Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Cells ; 13(13)2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38995007

ABSTRACT

Primary cilia are finger-like sensory organelles that extend from the bodies of most cell types and have a distinct lipid and protein composition from the plasma membrane. This partitioning is maintained by a diffusion barrier that restricts the entry of non-ciliary proteins, and allows the selective entry of proteins harboring a ciliary targeting sequence (CTS). However, CTSs are not stereotyped and previously reported sequences are insufficient to drive efficient ciliary localisation across diverse cell types. Here, we describe a short peptide sequence that efficiently targets transmembrane proteins to primary cilia in all tested cell types, including human neurons. We generate human-induced pluripotent stem cell (hiPSC) lines stably expressing a transmembrane construct bearing an extracellular HaloTag and intracellular fluorescent protein, which enables the bright, specific labeling of primary cilia in neurons and other cell types to facilitate studies of cilia in health and disease. We demonstrate the utility of this resource by developing an image analysis pipeline for the automated measurement of primary cilia to detect changes in their length associated with altered signaling or disease state.


Subject(s)
Cilia , Induced Pluripotent Stem Cells , Membrane Proteins , Cilia/metabolism , Humans , Membrane Proteins/metabolism , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Animals , Neurons/metabolism , Amino Acid Sequence , Cell Line , Protein Transport
2.
Curr Biol ; 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38944034

ABSTRACT

Rapid eye movement (REM) sleep has been hypothesized to promote emotional resilience, but any neuronal circuits mediating this have not been identified. We find that in mice, somatostatin (Som) neurons in the entopeduncular nucleus (EPSom)/internal globus pallidus are predominantly active during REM sleep. This unique REM activity is both necessary and sufficient for maintaining normal REM sleep. Inhibiting or exciting EPSom neurons reduced or increased REM sleep duration, respectively. Activation of the sole downstream target of EPSom neurons, Vglut2 cells in the lateral habenula (LHb), increased sleep via the ventral tegmental area (VTA). A simple chemogenetic scheme to periodically inhibit the LHb over 4 days selectively removed a significant amount of cumulative REM sleep. Chronic, but not acute, REM reduction correlated with mice becoming anxious and more sensitive to aversive stimuli. Therefore, we suggest that cumulative REM sleep, in part generated by the EP → LHb → VTA circuit identified here, could contribute to stabilizing reactions to habitual aversive stimuli.

4.
Front Neurosci ; 15: 709825, 2021.
Article in English | MEDLINE | ID: mdl-34720852

ABSTRACT

When mice are exposed to external warmth, nitric oxide synthase (NOS1) neurons in the median and medial preoptic (MnPO/MPO) hypothalamus induce sleep and concomitant body cooling. However, how these neurons regulate baseline sleep and body temperature is unknown. Using calcium photometry, we show that NOS1 neurons in MnPO/MPO are predominantly NREM and REM active, especially at the boundary of wake to NREM transitions, and in the later parts of REM bouts, with lower activity during wakefulness. In addition to releasing nitric oxide, NOS1 neurons in MnPO/MPO can release GABA, glutamate and peptides. We expressed tetanus-toxin light-chain in MnPO/MPO NOS1 cells to reduce vesicular release of transmitters. This induced changes in sleep structure: over 24 h, mice had less NREM sleep in their dark (active) phase, and more NREM sleep in their light (sleep) phase. REM sleep episodes in the dark phase were longer, and there were fewer REM transitions between other vigilance states. REM sleep had less theta power. Mice with synaptically blocked MnPO/MPO NOS1 neurons were also warmer than control mice at the dark-light transition (ZT0), as well as during the dark phase siesta (ZT16-20), where there is usually a body temperature dip. Also, at this siesta point of cooled body temperature, mice usually have more NREM, but mice with synaptically blocked MnPO/MPO NOS1 cells showed reduced NREM sleep at this time. Overall, MnPO/MPO NOS1 neurons promote both NREM and REM sleep and contribute to chronically lowering body temperature, particularly at transitions where the mice normally enter NREM sleep.

