Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Neuroimmunomodulation ; 20(6): 323-33, 2013.
Article in English | MEDLINE | ID: mdl-23948712

ABSTRACT

BACKGROUND: Within hours of intranasal challenge, mouse-adapted H1N1 A/Puerto Rico/8/34 (PR8) influenza genomic RNA is found in the olfactory bulb (OB) and OB pro-inflammatory cytokines are up-regulated. Severing the olfactory tract delays the acute-phase response (APR) and the APR is attenuated by immunization. OBJECTIVES: To determine if immunization affects OB localization of influenza or the molecular brain mechanisms regulating APR. METHODS: Male mice were immunized with PR8 influenza, then OB viral RNA, APR, and influenza-related cytokine responses were determined after homologous viral challenge. RESULTS: Immunization did not prevent influenza OB viral invasion within 24 h of viral challenge. However, it greatly attenuated OB viral RNA 6 days after viral challenge and the APR including hypothermia and body weight loss responses. Within the OB, 24 h after influenza challenge, prior immunization blocked virus-induced up-regulation of toll-like receptor 7 and interferon (IFN) γ mRNAs. At this time, hypothalamic (HT) growth hormone-releasing hormone receptor and tumor necrosis factor-α mRNAs were greatly enhanced in immunized but not in positive control mice. By 6 days after viral challenge, OB and HT mRNAs returned towards baseline values. In the lung, mRNA up-regulation was greater than that in the brain and maximized 6 days after challenge. Lung IFNγ mRNA decreased at 24 h but increased 6 days after challenge in the positive compared to negative controls. Immunization prevented the up-regulation of most of the flu-related mRNAs measured in lungs. CONCLUSION: Collectively, these data suggest a role for OB and HT involvement in immunization protection against influenza infection.


Subject(s)
Acute-Phase Reaction/immunology , Hypothalamus/immunology , Neuroimmunomodulation/physiology , Olfactory Bulb/immunology , Orthomyxoviridae Infections/immunology , Vaccination , Animals , Cytokines/biosynthesis , Cytokines/immunology , Influenza A Virus, H1N1 Subtype , Influenza Vaccines/immunology , Male , Mice , Mice, Inbred C57BL , RNA, Viral/analysis
2.
Brain Behav Immun ; 26(1): 83-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21821116

ABSTRACT

Mice with a dysfunctional myxovirus resistance-1 (dMx1) gene transport intranasally-instilled PR8 influenza virus to the olfactory bulb (OB) within 4 h post-infection. To determine if the presence of a functional Mx1 (fMx1) gene would influence this brain viral localization and/or disease, we infected mature C57BL/6 dMx1 and fMx1 mice under the same conditions and observed sickness behaviors, viral nucleoprotein (NP) RNA expression and innate immune mediator (IIM) mRNA expression in selected tissues at 15 and 96 h post-infection. Virus invaded the OB and lungs comparably in both sub-strains at 15 and 96 h as determined by nested PCR. In contrast, virus was present in blood and somatosensory cortex of dMx1, but not fMx1 mice at 96 h. At 15 h, sickness behaviors were comparable in both sub-strains. By 96 h dMx1, but not fMx1, were moribund. In both 15 and 96 h lungs, viral NP was significantly elevated in the dMx1 mice compared to the fMx1 mice, as determined by quantitative PCR. OB expression of most IIM mRNAs was similar at both time periods in both sub-strains. In contrast, lung IIM mRNAs were elevated in fMx1 at 15 h, but by 96 h were consistently reduced compared to dMx1 mice. In conclusion, functional Mx1 did not alter OB invasion by virus but attenuated illness compared to dMx1 mice. Inflammation was similar in OBs and lungs of both strains at 15 h but by 96 h it was suppressed in lungs, but not in OBs, of fMx1 mice.


