Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Sleep Adv ; 4(1): zpad039, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37954093

RESUMEN

Herein the major accomplishments, trials and tribulations, and epiphanies experienced by James M. Krueger over the course of his career in sleep research are presented. They include the characterization of a) the supranormal EEG delta waves occurring during NREMS post sleep loss, b) Factor S as a muramyl peptide, c) the physiological roles of cytokines in sleep regulation, d) multiple other sleep regulatory substances, e) the dramatic changes in sleep over the course of infectious diseases, and f) sleep initiation within small neuronal/glial networks. The theory that the preservation of brain plasticity is the primordial sleep function is briefly discussed. These accomplishments resulted from collaborations with many outstanding scientists including James M. Krueger's mentors (John Pappenheimer and Manfred Karnovsky) and collaborators later in life, including Charles Dinarello, Louis Chedid, Mark Opp, Ferenc Obal jr., Dave Rector, Ping Taishi, Linda Toth, Jeannine Majde, Levente Kapas, Eva Szentirmai, Jidong Fang, Chris Davis, Sandip Roy, Tetsuya Kushikata, Fabio Garcia-Garcia, Ilia Karatsoreos, Mark Zielinski, and Alok De, plus many students, e.g. Jeremy Alt, Kathryn Jewett, Erika English, and Victor Leyva-Grado.

2.
Neurobiol Sleep Circadian Rhythms ; 10: 100063, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33748539

RESUMEN

Night shift work is a risk factor for viral infection, suggesting that night shift schedules compromise host defense mechanisms. Prior studies have investigated changes in the temporal profiles of circulating cytokines important for priming and restraining the immune response to infectious challenges from night shift work, but not by way of a 24-h constant routine of continuous wakefulness devoid of behavioral or environmental influences. Hence the true endogenous pattern of cytokines, and the combined effect of sleep loss and circadian misalignment on these cytokines remains unknown. Here, 14 healthy young men and women underwent three days of either a simulated night shift or a simulated day shift schedule under dim light in a controlled in-laboratory environment. This was followed by a 24-h constant routine protocol during which venous blood was collected at 3-h intervals. Those who had been in the night shift schedule showed lower mean circulating TNF-α (t13 = -6.03, p < 0.001), without any significant differences in IL-1ß, IL-8 and IL-10, compared with those who had been in the day shift (i.e., control) schedule. Furthermore, circulating IL-6 increased with time awake in both shift work conditions (t13 = 6.03, p < 0.001), such that temporal changes in IL-6 were markedly shifted relative to circadian clock time in the night shift condition. These results indicate that night shift work compromises host defense by creating cytokine conditions that initially impede anti-viral immunity (lower TNF-α) and may eventually promote autoimmunity (mistimed rise in IL-6).

3.
Brain Behav Immun ; 94: 245-258, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33571627

RESUMEN

Tumor necrosis factor alpha (TNF) has sleep regulatory and brain development roles. TNF promotes sleep in vivo and in vitro while TNF inhibition diminishes sleep. Transmembrane (tm) TNF and the tmTNF receptors (Rs), are cleaved by tumor necrosis factor alpha convertase to produce soluble (s) TNF and sTNFRs. Reverse signaling occurs in cells expressing tmTNF upon sTNFR binding. sTNFR administration in vivo inhibits sleep, thus we hypothesized that a wake-like state in vitro would be induced by sTNFR-tmTNF reverse signaling. Somatosensory cortical neuron/glia co-cultures derived from male and female mice lacking both TNFRs (TNFRKO), or lacking TNF (TNFKO) and wildtype (WT) mice were plated onto six-well multi-electrode arrays. Daily one-hour electrophysiological recordings were taken on culture days 4 through 14. sTNFR1 (0.0, 0.3, 3, 30, 60, and 120 ng/µL) was administered on day 14. A final one-hour recording was taken on day 15. Four measures were characterized that are also used to define sleep in vivo: action potentials (APs), burstiness index (BI), synchronization of electrical activity (SYN), and slow wave power (SWP; 0.25-3.75 Hz). Development rates of these emergent electrophysiological properties increased in cells from mice lacking TNF or both TNFRs compared to cells from WT mice. Decreased SWP, after the three lowest doses (0.3, 3 and 30 ng/µL) of the sTNFR1, indicate a wake-like state in cells from TNFRKO mice. A wake-like state was also induced after 3 ng/µl sTNFR1 treatment in cells from TNFKO mice, which express the TNFR1 ligand, lymphotoxin alpha. Cells from WT mice showed no treatment effects. Results are consistent with prior studies demonstrating involvement of TNF in brain development, TNF reverse signaling, and sleep regulation in vivo. Further, the current demonstration of sTNFR1 induction of a wake-like state in vitro is consistent with the idea that small neuronal/glial circuits manifest sleep- and wake-like states analogous to those occurring in vivo. Finally, that sTNF forward signaling enhances sleep while sTNFR1 reverse signaling enhances a wake-like state is consistent with other sTNF/tmTNF/sTNFR1 brain actions having opposing activities.