5.
Mol Psychiatry ; 26(9): 5213-5228, 2021 09.
Article in English | MEDLINE | ID: mdl-32555422

ABSTRACT

The ventral tegmental area (VTA), an important source of dopamine, regulates goal- and reward-directed and social behaviors, wakefulness, and sleep. Hyperactivation of dopamine neurons generates behavioral pathologies. But any roles of non-dopamine VTA neurons in psychiatric illness have been little explored. Lesioning or chemogenetically inhibiting VTA GABAergic (VTAVgat) neurons generated persistent wakefulness with mania-like qualities: locomotor activity was increased; sensitivity to D-amphetamine was heightened; immobility times decreased on the tail suspension and forced swim tests; and sucrose preference increased. Furthermore, after sleep deprivation, mice with lesioned VTAVgat neurons did not catch up on lost sleep, even though they were starting from a sleep-deprived baseline, suggesting that sleep homeostasis was bypassed. The mania-like behaviors, including the sleep loss, were reversed by valproate, and re-emerged when treatment was stopped. Lithium salts and lamotrigine, however, had no effect. Low doses of diazepam partially reduced the hyperlocomotion and fully recovered the immobility time during tail suspension. The mania like-behaviors mostly depended on dopamine, because giving D1/D2/D3 receptor antagonists reduced these behaviors, but also partially on VTAVgat projections to the lateral hypothalamus (LH). Optically or chemogenetically inhibiting VTAVgat terminals in the LH elevated locomotion and decreased immobility time during the tail suspension and forced swimming tests. VTAVgat neurons help set an animal's (and perhaps human's) mental and physical activity levels. Inputs inhibiting VTAVgat neurons intensify wakefulness (increased activity, enhanced alertness and motivation), qualities useful for acute survival. In the extreme, however, decreased or failed inhibition from VTAVgat neurons produces mania-like qualities (hyperactivity, hedonia, decreased sleep).


Subject(s)
GABAergic Neurons , Ventral Tegmental Area , Animals , Dopaminergic Neurons , Hypothalamic Area, Lateral , Mania , Mice
6.
Curr Opin Physiol ; 15: 7-13, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32617439

ABSTRACT

In homeothermic animals sleep preparatory behaviours often promote thermal efficiency, including warmth-seeking, adopting particular postures (curling up, head tucking) and nest building, all promoting warmer skin microclimates. Skin warmth induces NREM sleep and body cooling via circuitry that connects skin sensation to the preoptic hypothalamus. Coupling sleep induction and lower body temperature could serve to minimise energy expenditure or allow energy reallocation. Cooling during NREM sleep may also induce transcriptional changes in genes whose products facilitate housekeeping functions or measure the time spent sleeping.

7.
Curr Biol ; 29(19): 3315-3322.e3, 2019 10 07.
Article in English | MEDLINE | ID: mdl-31543455

ABSTRACT

Our urge to sleep increases with time spent awake, until sleep becomes inescapable. The sleep following sleep deprivation is longer and deeper, with an increased power of delta (0.5-4 Hz) oscillations, a phenomenon termed sleep homeostasis [1-4]. Although widely expressed genes regulate sleep homeostasis [1, 4-10] and the process is tracked by somnogens and phosphorylation [1, 3, 7, 11-14], at the circuit level sleep homeostasis has remained mysterious. Previously, we found that sedation induced with α2-adrenergic agonists (e.g., dexmedetomidine) and sleep homeostasis both depend on the preoptic (PO) hypothalamus [15, 16]. Dexmedetomidine, increasingly used for long-term sedation in intensive care units [17], induces a non-rapid-eye-movement (NREM)-like sleep but with undesirable hypothermia [18, 19]. Within the PO, various neuronal subtypes (e.g., GABA/galanin and glutamate/NOS1) induce NREM sleep [20-22] and concomitant body cooling [21, 22]. This could be because NREM sleep's restorative effects depend on lower body temperature [23, 24]. Here, we show that mice with lesioned PO galanin neurons have reduced sleep homeostasis: in the recovery sleep following sleep deprivation there is a diminished increase in delta power, and the mice catch up little on lost sleep. Furthermore, dexmedetomidine cannot induce high-power delta oscillations or sustained hypothermia. Some hours after dexmedetomidine administration to wild-type mice there is a rebound in delta power when they enter normal NREM sleep, reminiscent of emergence from torpor. This delta rebound is reduced in mice lacking PO galanin neurons. Thus, sleep homeostasis and dexmedetomidine-induced sedation require PO galanin neurons and likely share common mechanisms.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/pharmacology , Dexmedetomidine/pharmacology , Galanin/metabolism , Hypnotics and Sedatives/pharmacology , Neurons/physiology , Sleep Deprivation/metabolism , Sleep/physiology , Animals , Female , Homeostasis , Male , Mice , Neurons/drug effects , Sleep/drug effects
9.
Front Neurosci ; 13: 336, 2019.
Article in English | MEDLINE | ID: mdl-31105512