Subject(s)
Brain/physiopathology , Brain/virology , GTP-Binding Proteins/genetics , Influenza A virus , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/physiopathology , Animals , Blood/virology , Body Temperature/physiology , Body Weight/physiology , Illness Behavior , Lung/virology , Male , Mice , Mice, Inbred C57BL , Motor Activity/physiology , Myxovirus Resistance Proteins , Olfactory Bulb/virology , Orthomyxoviridae Infections/virology , RNA, Viral/biosynthesis , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction , Somatosensory Cortex/virology
3.
J Appl Physiol (1985) ; 111(3): 665-72, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21719725

ABSTRACT

MicroRNA (miRNA) levels in brain are altered by sleep deprivation; however, the direct effects of any miRNA on sleep have not heretofore been described. We report herein that intracerebroventricular application of a miRNA-132 mimetic (preMIR-132) decreased duration of non-rapid-eye-movement sleep (NREMS) while simultaneously increasing duration of rapid eye movement sleep (REMS) during the light phase. Further, preMIR-132 decreased electroencephalographic (EEG) slow-wave activity (SWA) during NREMS, an index of sleep intensity. In separate experiments unilateral supracortical application of preMIR-132 ipsilaterally decreased EEG SWA during NREMS but did not alter global sleep duration. In addition, after ventricular or supracortical injections of preMIR-132, the mimetic-induced effects were state specific, occurring only during NREMS. After local supracortical injections of the mimetic, cortical miRNA-132 levels were higher at the time sleep-related EEG effects were manifest. We also report that spontaneous cortical levels of miRNA-132 were lower at the end of the sleep-dominant light period compared with at the end of the dark period in rats. Results suggest that miRNAs play a regulatory role in sleep and provide a new tool for investigating sleep regulation.


Subject(s)
Brain/metabolism , MicroRNAs/metabolism , Sleep , Animals , Circadian Rhythm , Electroencephalography , Injections, Intraventricular , Male , Molecular Mimicry , Oligonucleotides/administration & dosage , Oligonucleotides/metabolism , Photoperiod , Rats , Rats, Sprague-Dawley , Sleep, REM , Time Factors
4.
Brain Behav Immun ; 24(2): 306-15, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19861156

ABSTRACT

Certain sickness behaviors occur consistently in influenza-infected humans and mice. These include body temperature changes, somnolence, and anorexia. Several cytokines serve as mediators of the influenza acute phase response (APR), including these sickness behaviors, and one likely inducer of these cytokines is dsRNA produced during viral replication. TLR3 is known to be one of the host cellular components capable of recognizing dsRNA and activating cytokine synthesis. To determine the role of TLR3-detected viral dsRNA in the causation of viral symptoms, TLR3-deficient mice (TLR3 knockouts, or KOs) were infected with a marginally-lethal dose of mouse-adapted X-31 influenza virus. TLR3 KOs and their wild-type (WT) controls were monitored for baseline body temperature, locomotor activity, and sleep profiles prior to infection. Both mouse strains were then infected and monitored for changes in these sickness behaviors plus body weight changes and mortality for up to 14days post-infection. Consistent with the observations that influenza pathology is reduced in TLR3 KOs, we showed that hypothermia after post-infection day 5 and the total loss of body weight were attenuated in the TLR3 KOs. Sleep changes characteristic of this infection model [particularly increased non-rapid-eye-movement sleep (NREMS)] were also attenuated in TLR3 KOs and returned to baseline values more rapidly. Locomotor activity suppression was similar in both strains. Therefore virus-associated dsRNA detected by TLR3 appears to play a substantial role in mediating several aspects of the influenza syndrome in mice.


Subject(s)
Behavior, Animal/physiology , Influenza A Virus, H1N1 Subtype , Orthomyxoviridae Infections/psychology , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/physiology , Animals , Body Temperature/physiology , Body Weight/physiology , Electroencephalography , Electromyography , Influenza A Virus, H1N1 Subtype/isolation & purification , Male , Mice , Mice, Knockout , Motor Activity/physiology , Orthomyxoviridae Infections/mortality , Sleep/physiology , Sleep Stages/physiology
5.
J Appl Physiol (1985) ; 105(4): 1187-98, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18687977