Asunto(s)
Receptores Tipo II del Factor de Necrosis Tumoral , Factor de Necrosis Tumoral alfa , Animales , Femenino , Masculino , Ratones , Neuroglía , Neuronas , Receptores Tipo I de Factores de Necrosis Tumoral , Transducción de Señal
4.
Artículo en Inglés | MEDLINE | ID: mdl-32529121

RESUMEN

Circadian rhythms evolved within single cell organisms and serve to regulate rest-activity cycles in most single-cell and multiple-cell organisms. In contrast, sleep is a network emergent property found in animals with a nervous system. Rhythms and sleep are much entangled involving shared regulatory molecules such as adenosine, ATP, cytokines, neurotrophins, and nitric oxide. These molecules are activity-dependent and act locally to initiate regulatory events involved in rhythms, sleep, and plasticity.

5.
J Appl Physiol (1985) ; 128(6): 1506-1522, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32324480

RESUMEN

Sleep regulation involves interleukin-1ß (IL1) family members, TNF, and circadian clock genes. Previously, we characterized spontaneous sleep and sleep after 8 h of sleep deprivation (SD) ending at zeitgeber time (ZT)4 and ZT16 in wild-type (WT) and IL1 receptor accessory protein (AcP)- and brain-specific AcP (AcPb)-knockout (KO) mice. Here, we applied quantitative reverse transcriptase polymerase chain reaction and Spearman gene pair expression correlation methods to characterize IL1, IL1 receptor 1 (IL1R1), AcP, AcPb, Period 1 (Per1), Clock, adenosine deaminase (Ada), peptidoglycan recognition protein 1 (Pglyrp1), and TNF mRNA expressions under conditions with distinct sleep phenotypes. In WT mice, IL1, IL1R1, AcP, Ada, and Clock mRNAs were higher at ZT4 (mid-sleep period) than at ZT16. mRNA expressions differed substantially in AcP and AcPb KO mice at those times. After SD ending at ZT4, only WT mice had a non-rapid eye movement sleep (NREMS) rebound, and AcPb and IL1R1 mRNA increases were unique to WT mice. In AcPb KO mice, which have spontaneous high EEG slow wave power, AcP and Pglyrp1 mRNAs were elevated relative to WT mice at ZT4. At ZT4, the AcPb KO - WT Spearman correlation difference networks showed high positive correlations between IL1R1 and IL1, Per1, and Clock and high negative correlations between TNF and Pglyrp1 and Ada. At ZT16, the WT mice gene pair expression network was mostly negative, whereas in AcP KO mice, which have substantially more rapid eye movement sleep than WT mice, it was all positive. We conclude that gene pair expression correlations depend on the presence of AcP and AcPb.NEW & NOTEWORTHY Spearman gene pair expression correlations depend upon the presence or absence of interleukin-1 receptor accessory protein and upon sleep phenotype.