ABSTRACT

Mammals have evolved a range of behavioural and neurological mechanisms that coordinate cycles of thermoregulation and sleep. Whether diurnal or nocturnal, sleep onset and a reduction in core temperature occur together. Non-rapid eye movement (NREM) sleep episodes are also accompanied by core and brain cooling. Thermoregulatory behaviours, like nest building and curling up, accompany this circadian temperature decline in preparation for sleeping. This could be a matter of simply comfort as animals seek warmth to compensate for lower temperatures. However, in both humans and other mammals, direct skin warming can shorten sleep-latency and promote NREM sleep. We discuss the evidence that body cooling and sleep are more fundamentally connected and that thermoregulatory behaviours, prior to sleep, form warm microclimates that accelerate NREM directly through neuronal circuits. Paradoxically, this warmth might also induce vasodilation and body cooling. In this way, warmth seeking and nesting behaviour might enhance the circadian cycle by activating specific circuits that link NREM initiation to body cooling. We suggest that these circuits explain why NREM onset is most likely when core temperature is at its steepest rate of decline and why transitions to NREM are accompanied by a decrease in brain temperature. This connection may have implications for energy homeostasis and the function of sleep.

10.
Sleep ; 42(5)2019 05 01.
Article in English | MEDLINE | ID: mdl-30722053

ABSTRACT

Acute chemogenetic inhibition of histamine (HA) neurons in adult mice induced nonrapid eye movement (NREM) sleep with an increased delta power. By contrast, selective genetic lesioning of HA neurons with caspase in adult mice exhibited a normal sleep-wake cycle overall, except at the diurnal start of the lights-off period, when they remained sleepier. The amount of time spent in NREM sleep and in the wake state in mice with lesioned HA neurons was unchanged over 24 hr, but the sleep-wake cycle was more fragmented. Both the delayed increase in wakefulness at the start of the night and the sleep-wake fragmentation are similar phenotypes to histidine decarboxylase knockout mice, which cannot synthesize HA. Chronic loss of HA neurons did not affect sleep homeostasis after sleep deprivation. However, the chronic loss of HA neurons or chemogenetic inhibition of HA neurons did notably reduce the ability of the wake-promoting compound modafinil to sustain wakefulness. Thus, part of modafinil's wake-promoting actions arise through the HA system.


Subject(s)
Histamine/genetics , Modafinil/therapeutic use , Neurons/drug effects , Sleep Deprivation/genetics , Wakefulness-Promoting Agents/therapeutic use , Wakefulness/drug effects , Animals , Electroencephalography/drug effects , Electroencephalography/methods , Genetic Vectors/administration & dosage , Histamine/deficiency , Homeostasis/drug effects , Homeostasis/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Modafinil/pharmacology , Neurons/physiology , Sleep/drug effects , Sleep/physiology , Sleep Deprivation/drug therapy , Sleep Deprivation/metabolism , Wakefulness/physiology , Wakefulness-Promoting Agents/pharmacology
11.
Nat Neurosci ; 22(1): 106-119, 2019 01.
Article in English | MEDLINE | ID: mdl-30559475

