the MLR from the substantia nigra via the MEA. These findings offer a new perspective on the neuroanatomic basis of the MLR, and suggest that MLR dysfunction may contribute to the postural and gait abnormalities in Parkinsonism.

9.
J Comp Neurol ; 523(6): 907-20, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25424719

ABSTRACT

In patients with obstructive sleep apnea, airway obstruction during sleep produces hypercapnia, which in turn activates respiratory muscles that pump air into the lungs (e.g., the diaphragm) and that dilate and stabilize the upper airway (e.g., the genioglossus). We hypothesized that these responses are facilitated by glutamatergic neurons in the parabrachial complex (PB) that respond to hypercapnia and project to premotor and motor neurons that innervate the diaphragm and genioglossus muscles. To test this hypothesis, we combined c-Fos immunohistochemistry with in situ hybridization for vGluT2 or GAD67 or with retrograde tracing from the ventrolateral medullary region that contains phrenic premotor neurons, the phrenic motor nucleus in the C3-C5 spinal ventral horn, or the hypoglossal motor nucleus. We found that hypercapnia (10% CO2 for 2 hours) activated c-Fos expression in neurons in the external lateral, lateral crescent (PBcr), and Kölliker-Fuse (KF) PB subnuclei and that most of these neurons were glutamatergic and virtually none γ-aminobutyric acidergic. Numerous CO2 -responsive neurons in the KF and PBcr were labeled after retrograde tracer injection into the ventrolateral medulla or hypoglossal motor nuclei, and in the KF after injections into the spinal cord, making them candidates for mediating respiratory-facilitatory and upper-airway-stabilizing effects of hypercapnia.


Subject(s)
Glutamic Acid/metabolism , Hypercapnia/metabolism , Motor Neurons/physiology , Neural Pathways/physiology , Parabrachial Nucleus/cytology , Respiration/immunology , Animals , Calcitonin Gene-Related Peptide/metabolism , Cholera Toxin/metabolism , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-fos/metabolism , Vesicular Glutamate Transport Protein 2/genetics , Vesicular Glutamate Transport Protein 2/metabolism
10.
Sleep ; 37(3): 561-9, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24587579

ABSTRACT

STUDY OBJECTIVES: Rapid eye movement (REM)-induced hypotonia of the major upper airway dilating muscle (genioglossus) potentially contributes to the worsening of obstructive sleep apnea that occurs during this stage. No prior human single motor unit (SMU) study of genioglossus has examined this possibility to our knowledge. We hypothesized that genioglossus SMUs would reduce their activity during stable breathing in both tonic and phasic REM compared to stage N2 sleep. Further, we hypothesized that hypopneas occurring in REM would be associated with coincident reductions in genioglossus SMU activity. DESIGN: The activity of genioglossus SMUs was studied in (1) neighboring epochs of stage N2, and tonic and phasic REM; and (2) during hypopneas occurring in REM. SETTING: Sleep laboratory. PARTICIPANTS: 29 subjects (38 ± 13 y) (17 male). INTERVENTION: Natural sleep, including REM sleep and REM hypopneas. MEASUREMENT AND RESULTS: Subjects slept overnight with genioglossus fine-wire intramuscular electrodes and full polysomnography. Forty-two SMUs firing during one or more of stage N2, tonic REM, or phasic REM were sorted. Twenty inspiratory phasic (IP), 17 inspiratory tonic (IT), and five expiratory tonic (ET) SMUs were characterized. Fewer units were active during phasic REM (23) compared to tonic REM (30) and stage N2 (33). During phasic REM sleep, genioglossus IP and IT SMUs discharged at slower rates and for shorter durations than during stage N2. For example, the SMU peak frequency during phasic REM 5.7 ± 6.6 Hz (mean ± standard deviation) was less than both tonic REM 12.3 ± 9.7 Hz and stage N2 16.1 ± 10.0 Hz (P < 0.001). The peak firing frequencies of IP/IT SMUs decreased from the last breath before to the first breath of a REM hypopnea (11.8 ± 10.9 Hz versus 5.7 ± 9.4 Hz; P = 0.001). CONCLUSION: Genioglossus single motor unit activity is significantly reduced in REM sleep, particularly phasic REM. Single motor unit activity decreases abruptly at the onset of REM hypopneas.