Asunto(s)
Privación de Sueño , Sueño , Animales , Interleucina-1beta , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/genética , Receptores de Interleucina-1 , Sueño/genética , Privación de Sueño/genética
6.
J Appl Physiol (1985) ; 127(3): 770-780, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31295066

RESUMEN

Interleukin-1ß (IL1) is a sleep regulatory substance. The IL1/IL1 type 1 receptor complex requires a receptor accessory protein (AcP) to signal. There are three isoforms of AcP. In the current experiments, mice lacking a neuron-specific isoform, called AcPb knockout (AcPb KO), or mice lacking AcP + AcPb isoforms (AcP KO) or wild-type (WT) mice were used. Spontaneous sleep and sleep responses to sleep deprivation (SD) between zeitgeber time (ZT) 20-ZT4 and ZT8-ZT16 were characterized. Furthermore, somatosensory cortical protein extracts were examined for phosphorylated (p) proto-oncogene tyrosine-protein kinase sarcoma (Src) and p38MAPK levels at ZT4 and ZT16 and after SD. Spontaneous sleep was similar in the three strains, except rapid eye movement sleep (REMS) duration between ZT12-ZT16 was greater in AcP KO than WT mice. After SD at ZT4, only WT mice had non-REMS (NREMS) rebounds. All mouse strains lacked an NREMS rebound after SD at ZT16. All strains after both SD periods had REMS rebounds. AcPb KO mice, but not AcP KO mice, had greater EEG delta wave (0.5-4 Hz) power during NREMS than WT mice. p-Src was very low at ZT16 but high at ZT4, whereas p-p38MAPK was low at ZT4 and high at ZT16. p-p38MAPK levels were not sensitive to SD. In contrast, p-Src levels were less after SD at the P = 0.08 level of significance in the strains lacking AcPb. We conclude that AcPb is required for NREMS responses to sleep loss, but not for SD-induced EEG delta wave or REMS responses.NEW & NOTEWORTHY Interleukin-1ß (IL1), a well-characterized sleep regulatory substance, requires an IL1 receptor accessory protein (AcP); one of its isoforms is neuron-specific (called AcPb). We showed that in mice, AcPb is required for nonrapid eye movement sleep responses following 8 h of sleep loss ending 4 h after daybreak but did not affect rapid eye movement sleep rebound. Sleep loss reduced phosphorylation of proto-oncogene tyrosine-protein kinase sarcoma but not of the less sensitive p38MAPK, downstream IL1 signaling molecules.


Asunto(s)
Receptores de Interleucina-1/metabolismo , Privación de Sueño/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Familia-src Quinasas/metabolismo , Animales , Electroencefalografía , Homeostasis , Masculino , Ratones Noqueados , Fases del Sueño
7.
Artículo en Inglés | MEDLINE | ID: mdl-31106280

RESUMEN

Small in vitro neuronal/glial networks exhibit sleep-like states. Sleep regulatory substance interleukin-1ß (IL1) signals via its type I receptor and a receptor accessory protein (AcP). AcP has a neuron-specific isoform called AcPb. After sleep deprivation, AcPb, but not AcP, upregulates in brain, and mice lacking AcPb lack sleep rebound. Herein we used action potentials (APs), AP burstiness, synchronization of electrical activity (SYN), and delta wave (0.5-3.75 Hz) power to characterize cortical culture network state. Homologous parameters are used in vivo to characterize sleep. Cortical cells from 1-2-day-old pups from AcP knockout (KO, lacking both AcP and AcPb), AcPb KO (lacking only AcPb), and wild type (WT) mice were cultured separately on multi-electrode arrays. Recordings of spontaneous activity were taken each day during days 4-14 in vitro. In addition, cultures were treated with IL1, or in separate experiments, stimulated electrically to determine evoked response potentials (ERPs). In AcP KO cells, the maturation of network properties accelerated compared to those from cells lacking only AcPb. In contrast, the lack of AcPb delayed spontaneous network emergence of sleep-linked properties. The addition of IL1 enhanced delta wave power in WT cells but not in AcP KO or AcPb KO cells. The ontology of electrically-induced ERPs was delayed in AcP KO cells. We conclude IL1 signaling has a critical role in the emergence of sleep-linked network behavior with AcP playing a dominant role in the slowing of development while AcPb enhances development rates of sleep-linked emergent network properties.