ABSTRACT

We screened for novel circuits in the mouse brain that promote wakefulness. Chemogenetic activation experiments and electroencephalogram recordings pointed to glutamatergic/nitrergic (NOS1) and GABAergic neurons in the ventral tegmental area (VTA). Activating glutamatergic/NOS1 neurons, which were wake- and rapid eye movement (REM) sleep-active, produced wakefulness through projections to the nucleus accumbens and the lateral hypothalamus. Lesioning the glutamate cells impaired the consolidation of wakefulness. By contrast, activation of GABAergic VTA neurons elicited long-lasting non-rapid-eye-movement-like sleep resembling sedation. Lesioning these neurons produced an increase in wakefulness that persisted for at least 4 months. Surprisingly, these VTA GABAergic neurons were wake- and REM sleep-active. We suggest that GABAergic VTA neurons may limit wakefulness by inhibiting the arousal-promoting VTA glutamatergic and/or dopaminergic neurons and through projections to the lateral hypothalamus. Thus, in addition to its contribution to goal- and reward-directed behaviors, the VTA has a role in regulating sleep and wakefulness.


Subject(s)
GABAergic Neurons/physiology , Glutamic Acid/metabolism , Neurons/physiology , Sleep/physiology , Ventral Tegmental Area/physiology , Wakefulness/physiology , Animals , Dopaminergic Neurons/physiology , Mice , Nitric Oxide Synthase Type I/metabolism , Sleep, REM/physiology , Ventral Tegmental Area/metabolism
12.
Curr Biol ; 28(14): 2263-2273.e4, 2018 07 23.
Article in English | MEDLINE | ID: mdl-30017485

ABSTRACT

Mammals, including humans, prepare for sleep by nesting and/or curling up, creating microclimates of skin warmth. To address whether external warmth induces sleep through defined circuitry, we used c-Fos-dependent activity tagging, which captures populations of activated cells and allows them to be reactivated to test their physiological role. External warming tagged two principal groups of neurons in the median preoptic (MnPO)/medial preoptic (MPO) hypothalamic area. GABA neurons located mainly in MPO produced non-rapid eye movement (NREM) sleep but no body temperature decrease. Nitrergic-glutamatergic neurons in MnPO-MPO induced both body cooling and NREM sleep. This circuitry explains how skin warming induces sleep and why the maximal rate of core body cooling positively correlates with sleep onset. Thus, the pathways that promote NREM sleep, reduced energy expenditure, and body cooling are inextricably linked, commanded by the same neurons. This implies that one function of NREM sleep is to lower brain temperature and/or conserve energy.


Subject(s)
Body Temperature Regulation/physiology , Neurons/physiology , Preoptic Area/physiology , Sleep/physiology , Adaptation, Physiological , Animals , Cold Temperature , Hot Temperature , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-fos/metabolism
13.
Curr Biol ; 28(4): 580-587.e5, 2018 02 19.
Article in English | MEDLINE | ID: mdl-29398217

ABSTRACT

The lateral habenula has been widely studied for its contribution in generating reward-related behaviors [1, 2]. We have found that this nucleus plays an unexpected role in the sedative actions of the general anesthetic propofol. The lateral habenula is a glutamatergic, excitatory hub that projects to multiple targets throughout the brain, including GABAergic and aminergic nuclei that control arousal [3-5]. When glutamate release from the lateral habenula in mice was genetically blocked, the ability of propofol to induce sedation was greatly diminished. In addition to this reduced sensitivity to propofol, blocking output from the lateral habenula caused natural non-rapid eye movement (NREM) sleep to become highly fragmented, especially during the rest ("lights on") period. This fragmentation was largely reversed by the dual orexinergic antagonist almorexant. We conclude that the glutamatergic output from the lateral habenula is permissive for the sedative actions of propofol and is also necessary for the consolidation of natural sleep.


Subject(s)
Glutamic Acid/metabolism , Habenula/drug effects , Hypnotics and Sedatives/pharmacology , Neural Pathways/drug effects , Propofol/pharmacology , Anesthetics, Intravenous/metabolism , Animals , HEK293 Cells , Habenula/physiology , Humans , Male , Mice , Neural Pathways/physiology
14.
J Neurosci ; 36(44): 11171-11184, 2016 11 02.
Article in English | MEDLINE | ID: mdl-27807161