ABSTRACT

Tumor necrosis factor-alpha (TNF-alpha) is associated with sleep regulation in health and disease. Previous studies assessed sleep in mice genetically deficient in the TNF-alpha 55-kDa receptor. In this study, spontaneous and influenza virus-induced sleep profiles were assessed in mice deficient in both the 55-kDa and 75-kDa TNF-alpha receptors [TNF-2R knockouts (KO)] and wild-type (WT) strain controls. Under baseline conditions the TNF-2R KO mice had less non-rapid eye movement sleep (NREMS) than WTs during the nighttime and more rapid eye movement sleep (REMS) than controls during the daytime. The differences between nighttime maximum and daytime minimum values of electroencephalogram (EEG) delta power during NREMS were greater in the TNF-2R KO mice than in WTs. Viral challenge (mouse-adapted influenza X-31) enhanced NREMS and decreased REMS in both strains roughly to the same extent. EEG delta power responses to viral challenge differed substantially between strains; the WT animals increased, whereas the TNF-2R KO mice decreased their EEG delta wave power during NREMS. There were no differences between strains in body temperatures or locomotor activity in uninfected mice or after viral challenge. Analyses of cortical mRNAs confirmed that the TNF-2R KO mice lacked both TNF-alpha receptors; these mice also had higher levels of orexin mRNA and reduced levels of the purine P2X7 receptor compared with WTs. Results reinforce the hypothesis that TNF-alpha is involved in physiological sleep regulation but plays a limited role in the acute-phase response induced by influenza virus.


Subject(s)
Cerebral Cortex/metabolism , Orthomyxoviridae Infections/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Sleep Stages , Tumor Necrosis Factor-alpha/metabolism , Animals , Body Temperature , Cerebral Cortex/physiopathology , Cerebral Cortex/virology , Disease Models, Animal , Electroencephalography , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Knockout , Motor Activity , Neuropeptides/metabolism , Orexins , Orthomyxoviridae Infections/physiopathology , Orthomyxoviridae Infections/virology , RNA, Messenger/metabolism , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2X7 , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type II/deficiency , Receptors, Tumor Necrosis Factor, Type II/genetics , Time Factors
6.
J Neurovirol ; 13(5): 399-409, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17994424

ABSTRACT

Influenza pneumonitis causes severe systemic symptoms in mice, including hypothermia and excess sleep. The association of extrapulmonary virus, particularly virus in the brain, with the onset of such disease symptoms has not been investigated. Mature C57BL/6 male mice were infected intranasally with mouse-adapted human influenza viruses (PR8 or X-31) under inhalation, systemic, or no anesthesia. Core body temperatures were monitored continuously by radiotelemetry, and tissues (lung, brain, olfactory bulb, spleen, blood) were harvested at the time of onset of hypothermia (13 to 24 h post infection [PI]) or at 4 or 7 h PI. Whole RNA from all tissues was examined by one or more of three reverse transcriptase-polymerase chain reaction (RT-PCR) procedures using H1N1 nucleoprotein (NP) primers for minus polarity RNA (genomic or vRNA) or plus polarity RNA (replication intermediates). Selected cytokines were assayed at 4, 7, and 15 h in the olfactory bulb (OB). Minus and plus RNA strands were readily detected in OBs as early as 4 h PI by nested RT-PCR. Anesthesia was not required for viral invasion of the OB. Cytokine mRNAs were also significantly elevated in the OB at 7 and 15 h PI in infected mice. Controls receiving boiled virus expressed only input vRNA and that only in lung. Immunohistochemistry demonstrated localization of H1N1 and NP antigens in olfactory nerves and the glomerular layer of the OB. Therefore a mouse-adapted human influenza virus strain, not known to be neurotropic, was detected in the mouse OB within 4 h PI where it appeared to induce replication intermediates and cytokines.


Subject(s)
Influenza A Virus, H1N1 Subtype/genetics , Olfactory Bulb/virology , Orthomyxoviridae Infections/virology , Animals , Body Temperature , Cytokines/genetics , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H1N1 Subtype/physiology , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Viral/genetics , RNA, Viral/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction
7.
Neurosci Lett ; 422(1): 68-73, 2007 Jul 05.
Article in English | MEDLINE | ID: mdl-17597302