8.
Sleep Med Rev ; 43: 14-21, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30502497

RESUMEN

The historic sleep regulatory paradigm invokes "top-down" imposition of sleep on the brain by sleep regulatory circuits. While remaining conceptually useful, many sleep phenomena are difficult to explain using that paradigm, including, unilateral sleep, sleep-walking, and poor performance after sleep deprivation. Further, all animals sleep after non-lethal brain lesions, regardless of whether the lesion includes sleep regulatory circuits, suggesting that sleep is a fundamental property of small viable neuronal/glial networks. That small areas of the brain can exhibit non-rapid eye movement sleep-like states is summarized. Further, sleep-like states in neuronal/glial cultures are described. The local sleep states, whether in vivo or in vitro, share electrophysiological properties and molecular regulatory components with whole animal sleep and exhibit sleep homeostasis. The molecular regulatory components of sleep are also involved in plasticity and inflammation. Like sleep, these processes, are initiated by local cell-activity dependent events, yet have at higher levels of tissue organization whole body functions. While there are large literatures dealing with local initiation and regulation of plasticity and inflammation, the literature surrounding local sleep is in its infancy and clinical applications of the local sleep concept are absent. Regardless, the local use-dependent sleep paradigm can advise and advance future research and clinical applications.


Asunto(s)
Modelos Neurológicos , Red Nerviosa/fisiología , Neuroglía/fisiología , Neuronas/fisiología , Sueño/fisiología , Animales , Encéfalo/fisiología , Homeostasis , Humanos , Técnicas In Vitro , Privación de Sueño
9.
Sleep Med Rev ; 40: 69-78, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29153862

RESUMEN

This review details tumor necrosis factor alpha (TNF) biology and its role in sleep, and describes how TNF medications influence sleep/wake activity. Substantial evidence from healthy young animals indicates acute enhancement or inhibition of endogenous brain TNF respectively promotes and inhibits sleep. In contrast, the role of TNF in sleep in most human studies involves pathological conditions associated with chronic elevations of systemic TNF and disrupted sleep. Normalization of TNF levels in such patients improves sleep. A few studies involving normal healthy humans and their TNF levels and sleep are consistent with the animal studies but are necessarily more limited in scope. TNF can act on established sleep regulatory circuits to promote sleep and on the cortex within small networks, such as cortical columns, to induce sleep-like states. TNF affects multiple synaptic functions, e.g., its role in synaptic scaling is firmly established. The TNF-plasticity actions, like its role in sleep, can be local network events suggesting that sleep and plasticity share biochemical regulatory mechanisms and thus may be inseparable from each other. We conclude that TNF is involved in sleep regulation acting within an extensive tightly orchestrated biochemical network to niche-adapt sleep in health and disease.


Asunto(s)
Encéfalo/fisiología , Sueño/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Animales , Humanos , Plasticidad Neuronal , Factor de Necrosis Tumoral alfa/sangre
10.
Artículo en Inglés | MEDLINE | ID: mdl-28070566

RESUMEN

Multiple interactions between the immune system and sleep are known, including the effects of microbial challenge on sleep or the effects of sleep loss on facets of the immune response. Cytokines regulate, in part, sleep and immune responses. Here we examine the role of an anti-inflammatory cytokine, interleukin-37 (IL-37) on sleep in a mouse strain that expresses human IL-37b (IL37tg mice). Constitutive expression of the IL-37 gene in the brains of these mice under resting conditions is low; however, upon an inflammatory stimulus, expression increases dramatically. We measured sleep in three conditions; a) under baseline conditions and after 6 h of sleep loss, b) after bolus intraperitoneal administration of lipopolysaccharide (LPS) or IL-1ß and c) after intranasal influenza virus challenge. Under baseline conditions, the IL37tg mice had 7% more spontaneous non-rapid eye movement sleep (NREMS) during the light period than wild-type (WT) mice. After sleep deprivation both WT mice and IL37tg mice slept an extra 21% and 12%, respectively, during the first 6 h of recovery. NREMS responses after sleep deprivation did not significantly differ between WT mice and IL37tg mice. However, in response to either IL-1ß or LPS, the increases in time spent in NREMS were about four-fold greater in the WT mice than in the IL37tg mice. In contrast, in response to a low dose of mouse-adapted H1N1 influenza virus, sleep responses developed slowly over the 6 day recording period. By day 6, NREMS increased by 10% and REMS increased by 18% in the IL37tg mice compared to the WT mice. Further, by day 4 IL37tg mice lost less weight, remained more active, and retained their body temperatures closer to baseline values than WT mice. We conclude that conditions that promote IL-37 expression attenuate morbidity to severe inflammatory challenge.