ABSTRACT

Zolpidem, a GABAA receptor-positive modulator, is the gold-standard drug for treating insomnia. Zolpidem prolongs IPSCs to decrease sleep latency and increase sleep time, effects that depend on α2 and/or α3 subunit-containing receptors. Compared with natural NREM sleep, zolpidem also decreases the EEG power, an effect that depends on α1 subunit-containing receptors, and which may make zolpidem-induced sleep less optimal. In this paper, we investigate whether zolpidem needs to potentiate only particular GABAergic pathways to induce sleep without reducing EEG power. Mice with a knock-in F77I mutation in the GABAA receptor γ2 subunit gene are zolpidem-insensitive. Using these mice, GABAA receptors in the frontal motor neocortex and hypothalamic (tuberomammillary nucleus) histaminergic-neurons of γ2I77 mice were made selectively sensitive to zolpidem by genetically swapping the γ2I77 subunits with γ2F77 subunits. When histamine neurons were made selectively zolpidem-sensitive, systemic administration of zolpidem shortened sleep latency and increased sleep time. But in contrast to the effect of zolpidem on wild-type mice, the power in the EEG spectra of NREM sleep was not decreased, suggesting that these EEG power-reducing effects of zolpidem do not depend on reduced histamine release. Selective potentiation of GABAA receptors in the frontal cortex by systemic zolpidem administration also reduced sleep latency, but less so than for histamine neurons. These results could help with the design of new sedatives that induce a more natural sleep. SIGNIFICANCE STATEMENT: Many people who find it hard to get to sleep take sedatives. Zolpidem (Ambien) is the most widely prescribed "sleeping pill." It makes the inhibitory neurotransmitter GABA work better at its receptors throughout the brain. The sleep induced by zolpidem does not resemble natural sleep because it produces a lower power in the brain waves that occur while we are sleeping. We show using mouse genetics that zolpidem only needs to work on specific parts and cell types of the brain, including histamine neurons in the hypothalamus, to induce sleep but without reducing the power of the sleep. This knowledge could help in the design of sleeping pills that induce a more natural sleep.


Subject(s)
Neocortex/physiology , Neurons/physiology , Pyridines/administration & dosage , Receptors, GABA-A/metabolism , Sleep/drug effects , Sleep/physiology , Animals , Dose-Response Relationship, Drug , Female , Histamine Agents/administration & dosage , Hypnotics and Sedatives/administration & dosage , Male , Mice , Mice, Inbred C57BL , Neocortex/cytology , Neocortex/drug effects , Neurons/cytology , Neurons/drug effects , Sleep Aids, Pharmaceutical/administration & dosage , Zolpidem
15.
Nat Neurosci ; 18(4): 553-561, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25706476

ABSTRACT

Do sedatives engage natural sleep pathways? It is usually assumed that anesthetic-induced sedation and loss of righting reflex (LORR) arise by influencing the same circuitry to lesser or greater extents. For the α2 adrenergic receptor agonist dexmedetomidine, we found that sedation and LORR were in fact distinct states, requiring different brain areas: the preoptic hypothalamic area and locus coeruleus (LC), respectively. Selective knockdown of α2A adrenergic receptors from the LC abolished dexmedetomidine-induced LORR, but not sedation. Instead, we found that dexmedetomidine-induced sedation resembled the deep recovery sleep that follows sleep deprivation. We used TetTag pharmacogenetics in mice to functionally mark neurons activated in the preoptic hypothalamus during dexmedetomidine-induced sedation or recovery sleep. The neuronal ensembles could then be selectively reactivated. In both cases, non-rapid eye movement sleep, with the accompanying drop in body temperature, was recapitulated. Thus, α2 adrenergic receptor-induced sedation and recovery sleep share hypothalamic circuitry sufficient for producing these behavioral states.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/pharmacology , Deep Sedation , Dexmedetomidine/pharmacology , Hypnotics and Sedatives/pharmacology , Hypothalamus/drug effects , Sleep/drug effects , Animals , Electroencephalography , Hypothalamus/physiology , Hypothermia/chemically induced , Locus Coeruleus/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pharmacogenetics
17.
Glycobiology ; 24(12): 1291-300, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25028392