ABSTRACT

MicroRNAs (miRNAs) are small ( approximately 22 nucleotides) non-coding RNA strands that base pair with mRNA to degrade it or inhibit its translation. Because sleep and sleep loss induce changes in many mRNA species, we hypothesized that sleep loss would also affect miRNA levels in the brain. Rats were sleep-deprived for 8h then decapitated; hippocampus, prefrontal and somatosensory cortices and hypothalamus tissues were harvested and frozen in liquid nitrogen. miRNA was extracted and then characterized using microarrays. Several let-7 miRNA microarray results using hippocampus and prefrontal cortex samples were verified by PCR. From the array data it was determined that about 50 miRNA species were affected by sleep loss. For example, in the hippocampus of sleep-deprived rats, miRNA expression increased compared to cage control samples. In contrast, the majority of miRNA species in the somatosensory and prefrontal cortices decreased, while in the hypothalamus miRNA species were both up- and down-regulated after sleep deprivation. The number of miRNA species affected by sleep loss, their differential expression in separate brain structures and their predicted targets suggest that they have a role in site-specific sleep mechanisms. Current results are, to our knowledge, the first demonstration of the homeostatic process, sleep, altering brain miRNA levels.


Subject(s)
Brain Chemistry/physiology , Gene Expression Regulation/physiology , MicroRNAs/metabolism , Sleep Wake Disorders/metabolism , Animals , Homeostasis/drug effects , Male , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Spectrophotometry, Ultraviolet
8.
Am J Physiol Regul Integr Comp Physiol ; 293(2): R922-30, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17537840

ABSTRACT

Growth hormone-releasing hormone (GHRH), its receptor (GHRHR), and other members of the somatotropic axis are involved in non-rapid eye movement sleep (NREMS) regulation. Previously, studies established the involvement of hypothalamic GHRHergic mechanisms in NREMS regulation, but cerebral cortical GHRH mechanisms in sleep regulation remained uninvestigated. Here, we show that unilateral application of low doses of GHRH to the surface of the rat somatosensory cortex ipsilaterally decreased EEG delta wave power, while higher doses enhanced delta power. These actions of GHRH on EEG delta wave power occurred during NREMS but not during rapid eye movement sleep. Further, the cortical forms of GHRH and GHRHR were identical to those found in the hypothalamus and pituitary, respectively. Cortical GHRHR mRNA and protein levels did not vary across the day-night cycle, whereas cortical GHRH mRNA increased with sleep deprivation. These results suggest that cortical GHRH and GHRHR have a role in the regulation of localized EEG delta power that is state dependent, as well as in their more classic hypothalamic role in NREMS regulation.


Subject(s)
Delta Rhythm , Growth Hormone-Releasing Hormone/physiology , Receptors, Neuropeptide/physiology , Receptors, Pituitary Hormone-Regulating Hormone/physiology , Sleep Stages/physiology , Somatosensory Cortex/physiology , Animals , Growth Hormone-Releasing Hormone/genetics , Growth Hormone-Releasing Hormone/pharmacology , Hypothalamus/physiology , Male , Microinjections , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Neuropeptide/genetics , Receptors, Pituitary Hormone-Regulating Hormone/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sleep Deprivation/physiopathology , Sleep Stages/drug effects , Somatosensory Cortex/drug effects
9.
Brain Behav Immun ; 21(3): 311-22, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17098395

ABSTRACT

The role of type I interferons (IFNs) in mediation of acute viral symptoms (fever, somnolence, anorexia, etc.) is unknown. To determine the role of type I IFN in selected symptom development, body temperature and sleep responses to a marginally lethal dose of X-31 influenza virus were examined in mice with a targeted mutation of the IFN receptor type I (IFN-RI knockouts) and compared to wild-type 129 SvEv control mice. Mice were monitored for 48 h to determine baseline temperature and sleep profiles prior to infection, and then for 9 days following infection. Hypothermic responses to virus were perceptible beginning at 64 h post-infection (PI) and were more marked in KO mice until 108 h, when hypothermia became more exaggerated in wild-type controls. Temperatures of wild-type mice continued to decline through day 9 while temperatures in IFN-RI KO mice stabilized. Time spent in non-rapid eye movement sleep (NREMS) increased in KO mice when hypothermia was marked and then returned to baseline levels, while NREMS continued to increase in wild-type mice through day 9. Other sleep parameters [time spent in rapid eye movement sleep (REMS), relative NREMS EEG slow wave activity, NREMS EEG power density] were all reduced in wild-type mice compared to KOs from days 3 to 8 while REMS low frequency EEG power density increased in wild-type relative to KOs. In conclusion, our results indicate that the presence of functional type I IFN slightly ameliorates disease symptoms early in the X-31 infection while exacerbating disease symptoms later in the infection.