11.
Crit Rev Immunol ; 37(2-6): 291-315, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29773024

RESUMEN

Excessive sleepiness and fever are constitutional symptoms associated with systemic infection. Although fevers have been investigated for many years, sleep responses to infectious challenge have only recently been investigated. Inoculation of animals with bacterial, viral, protozoan and fungal organisms result in complex sleep responses dependent upon the microbial agent and route of administration. The general pattern is characterized by an initial robust increase in non-rapid eye movement sleep (NREMS) followed by a period of NREMS inhibition. REMS is inhibited after infectious challenge. The sleep responses are accompanied by fever but the two responses are, in part, independent from each other. Sleep responses, like fevers, may be beneficial to host defense although this area is relatively uninvestigated. Microbial products likely responsible for sleep and fever responses include bacterial muramyl peptides and endotoxin, and viral double stranded RNA. These microbial products induce sleep and fever responses in animal models. The exact mechanism of how these structurally diverse microbial products elicit sleep and fever remain unknown; however these substances share the ability to induce cytokine production. Cytokines such as interleukin-1 (IL-1), tumor necrosis factor, acidic fibroblast growth factor (FGF), and interferon-α (IFN-α) are somnogenic whether given directly into brain or intravenously. Other cytokines lack somnogenic activity, e.g., IL-2, IL-6, IFNß and basic FGF. The somnogenic actions of cytokines probably involve growth hormone-releasing hormone (GHRH) and nitric oxide. Anti-GHRH or inhibition of NO production inhibits normal sleep and inhibits IL-1-induced sleep. In conclusion, cytokines are likely key mediators of fever and sleep responses to infection. The microbial-cytokine altered sleep likely results from an amplification of physiological sleep mechanisms which include cytokines, several neuropeptides and neurotransmitters such as nitric oxide.


Asunto(s)
Fiebre/inmunología , Interacciones Microbiota-Huesped/inmunología , Interacciones Huésped-Parásitos/inmunología , Infecciones/inmunología , Somnolencia , Acetilmuramil-Alanil-Isoglutamina/inmunología , Acetilmuramil-Alanil-Isoglutamina/metabolismo , Animales , Encéfalo/inmunología , Encéfalo/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Endotoxinas/inmunología , Endotoxinas/metabolismo , Fiebre/microbiología , Fiebre/parasitología , Fiebre/virología , Hormona Liberadora de Hormona del Crecimiento/inmunología , Hormona Liberadora de Hormona del Crecimiento/metabolismo , Humanos , Infecciones/microbiología , Infecciones/parasitología , Infecciones/virología , Óxido Nítrico/inmunología , Óxido Nítrico/metabolismo , Sueño/inmunología
12.
Am J Physiol Regul Integr Comp Physiol ; 311(6): R1004-R1012, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27707719

RESUMEN

The ionotropic purine type 2X7 receptor (P2X7R) is a nonspecific cation channel implicated in sleep regulation and brain cytokine release. Many endogenous rhythms covary with sleep, including locomotor activity and core body temperature. Furthermore, brain-hypothalamic cytokines and purines play a role in the regulation of these physiological parameters as well as sleep. We hypothesized that these parameters are also affected by the absence of the P2X7 receptor. Herein, we determine spontaneous expression of body temperature and locomotor activity in wild-type (WT) and P2X7R knockout (KO) mice and how they are affected by sleep deprivation (SD). We also compare hypothalamic, hippocampal, and cortical cytokine- and purine-related receptor and enzyme mRNA expressions before and after SD in WT and P2X7RKO mice. Next, in a hypothesis-generating survey of hypothalamic long noncoding (lnc) RNAs, we compare lncRNA expression levels between strains and after SD. During baseline conditions, P2X7RKO mice had attenuated temperature rhythms compared with WT mice, although locomotor activity patterns were similar in both strains. After 6 h of SD, body temperature and locomotion were enhanced to a greater extent in P2X7RKO mice than in WT mice during the initial 2-3 h after SD. Baseline mRNA levels of cortical TNF-α and P2X4R were higher in the KO mice than WT mice. In response to SD, the KO mice failed to increase hypothalamic adenosine deaminase and P2X4R mRNAs. Further, hypothalamic lncRNA expressions varied by strain, and with SD. Current data are consistent with a role for the P2X7R in thermoregulation and lncRNA involvement in purinergic signaling.