ABSTRACT

Trehalose dimycolate, an unusual glycolipid in the outer membrane of Mycobacterium tuberculosis, stimulates macrophages by binding to the macrophage receptor mincle. This stimulation plays an important role both in infection by mycobacteria and in the use of derivatives of mycobacteria as adjuvants to enhance the immune response. The mechanism of trehalose dimycolate binding to the C-type carbohydrate-recognition domain in human mincle has been investigated using a series of synthetic analogs of trehalose dimycolate and site-directed mutagenesis of the human protein. The results support a mechanism of binding acylated trehalose derivatives to human mincle that is very similar to the mechanism of binding to bovine mincle, in which one glucose residue in the trehalose headgroup of the glycolipid is ligated to the principle Ca(2+)-binding site in the carbohydrate-recognition domain, with specificity for the disaccharide resulting from interactions with the second glucose residue. Acyl chains attached to the 6-OH groups of trehalose enhance affinity, with the affinity dependent on the length of the acyl chains and the presence of a hydrophobic groove adjacent to the sugar-binding sites. The results indicate that the available crystal structure of the carbohydrate-recognition domain of human mincle is unlikely to be in a fully active conformation. Instead, the ligand-binding conformation probably resembles closely the structure observed for bovine mincle in complex with trehalose. These studies provide a basis for targeting human mincle as a means of inhibiting interactions with mycobacteria and as an approach to harnessing the ability of mincle to stimulate the immune response.


Subject(s)
Cord Factors/chemistry , Lectins, C-Type/chemistry , Mycobacterium tuberculosis/chemistry , Receptors, Immunologic/chemistry , Animals , Binding Sites , Calcium/chemistry , Cattle , Cord Factors/chemical synthesis , Humans , Hydrogen-Ion Concentration , Lectins, C-Type/metabolism , Models, Molecular , Protein Conformation , Receptors, Immunologic/metabolism
18.
PLoS Negl Trop Dis ; 5(8): e1234, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21829735

ABSTRACT

BACKGROUND: The clinical signs of active trachoma are often present in the absence of ocular Chlamydia trachomatis infection in low prevalence and mass treated settings. Treatment decisions are currently based on the prevalence of clinical signs, and this may result in the unnecessary distribution of mass antibiotic treatment. We aimed to evaluate the diagnostic accuracy of a prototype point-of-care (POC) test, developed for field diagnosis of ocular C. trachomatis, in low prevalence settings of The Gambia and Senegal. METHODOLOGY/PRINCIPAL FINDINGS: Three studies were conducted, two in The Gambia and one in Senegal. Children under the age of 10 years were screened for the clinical signs of trachoma. Two ocular swabs were taken from the right eye. The first swab was tested by the POC test in the field and the result independently graded by two readers. The second swab was tested for the presence of C. trachomatis by Amplicor Polymerase Chain Reaction. In Senegal, measurements of humidity and temperature in the field were taken. A total of 3734 children were screened, 950 in the first and 1171 in the second Gambian study, and 1613 in Senegal. The sensitivity of the prototype POC test ranged between 33.3-67.9%, the specificity between 92.4-99.0%, the positive predictive value between 4.3-21.0%, and the negative predictive value between 98.0-99.8%. The rate of false-positives increased markedly at temperatures above 31.4°C and relative humidities below 11.4%. CONCLUSIONS/SIGNIFICANCE: In its present format, this prototype POC test is not suitable for field diagnosis of ocular C. trachomatis as its specificity decreases in hot and dry conditions: the environment in which trachoma is predominantly found. In the absence of a suitable test for infection, trachoma diagnosis remains dependent on clinical signs. Under current WHO recommendations, this is likely resulting in the continued mass treatment of non-infected communities.


Subject(s)
Chlamydia trachomatis/isolation & purification , Point-of-Care Systems , Reagent Kits, Diagnostic , Trachoma/diagnosis , Child , Child, Preschool , Eye/microbiology , False Positive Reactions , Gambia , Humans , Humidity , Infant , Logistic Models , Polymerase Chain Reaction , Predictive Value of Tests , Reproducibility of Results , Senegal , Temperature , Trachoma/microbiology
19.
Inhal Toxicol ; 22(8): 669-78, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20462393