Subject(s)
Interferon Type I/metabolism , Orthomyxoviridae Infections/immunology , Orthomyxoviridae/immunology , Receptors, Interferon/metabolism , Sleep Stages/immunology , Analysis of Variance , Animals , Body Temperature Regulation/immunology , Interferon Type I/immunology , Male , Mice , Mice, Knockout , Receptors, Interferon/genetics , Receptors, Interferon/immunology
10.
Brain Behav Immun ; 21(1): 60-7, 2007 Jan.
Article in English | MEDLINE | ID: mdl-15951155

ABSTRACT

Influenza virus infection up-regulates cytokines such as interleukin-1beta (IL-1beta) and activates the somatotropic axis and the hypothalamic-pituitary axis. Mice with deficits in growth hormone releasing hormone (GHRH) signaling (lit/lit mice) respond to influenza virus challenge with a progressive decrease in sleep and lower survival rates. Current experiments characterize plasma glucocorticoid responses and hypothalamic and lung mRNA expression of sleep-related genes in lit/lit mice and their heterozygous controls after influenza virus challenge. lit/lit mice had higher basal and post-infection plasma corticosterone levels compared to controls. In contrast, the heterozygous mice increased hypothalamic GHRH-receptor, CRH-type 2 receptor, IL-1beta, and tumor necrosis factor-alpha (TNF-alpha) mRNAs after virus treatment while the lit/lit mice failed to up-regulate these substances. In contrast, lung levels of IL-1beta and TNF-alpha mRNAs were greater in the lit/lit mice. These data are consistent with the hypothesis that the sleep response to influenza infection is mediated, in part, by an up-regulation of hypothalamic sleep-related transcripts and they also show that a primary deficit in GHRH signaling is associated with enhanced corticosterone secretion and attenuated hypothalamic cytokine response to infection.


Subject(s)
Corticosterone/blood , Cytokines/metabolism , Hypothalamus/immunology , Influenza A Virus, H1N1 Subtype/immunology , Lung/immunology , Orthomyxoviridae Infections/immunology , Receptors, Neuropeptide/physiology , Receptors, Pituitary Hormone-Regulating Hormone/physiology , Analysis of Variance , Animals , Circadian Rhythm/immunology , Corticosterone/immunology , Cytokines/immunology , Gene Expression Profiling , Growth Hormone-Releasing Hormone/deficiency , Hypothalamus/metabolism , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/blood , Orthomyxoviridae Infections/complications , Orthomyxoviridae Infections/virology , RNA, Messenger/analysis , Sleep/immunology , Sleep Wake Disorders/etiology , Sleep Wake Disorders/physiopathology , Up-Regulation
11.
Brain Behav Immun ; 20(3): 290-9, 2006 May.
Article in English | MEDLINE | ID: mdl-16243480

ABSTRACT

Type I interferons (IFNs) include IFNalpha and IFNbeta, both of which are elevated in acute viral infections and both of which have been shown to induce symptoms such as fever and somnolence when administered in pharmacological doses. To investigate the role of type I IFNs in mediation of acute respiratory viral symptoms we examined sleep and body temperature responses in mice with a targeted mutation of the IFN receptor type I (IFN-RI knockouts). IFN-RI knockouts (KOs) or wild-type 129 SvEv controls were challenged intratracheally (IT) with combined poly[rI.rC] (synthetic double-stranded RNA) and IFNgamma, a model that simulates an acute viral infection with respect to body temperature and locomotor activity responses. Control mice of both strains were treated with IT IFNgamma alone. Hypothermic responses to IT poly[rI.rC]/IFNgamma were more exaggerated in the IFN-RI KO mice than in wild-type. The non-rapid eye movement sleep (NREMS) response to IT poly[rI.rC]/IFNgamma was increased earlier in the IFN-RI KO mice than in wild-type, though the total time spent in NREMS was reduced in the KOs compared to wild-type and the return to baseline NREMS was faster in the KOs. The quality of NREMS also was altered more extensively in the wild-type than in the KO mice. Spontaneous rapid eye movement sleep (REMS) was suppressed in IFN-RI KOs as previously reported, but was not substantially altered in either mouse strain by IT poly[rI.rC]/IFNgamma challenge. Our results implicate type I IFNs as inhibitors of the hypothermic response and enhancers of the NREMS response to IT poly[rI.rC]/IFNgamma, a model of acute viral infection.