Asunto(s)
Temperatura Corporal , Encéfalo/metabolismo , Locomoción , ARN Largo no Codificante/metabolismo , ARN Mensajero/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Privación de Sueño/fisiopatología , Animales , Conducta Animal , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
14.
Sleep ; 39(7): 1467-79, 2016 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-27166236

RESUMEN

STUDY OBJECTIVES: Animal sleep deprivation (SDEP), in contrast to human SDEP, is involuntary and involves repeated exposure to aversive stimuli including the inability of the animal to control the waking stimulus. Therefore, we explored intracranial self-stimulation (ICSS), an operant behavior, as a method for voluntary SDEP in rodents. METHODS: Male Sprague-Dawley rats were implanted with electroencephalography/electromyography (EEG/EMG) recording electrodes and a unilateral bipolar electrode into the lateral hypothalamus. Rats were allowed to self-stimulate, or underwent gentle handling-induced SDEP (GH-SDEP), during the first 6 h of the light phase, after which they were allowed to sleep. Other rats performed the 6 h ICSS and 1 w later were subjected to 6 h of noncontingent stimulation (NCS). During NCS the individual stimulation patterns recorded during ICSS were replayed. RESULTS: After GH-SDEP, ICSS, or NCS, time in nonrapid eye movement (NREM) sleep and rapid eye movement (REM) sleep increased. Further, in the 24 h after SDEP, rats recovered all of the REM sleep lost during SDEP, but only 75% to 80% of the NREM sleep lost, regardless of the SDEP method. The magnitude of EEG slow wave responses occurring during NREM sleep also increased after SDEP treatments. However, NREM sleep EEG slow wave activity (SWA) responses were attenuated following ICSS, compared to GH-SDEP and NCS. CONCLUSIONS: We conclude that ICSS and NCS can be used to sleep deprive rats. Changes in rebound NREM sleep EEG SWA occurring after ICSS, NCS, and GH-SDEP suggest that nonspecific effects of the SDEP procedure differentially affect recovery sleep phenotypes.


Asunto(s)
Autoestimulación , Privación de Sueño/psicología , Animales , Electroencefalografía , Electromiografía , Masculino , Ratas , Ratas Sprague-Dawley , Sueño/fisiología , Privación de Sueño/fisiopatología , Vigilia/fisiología
15.
Sleep Med Rev ; 28: 46-54, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26447948

RESUMEN

Sleep function remains controversial. Individual perspectives frame the issue of sleep function differently. We briefly illustrate how sleep measurement and the evolution, tissue organization levels, molecular mechanisms, and regulation of sleep could influence one's view of sleep function. Then we discuss six viable theories of sleep function. Sleep serves host-defense mechanisms and conserves caloric expenditures, but these functions likely are opportunistic functions evolving later in evolution. That sleep replenishes brain energy stores and that sleep serves a glymphatic function by removing toxic byproducts of waking activity are attractive ideas, but lack extensive supporting experimental evidence. That sleep restores performance is experimentally demonstrated and has obvious evolutionary value. However, this hypothesis lacks experimentally verified mechanisms although ideas relating to this issue are presented. Finally, the ideas surrounding the broad hypothesis that sleep serves a connectivity/plasticity function are many and attractive. There is experimental evidence that connectivity changes with sleep, sleep loss, and with changing afferent input, and that those changes are linked to sleep regulatory mechanisms. In our view, this is the leading contender for the primordial function of sleep. However, much refinement of ideas and innovative experimental approaches are needed to clarify the sleep-connectivity relationship.