ABSTRACT

Particles depositing on alveolar walls is of concern due to their potential to migrate through the blood-gas barrier. Whole-lung dosimetry models do not account for the flow field inside the alveoli and therefore may not accurately describe alveolar deposition. Studies that quantify the flow patterns in realistic geometries are limited and results inconsistent. This study aims to better understand the fluid characteristics in the terminal air sacs; specifically, alveolar mouth to depth flow rate ratio, penetration depth of residual air, and diffusive versus convective particle motion. A terminating alveolar sac with expanding alveolar walls was constructed using 13 truncated sphere-shaped alveoli, with dimensions consistent with published morphometry data. The flow field was governed by a measured in vivo breathing curve for normal volumes over periods of 2 and 4 seconds, analyzed numerically and compared to previous literature. Recirculation was not present, consistent with prior studies. Flow rate ratios (0.18-0.36) were within the range (0.057-1) previously reported. Penetration depths were less than 33% into the air sac during inhalation, decreasing in length for air inside the sac to zero near the wall. Péclet numbers indicated diffusion dominated flow for all submicron-sized particles. However, convection was significant at the duct entrance for particles >0.5 micron and inside the sac for particles >1 micron. Wall motion induced convection may not always be negligible, and if neglected could affect the accuracy of deposition predictions for certain particle sizes and flow conditions.


Subject(s)
Air Pollutants/pharmacokinetics , Air Sacs/physiology , Particulate Matter/pharmacokinetics , Pulmonary Alveoli/physiology , Rheology/methods , Air Sacs/anatomy & histology , Animals , Convection , Humans , Inhalation Exposure , Mice , Microscopy, Electron, Scanning , Models, Anatomic , Models, Biological , Particle Size , Pulmonary Alveoli/anatomy & histology , Rats , Respiration , Spirometry
20.
Vet Anaesth Analg ; 32(4): 184-92, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16008715

ABSTRACT

OBJECTIVE: To compare the peri- and post-operative (72 hours) analgesic effects of injectable and orally administered carprofen and meloxicam for ovariohysterectomy in dogs. STUDY DESIGN: Prospective, randomized clinical study. ANIMALS: Forty-three dogs undergoing elective ovariohysterectomy. MATERIALS AND METHODS: Dogs were randomly assigned to receive pre-operative carprofen, meloxicam or sterile saline by subcutaneous injection. Pre-anaesthetic medication was intramuscular acepromazine (0.02 mg kg(-1)) and methadone (0.2 mg kg(-1)). Anaesthesia was induced with either thiopentone or propofol injected to effect, and maintained with isoflurane in oxygen. Visual analogue scores (VAS) for pain and sedation were recorded at 1, 2, 3, 4 and 6 hours following tracheal extubation. Oral medication with the same treatment was continued post-operatively for 3 days, with VAS scores for pain being recorded before, and 2 hours after treatment on each day. Differences between group age, body mass, duration of general anaesthesia, time from treatment injection to tracheal extubation and time from treatment injection to first oral treatment were analysed using one-way analysis of variance and Kruskal-Wallis test. Visual analogue scores for pain and sedation were analysed using a re-randomization method. The significance level was set at p < 0.05. RESULTS: Meloxicam-treated subjects had lower mean VAS than the control group at 2 and 6 hours following tracheal extubation. Control group VAS were more varied than meloxicam scores (at 6 hours) and carprofen scores (at 3 and 6 hours). On the first post-operative day, pre- to post-treatment VAS scores decreased significantly after meloxicam. On day 3, scores in the meloxicam-treated group were significantly lower than control values after treatment. Changes in pre- to post-treatment VAS were greater in animals receiving either meloxicam or carprofen compared with those given saline. CONCLUSIONS AND CLINICAL RELEVANCE: Both carprofen and meloxicam provided satisfactory analgesia for 72 hours following ovariohysterectomy in dogs.


Subject(s)
Analgesics, Non-Narcotic/administration & dosage , Carbazoles/administration & dosage , Dogs/surgery , Pain, Postoperative/veterinary , Thiazines/administration & dosage , Thiazoles/administration & dosage , Anesthesia/veterinary , Animals , Dogs/physiology , Female , Hysterectomy/veterinary , Meloxicam , Ovariectomy/veterinary , Pain Measurement/veterinary , Pain, Postoperative/prevention & control , Prospective Studies , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...