Subject(s)
Muscle Hypotonia/physiopathology , Sleep Apnea, Obstructive/physiopathology , Sleep, REM , Adult , Electromyography , Female , Humans , Male , Muscle, Skeletal/physiopathology , Polysomnography , Respiration , Tongue/physiopathology
11.
Curr Opin Neurobiol ; 23(5): 774-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23810448

ABSTRACT

Obstructive sleep apnea (OSA) is a disorder of repetitive sleep disruption caused by reduced or blocked respiratory airflow. Although an anatomically compromised airway accounts for the major predisposition to OSA, a patient's arousal threshold and factors related to the central control of breathing (ventilatory control stability) are also important. Arousal from sleep (defined by EEG desynchronization) may be the only mechanism that allows airway re-opening following an obstructive event. However, in many cases arousal is unnecessary and even worsens the severity of OSA. Mechanisms for arousal are poorly understood. However, accumulating data are elucidating the relevant neural pathways and neurotransmitters. For example, serotonin is critically required, but its site of action is unknown. Important neural substrates for arousal have been recently identified in the parabrachial complex (PB), a visceral sensory nucleus in the rostral pons. Moreover, glutamatergic signaling from the PB contributes to arousal caused by hypercapnia, one of the arousal-promoting stimuli in OSA. A major current focus of OSA research is to find means to maintain airway patency during sleep, without sleep interruption.


Subject(s)
Arousal/physiology , Brain/physiopathology , Neural Pathways/physiopathology , Sleep Apnea, Obstructive/physiopathology , Animals , Humans
12.
J Neurosci ; 33(18): 7627-40, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23637157

ABSTRACT

The mechanisms of arousal from apneas during sleep in patients suffering from obstructive sleep apnea are not well understood. However, we know that respiratory chemosensory pathways converge on the parabrachial nucleus (PB), which sends glutamatergic projections to a variety of forebrain structures critical to arousal, including the basal forebrain, lateral hypothalamus, midline thalamus, and cerebral cortex. We tested the role of glutamatergic signaling in this pathway by developing an animal model for repetitive CO2 arousals (RCAs) and investigating the effect of deleting the gene for the vesicular glutamate transporter 2 (Vglut2) from neurons in the PB. We used mice with lox P sequences flanking exon2 of the Vglut2 gene, in which adeno-associated viral vectors containing genes encoding Cre recombinase and green fluorescent protein were microinjected into the PB to permanently and selectively disrupt Vglut2 expression while labeling the affected neurons. We recorded sleep in these mice and then investigated the arousals during RCA. Vglut2 deletions that included the external lateral and lateral crescent subdivisions of the lateral PB more than doubled the latency to arousal and resulted in failure to arouse by 30 s in >30% of trials. By contrast, deletions that involved the medial PB subdivision had minimal effects on arousal during hypercapnia but instead increased non-rapid eye movement (NREM) sleep by ∼43% during the dark period, and increased delta power in the EEG during NREM sleep by ∼50%. Our results suggest that glutamatergic neurons in the lateral PB are necessary for arousals from sleep in response to CO2, while medial PB glutamatergic neurons play an important role in promoting spontaneous waking.


Subject(s)
Arousal , Brain Stem/physiology , Glutamic Acid/metabolism , Hypercapnia/physiopathology , Signal Transduction/physiology , Acoustic Stimulation , Analysis of Variance , Animals , Diphtheria Toxin/pharmacology , Electroencephalography , Electromyography , Eye Movements/physiology , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plethysmography , Reaction Time/physiology , Sleep/physiology , Time Factors , Vesicular Glutamate Transport Protein 2/deficiency , Vesicular Glutamate Transport Protein 2/genetics
13.
Neurobiol Dis ; 48(3): 348-55, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22776332