Asunto(s)
Sueño/fisiología , Encéfalo/metabolismo , Humanos
16.
Eur J Neurosci ; 42(4): 2078-90, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26036796

RESUMEN

We characterise sleep-like states in cultured neurons and glia during development in vitro as well as after electrical stimulation, the addition of tumor necrosis factor alpha (TNF), and the combination of TNF plus electrical stimulation. We also characterise optogenetic stimulation-induced ATP release and neuronal interleukin-1 and TNF expression in vitro demonstrating the activity dependence of these putative sleep-regulatory substances. Action potential (AP) burstiness, expressed as the burstiness index (BI), synchronization of slow electrical potentials between recording electrodes (SYN), and slow wave (SW) power (0.25-3.75 Hz) determined using fast Fourier analyses emerged as network properties, maturing after 2 weeks in culture. Homologous in vivo measures are used to characterise sleep. Electrical stimulation reduced the BI, SYN and SW power values during and/or after the stimulus period. One day later, homeostasis was evident from rebounds of SYN and SW power values to above baseline levels; the magnitude of the rebound was stimulus pattern-dependent. The addition of TNF enhanced BI, SYN and SW power values, suggesting the induction of a deeper sleep-like state. Electrical stimulation reversed these TNF effects, suggesting the network state was more wake-like. The day after TNF plus electrical stimulation, the changes in SYN and SW power values were dependent upon the stimulus patterns the cells received the day before. We conclude that sleep and wake states in cultured in vitro networks can be controlled and they share molecular regulatory mechanisms with local in vivo networks. Further, sleep is an activity-dependent emergent local network property.


Asunto(s)
Potenciales de Acción/fisiología , Estimulación Eléctrica , Neuroglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Potenciales de Acción/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Animales , Animales Recién Nacidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Fenómenos Biofísicos , Células Cultivadas , Corteza Cerebral/citología , Channelrhodopsins , Técnicas de Cocultivo , Citocinas/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Endogámicos C57BL , Red Nerviosa/efectos de los fármacos , Red Nerviosa/fisiología , Estimulación Luminosa , Transfección
18.
Sleep ; 38(3): 445-51, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25515099

RESUMEN

STUDY OBJECTIVES: The rat psychomotor vigilance task (rPVT) was developed as a rodent analog of the human psychomotor vigilance task (hPVT). We examined whether rPVT performance displays time-on-task effects similar to those observed on the hPVT. DESIGN: The rPVT requires rats to respond to a randomly presented light stimulus to obtain a water reward. Rats were water deprived for 22 h prior to each 30-min rPVT session to motivate performance. We analyzed rPVT performance over time on task and as a function of the response-stimulus interval, at baseline and after sleep deprivation. SETTING: The study was conducted in an academic research vivarium. PARTICIPANTS: Male Long-Evans rats were trained to respond to a 0.5 sec stimulus light within 3 sec of stimulus onset. Complete data were available for n = 20 rats. INTERVENTIONS: Rats performed the rPVT for 30 min at baseline and after 24 h total sleep deprivation by gentle handling. MEASUREMENTS AND RESULTS: Compared to baseline, sleep deprived rats displayed increased performance lapses and premature responses, similar to hPVT lapses of attention and false starts. However, in contrast to hPVT performance, the time-on-task performance decrement was not significantly enhanced by sleep deprivation. Moreover, following sleep deprivation, rPVT response times were not consistently increased after short response-stimulus intervals. CONCLUSIONS: The rPVT manifests similarities to the hPVT in global performance outcomes, but not in post-sleep deprivation effects of time on task and response-stimulus interval.


Asunto(s)
Desempeño Psicomotor , Tiempo de Reacción , Privación de Sueño/fisiopatología , Animales , Atención/fisiología , Agua Potable , Masculino , Motivación , Estimulación Luminosa , Ratas , Ratas Long-Evans , Recompensa , Factores de Tiempo
19.
Brain Behav Immun ; 47: 35-43, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25449578