Subject(s)
Body Temperature/immunology , Receptors, Interferon/physiology , Respiratory Tract Infections/immunology , Sleep Stages/immunology , Virus Diseases/immunology , Analysis of Variance , Animals , Interferon-gamma/immunology , Male , Mice , Mice, Knockout , Mutation , RNA, Double-Stranded/immunology , Receptors, Interferon/deficiency
12.
J Neurosci ; 25(44): 10282-9, 2005 Nov 02.
Article in English | MEDLINE | ID: mdl-16267236

ABSTRACT

Prolactin (PRL) is implicated in the modulation of spontaneous rapid eye movement sleep (REMS). Previous models of hypoprolactinemic animals were characterized by changes in REMS, although associated deficits made it difficult to ascribe changes in REMS to reduced PRL. In the current studies, male PRL knock-out (KO) mice were used; these mice lack functional PRL but have no known additional deficits. Spontaneous REMS was reduced in the PRL KO mice compared with wild-type or heterozygous littermates. Infusion of PRL for 11-12 d into PRL KO mice restored their REMS to that occurring in wild-type or heterozygous controls. Six hours of sleep deprivation induced a non-REMS and a REMS rebound in both PRL KO mice and heterozygous littermates, although the REMS rebound in the KOs was substantially less. Vasoactive intestinal peptide (VIP) induced REMS responses in heterozygous mice but not in KO mice. Similarly, an ether stressor failed to enhance REMS in the PRL KOs but did in heterozygous littermates. Finally, hypothalamic mRNA levels for PRL, VIP, neural nitric oxide synthase (NOS), inducible NOS, and the interferon type I receptor were similar in KO and heterozygous mice. In contrast, tyrosine hydroxylase mRNA was lower in the PRL KO mice than in heterozygous controls and was restored to control values by infusion of PRL, suggesting a functioning short-loop negative feedback regulation in PRL KO mice. Data support the notion that PRL is involved in REMS regulation.


Subject(s)
Prolactin/deficiency , Sleep, REM/genetics , Animals , Hypothalamus/drug effects , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Prolactin/blood , Prolactin/genetics , Sleep, REM/drug effects , Vasoactive Intestinal Peptide/pharmacology
13.
Brain Res Mol Brain Res ; 137(1-2): 213-22, 2005 Jun 13.
Article in English | MEDLINE | ID: mdl-15950780

ABSTRACT

Interleukin-1 beta (IL-1) and CREB have many CNS actions including sleep regulation and hippocampal-dependent learning. CREB acts in part via CREB-binding protein (CBP). We thus determined whether IL-1 could induce CBP gene expression. Initially, cultured hippocampal cells were treated with IL-1 and differential display reverse transcription was used to identify up- and down-regulated genes. We then sequenced rat CBP. Of the IL-1-upregulated genes, CBP and adenine nucleotide translocator-1 (ANT-1) were investigated in vivo. In these experiments, IL-1 was given to rats intraventricularly and sacrificed 2 h later; both CBP and ANT-1 transcripts were upregulated in the cerebral cortex and hypothalamus. We conclude that rat CBP shares many of the functional domains as human and murine CBP and that IL-1 upregulates genes previously associated with learning and sleep.


Subject(s)
Brain Chemistry/genetics , Brain/metabolism , Gene Expression Regulation/physiology , Interleukin-1/metabolism , Nuclear Proteins/genetics , RNA, Messenger/metabolism , Trans-Activators/genetics , Adenine Nucleotide Translocator 1/genetics , Animals , Brain/drug effects , Brain Chemistry/drug effects , CREB-Binding Protein , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Conserved Sequence/genetics , Gene Expression Regulation/drug effects , Humans , Hypothalamus/drug effects , Hypothalamus/metabolism , Injections, Intraventricular , Interleukin-1/pharmacology , Learning/physiology , Male , Mice , Molecular Sequence Data , Protein Structure, Tertiary/genetics , RNA, Messenger/drug effects , Rats , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Sleep/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
14.
J Appl Physiol (1985) ; 97(1): 17-28, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15220315

ABSTRACT

Influenza viral infection induces increases in non-rapid eye movement sleep and decreases in rapid eye movement sleep in normal mice. An array of cytokines is produced during the infection, and some of them, such as IL-1beta and TNF-alpha, are well-defined somnogenic substances. It is suggested that nitric oxide (NO) may mediate the sleep-promoting effects of these cytokines. In this study, we use mice with targeted disruptions of either the neuronal NO synthase (nNOS) or the inducible NO synthase (iNOS) gene, commonly referred to as nNOS or iNOS knockouts (KOs), to investigate sleep changes after influenza viral challenge. We report that the magnitude of viral-induced non-rapid eye movement sleep responses in both nNOS KOs and iNOS KOs was less than that of their respective controls. In addition, the duration of rapid eye movement sleep in nNOS KO mice did not decrease compared with baseline values. All strains of mice had similar viral titers and cytokine gene expression profiles in the lungs. Virus was not isolated from the brains of any strain. However, gene expression in the brain stem differed between nNOS KOs and their controls: mRNA for the interferon-induced gene 2',5'-oligoadenylate synthase 1a was elevated in nNOS KOs relative to their controls at 15 h, and IL-1beta mRNA was elevated in nNOS KOs relative to their controls at 48 h. Our results suggest that NO synthesized by both nNOS and iNOS plays a role in virus-induced sleep changes and that nNOS may modulate cytokine expression in the brain.


Subject(s)
Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/physiology , Orthomyxoviridae Infections/physiopathology , Orthomyxoviridae , Sleep/genetics , Sleep/physiology , 2',5'-Oligoadenylate Synthetase/biosynthesis , 2',5'-Oligoadenylate Synthetase/genetics , Animals , Brain Stem/enzymology , Brain Stem/metabolism , Electroencephalography , Gene Expression Regulation , Influenza A virus/genetics , Interleukin-1/biosynthesis , Interleukin-1/genetics , Lung/enzymology , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type I , Nitric Oxide Synthase Type II , Orthomyxoviridae Infections/enzymology , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Sleep, REM/genetics , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
15.
Life Sci ; 74(20): 2563-76, 2004 Apr 02.
Article in English | MEDLINE | ID: mdl-15010266

ABSTRACT

Double-stranded (ds)RNA is made as a by-product of viral replication. Synthetic dsRNA induces virtually all of the same systemic symptoms as acute viral infections, such as fever and malaise. In order to develop a model of respiratory viral infections (such as influenza) suitable for use in gene knockout mice (where the deleted gene may affect viral replication), we examined C57BL/6 mouse body temperature and locomotor activity responses to the synthetic dsRNA polyriboinosinic.polyribocytidylic acid (poly[rI.rC]) introduced via the intratracheal (IT) route. We compared the IT poly[rI.rC] responses to the well-characterized intraperitoneal (IP) poly[rI.rC] responses. IT poly[rI.rC] failed to induce an acute phase response (APR) in mice, in contrast to IP poly[rI.rC]. However, addition of interferon (IFN)gamma to the IT poly[rI.rC] inoculum induced sustained hypothermia and suppressed locomotor activity responses with similar kinetics to those responses seen in acute mouse influenza. We further examined cytokine, antiviral, muscarinic M2 receptor and inducible nitric oxide synthase gene expression at 5 hr in the lungs of IT challenged mice. These studies suggested that priming the lung with IFNgamma could enhance proinflammatory (IL1beta, IL6, TNFalpha) cytokine gene expression and suppress interferon gene expression compared to IT poly[rI.rC] alone. No differences were detected for the other genes examined. While further molecular characterization of the model is required, we demonstrate that IT challenge with combined poly[rI.rC] and IFNgamma closely simulates the APR to an acute respiratory virus, and may serve as a suitable model for analyzing the molecular basis of the viral APR in gene knockout mice.


Subject(s)
Acute-Phase Reaction , Interferon-gamma/metabolism , RNA, Double-Stranded/metabolism , Respiratory Tract Infections/immunology , Trachea/virology , Animals , Body Temperature , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Gene Expression Regulation , Lung/physiology , Male , Mice , Mice, Inbred C57BL , Motor Activity/physiology , Poly I-C/administration & dosage , Poly I-C/metabolism , RNA, Double-Stranded/administration & dosage , RNA, Viral , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...