ABSTRACT

Hospitalized patients can develop cognitive function decline, the mechanisms of which remain largely to be determined. Sleep disturbance often occurs in hospitalized patients, and neuroinflammation can induce learning and memory impairment. We therefore set out to determine whether sleep disturbance can induce neuroinflammation and impairment of learning and memory in rodents. Five to 6-month-old wild-type C57BL/6J male mice were used in the studies. The mice were placed in rocking cages for 24 h, and two rolling balls were present in each cage. The mice were tested for learning and memory function using the Fear Conditioning Test one and 7 days post-sleep disturbance. Neuroinflammation in the mouse brain tissues was also determined. Of the Fear Conditioning studies at one day and 7 days after sleep disturbance, twenty-four hour sleep disturbance decreased freezing time in the context test, which assesses hippocampus-dependent learning and memory; but not the tone test, which assesses hippocampus-independent learning and memory. Sleep disturbance increased pro-inflammatory cytokine IL-6 levels and induced microglia activation in the mouse hippocampus, but not the cortex. These results suggest that sleep disturbance induces neuroinflammation in the mouse hippocampus, and impairs hippocampus-dependent learning and memory in mice. Pending further studies, these findings suggest that sleep disturbance-induced neuroinflammation and impairment of learning and memory may contribute to the development of cognitive function decline in hospitalized patients.


Subject(s)
Cognition Disorders/etiology , Cognition Disorders/physiopathology , Hippocampus/physiopathology , Inflammation/etiology , Sleep/physiology , Animals , Blotting, Western , Cognition Disorders/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Immunohistochemistry , Inflammation/metabolism , Inflammation/physiopathology , Interleukin-6/biosynthesis , Learning/physiology , Male , Memory/physiology , Mice , Mice, Inbred C57BL
14.
Anesthesiology ; 116(1): 35-46, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22108392

ABSTRACT

BACKGROUND: Procedural sedation is frequently performed in spontaneously breathing patients, but hypnotics and opioids decrease respiratory drive and place the upper airway at risk for collapse. METHODS: In a randomized, controlled, cross-over, pharmaco-physiologic study in 12 rats, we conducted acute experiments to compare breathing and genioglossus electromyogram activity at equianesthetic concentrations of ketamine, a noncompetitive N-methyl-D-aspartate receptor antagonist that combines potent analgesic with hypnotic action effects, versus propofol. In 10 chronically instrumented rats resting in a plethysmograph, we measured these variables as well as electroencephalography during five conditions: quiet wakefulness, nonrapid-eye-movement sleep, rapid eye movement sleep, and low-dose (60 mg/kg intraperitoneally) and high-dose ketamine anesthesia (125 mg/kg intraperitoneally). RESULTS: Ketamine anesthesia was associated with markedly increased genioglossus activity (1.5 to fivefold higher values of genioglossus electromyogram) compared with sleep- and propofol-induced unconsciousness. Plethysmography revealed a respiratory stimulating effect: higher values of flow rate, respiratory rate, and duty-cycle (effective inspiratory time, 1.5-to-2-fold higher values). During wakefulness and normal sleep, the δ (f = 6.51, P = 0.04) electroencephalogram power spectrum was an independent predictor of genioglossus activity, indicating an association between electroencephalographic determinants of consciousness and genioglossus activity. Following ketamine administration, electroencephalogram power spectrum and genioglossus electroencephalogram was dissociated (P = 0.9 for the relationship between δ/θ power spectrum and genioglossus electromyogram). CONCLUSIONS: Ketamine is a respiratory stimulant that abolishes the coupling between loss-of-consciousness and upper airway dilator muscle dysfunction in a wide dose-range. Ketamine compared with propofol might help stabilize airway patency during sedation and anesthesia.


Subject(s)
Anesthetics, Dissociative/pharmacology , Ketamine/pharmacology , Respiration/drug effects , Respiratory Muscles/drug effects , Unconsciousness/chemically induced , Anesthesia , Anesthesia, Intravenous , Anesthetics, Intravenous/pharmacology , Animals , Behavior, Animal/drug effects , Electroencephalography/drug effects , Electromyography/drug effects , Injections, Intraperitoneal , Male , Neck/physiology , Propofol/pharmacology , Rats , Rats, Sprague-Dawley , Respiratory Mechanics/drug effects , Sleep/physiology , Wakefulness/physiology
15.
Brain Res ; 1426: 30-7, 2011 Dec 02.
Article in English | MEDLINE | ID: mdl-22041226

ABSTRACT

Neurons of the ventrolateral preoptic nucleus (VLPO) promote sleep and VLPO loss produces insomnia. Previous studies show that general anesthetics including isoflurane activate VLPO neurons, and may contribute to their sedative effects. However, it is not clear to what extent the activation of VLPO neurons contributes to general anesthesia. We tested whether destruction of the VLPO neurons would affect the onset, depth, or recovery from isoflurane's general anesthetic effects. The VLPO was ablated in 25 rats by bilateral local injection of orexin-saporin, and polysomnography was performed to measure baseline sleep loss and responses to isoflurane anesthesia at 1% and 2%. Eight rats received sham (saline) injections. We measured isoflurane effects on time to loss of righting reflex, onset of continuous slow wave activity, and burst suppression; burst-suppression ratio; and time to recovery of righting reflex and desynchronized EEG. VLPO neuron cell loss was quantified by post hoc histology. Loss of VLPO neurons as well as lesion size were associated with cumulative sleep loss (r=0.77 and r=0.62, respectively), and cumulative sleep loss was the strongest predictor of high sensitivity to anesthesia, expressed as decreased time to loss of righting reflex (-0.59), increased burst-suppression ratio (r=0.52) , and increased emergence time (r=0.54); an interaction-effect of isoflurane dose was observed (burst-suppression ratio: p<0.001). We conclude that the sleep loss caused by ablation of VLPO neurons sensitizes animals to the general anesthetic effects of isoflurane, but that the sedation produced by VLPO neurons themselves is not required for isoflurane anesthesia.


Subject(s)
Anesthetics, General/pharmacology , Isoflurane/pharmacology , Neurons/drug effects , Preoptic Area/physiology , Sleep/physiology , Anesthesia, General , Animals , Male , Neurons/physiology , Preoptic Area/cytology , Preoptic Area/drug effects , Rats , Rats, Sprague-Dawley
20.
Curr Opin Anaesthesiol ; 22(6): 730-7, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19752726

ABSTRACT

PURPOSE OF REVIEW: This review describes strategies to control perioperative muscle strength in patients undergoing ambulatory surgery. RECENT FINDINGS: Although it is impossible to improve muscle relaxation (defined as absence of electrical activity) of intact resting muscle by hypnotics, analgesia is required to prevent pain-evoked muscular contractions during surgery. Regional anesthesia, as well as hypnotics and opioids, promotes intraoperative muscle relaxation. Neuromuscular blocking agents (NMBAs) induce dose-dependent muscle relaxation, but their effects vary widely between individuals, and postoperative residual curarization (PORC) exposes patients to additional risk. Low doses of NMBAs should, therefore, be used, effects be monitored quantitatively by acceleromyography, and residual neuromuscular block be reversed. Acetylcholinesterase inhibitor reversal can cause respiratory side effects, so the lowest efficacious dose should be used: as little as 0.015-0.025 mg kg(-1) of neostigmine is required at a train-of-four count of four with minimal fade. Sugammadex encapsulates steroidal NMBAs. Sugammadex reversal is a viable approach to rapidly antagonize deep levels of neuromuscular block. SUMMARY: Optimal muscle relaxation for ambulatory surgery results from a judicious combination of regional anesthesia, opioids, and low doses of NMBAs. The effects of NMBAs should be monitored quantitatively by acceleromyography and reversed appropriately.


Subject(s)
Ambulatory Surgical Procedures , Muscle Strength/physiology , Perioperative Care , Anesthesia, Conduction , Humans , Muscle Relaxation/drug effects , Muscle Strength/drug effects , Neuromuscular Blockade , Neuromuscular Blocking Agents
SELECTION OF CITATIONS
SEARCH DETAIL
...