RESUMEN

Interleukin-1ß (IL1) is involved in sleep regulation and sleep responses induced by influenza virus. The IL1 receptor accessory protein (AcP) and an alternatively spliced isoform of AcP found primarily in neurons, AcPb, form part of the IL1 signaling complex. IL1-induced sleep responses depend on injection time. In rat cortex, both IL1 mRNA and AcPb mRNA peak at Zeitgeber Time (ZT) 0 then decline over the daylight hours. Sleep deprivation enhances cortical IL1 mRNA and AcPb mRNA levels, but not AcP mRNA. We used wild type (WT) and AcPb knockout (KO) mice and performed sleep deprivation between ZT10 and 20 or between ZT22 and 8 based on the time of day expression profiles of AcPb and IL1. We hypothesized that the magnitude of the responses to sleep loss would be strain- and time of day-dependent. In WT mice, NREMS and REMS rebounds occurred regardless of when they were deprived of sleep. In contrast, when AcPbKO mice were sleep deprived from ZT10 to 20 NREMS and REMS rebounds were absent. The AcPbKO mice expressed sleep rebound if sleep loss occurred from ZT22 to 8 although the NREMS responses were not as robust as those that occurred in WT mice. We also challenged mice with intranasal H1N1 influenza virus. WT mice exhibited the expected enhanced sleep responses. In contrast, the AcPbKO mice had less sleep after influenza challenge compared to their own baseline values and compared to WT mice. Body temperature and locomotor activity responses after viral challenge were lower and mortality was higher in AcPbKO than in WT mice. We conclude that neuron-specific AcPb plays a critical role in host defenses and sleep homeostasis.


Asunto(s)
Homeostasis/fisiología , Subtipo H1N1 del Virus de la Influenza A , Proteína Accesoria del Receptor de Interleucina-1/metabolismo , Neuronas/metabolismo , Privación de Sueño/metabolismo , Sueño/fisiología , Animales , Temperatura Corporal/inmunología , Temperatura Corporal/fisiología , Corteza Cerebral/metabolismo , Corteza Cerebral/virología , Homeostasis/inmunología , Proteína Accesoria del Receptor de Interleucina-1/genética , Ratones , Ratones Noqueados , Actividad Motora/inmunología , Actividad Motora/fisiología , Neuronas/virología , Sueño/inmunología , Privación de Sueño/virología
20.
Brain Behav Immun ; 36: 200-6, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24309634

RESUMEN

Ghrelin is an orexigenic hormone produced mainly by the gastrointestinal system and the brain. Much evidence also indicates a role for ghrelin in sleep and thermoregulation. Further, ghrelin was recently implicated in immune system modulation. Administration of bacterial lipopolysaccharide (LPS) induces fever, anorexia, and increased non-rapid-eye movement sleep (NREMS) and these actions are mediated primarily by proinflammatory cytokines. Ghrelin reduces LPS-induced fever, suppresses circulating levels of proinflammatory cytokines and reduces the severity and mortality of various models of experimental endotoxemia. In the present study, we determined the role of intact ghrelin signaling in LPS-induced sleep, feeding, and thermoregulatory responses in mice. Sleep-wake activity was determined after intraperitoneal, dark onset administration of 0.4, 2 and 10 µg LPS in preproghrelin knockout (KO) and wild-type (WT) mice. In addition, body temperature, motor activity and changes in 24-h food intake and body weight were measured. LPS induced dose-dependent increases in NREMS, and suppressed rapid-eye movement sleep, electroencephalographic slow-wave activity, motor activity, food intake and body weight in both Ppg KO and WT mice. Body temperature changes showed a biphasic pattern with a decrease during the dark period followed by an increase in the light phase. The effects of the low and middle doses of LPS were indistinguishable between the two genotypes. Administration of 10 µg LPS, however, induced significantly larger changes in NREMS and wakefulness amounts, body temperature, food intake and body weight in the Ppg KO mice. These findings support a role for ghrelin as an endogenous modulator of inflammatory responses and a central component of arousal and feeding circuits.


Asunto(s)
Ghrelina/fisiología , Conducta de Enfermedad/fisiología , Animales , Regulación de la Temperatura Corporal/efectos de los fármacos , Regulación de la Temperatura Corporal/fisiología , Peso Corporal/efectos de los fármacos , Peso Corporal/fisiología , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Electroencefalografía , Ghrelina/genética , Conducta de Enfermedad/efectos de los fármacos , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Fases del Sueño/efectos de los fármacos , Fases del Sueño/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA