Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Crit Rev Immunol ; 37(2-6): 291-315, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29773024

RESUMO

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.


Assuntos
Febre/imunologia , Interações entre Hospedeiro e Microrganismos/imunologia , Interações Hospedeiro-Parasita/imunologia , Infecções/imunologia , Sonolência , Acetilmuramil-Alanil-Isoglutamina/imunologia , Acetilmuramil-Alanil-Isoglutamina/metabolismo , Animais , Encéfalo/imunologia , Encéfalo/metabolismo , Citocinas/imunologia , Citocinas/metabolismo , Endotoxinas/imunologia , Endotoxinas/metabolismo , Febre/microbiologia , Febre/parasitologia , Febre/virologia , Hormônio Liberador de Hormônio do Crescimento/imunologia , Hormônio Liberador de Hormônio do Crescimento/metabolismo , Humanos , Infecções/microbiologia , Infecções/parasitologia , Infecções/virologia , Óxido Nítrico/imunologia , Óxido Nítrico/metabolismo , Sono/imunologia
2.
Brain Behav Immun ; 26(1): 83-9, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21821116

RESUMO

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.


Assuntos
Encéfalo/fisiopatologia , Encéfalo/virologia , Proteínas de Ligação ao GTP/genética , Vírus da Influenza A , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/fisiopatologia , Animais , Sangue/virologia , Temperatura Corporal/fisiologia , Peso Corporal/fisiologia , Comportamento de Doença , Pulmão/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora/fisiologia , Proteínas de Resistência a Myxovirus , Bulbo Olfatório/virologia , Infecções por Orthomyxoviridae/virologia , RNA Viral/biossíntese , RNA Viral/genética , Reação em Cadeia da Polimerase em Tempo Real , Córtex Somatossensorial/virologia
3.
Brain Behav Immun ; 24(2): 306-15, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19861156

RESUMO

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.


Assuntos
Comportamento Animal/fisiologia , Vírus da Influenza A Subtipo H1N1 , Infecções por Orthomyxoviridae/psicologia , Receptor 3 Toll-Like/genética , Receptor 3 Toll-Like/fisiologia , Animais , Temperatura Corporal/fisiologia , Peso Corporal/fisiologia , Eletroencefalografia , Eletromiografia , Vírus da Influenza A Subtipo H1N1/isolamento & purificação , Masculino , Camundongos , Camundongos Knockout , Atividade Motora/fisiologia , Infecções por Orthomyxoviridae/mortalidade , Sono/fisiologia , Fases do Sono/fisiologia
4.
J Appl Physiol (1985) ; 105(4): 1187-98, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18687977

RESUMO

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.


Assuntos
Córtex Cerebral/metabolismo , Infecções por Orthomyxoviridae/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Fases do Sono , Fator de Necrose Tumoral alfa/metabolismo , Animais , Temperatura Corporal , Córtex Cerebral/fisiopatologia , Córtex Cerebral/virologia , Modelos Animais de Doenças , Eletroencefalografia , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H3N2/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Camundongos Knockout , Atividade Motora , Neuropeptídeos/metabolismo , Orexinas , Infecções por Orthomyxoviridae/fisiopatologia , Infecções por Orthomyxoviridae/virologia , RNA Mensageiro/metabolismo , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2X7 , Receptores Tipo I de Fatores de Necrose Tumoral/deficiência , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo II do Fator de Necrose Tumoral/deficiência , Receptores Tipo II do Fator de Necrose Tumoral/genética , Fatores de Tempo
5.
J Neurotrauma ; 19(5): 503-57, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12042091

RESUMO

Traumatic brain injury (TBI) remains a major public health problem globally. In the United States the incidence of closed head injuries admitted to hospitals is conservatively estimated to be 200 per 100,000 population, and the incidence of penetrating head injury is estimated to be 12 per 100,000, the highest of any developed country in the world. This yields an approximate number of 500,000 new cases each year, a sizeable proportion of which demonstrate significant long-term disabilities. Unfortunately, there is a paucity of proven therapies for this disease. For a variety of reasons, clinical trials for this condition have been difficult to design and perform. Despite promising pre-clinical data, most of the trials that have been performed in recent years have failed to demonstrate any significant improvement in outcomes. The reasons for these failures have not always been apparent and any insights gained were not always shared. It was therefore feared that we were running the risk of repeating our mistakes. Recognizing the importance of TBI, the National Institute of Neurological Disorders and Stroke (NINDS) sponsored a workshop that brought together experts from clinical, research, and pharmaceutical backgrounds. This workshop proved to be very informative and yielded many insights into previous and future TBI trials. This paper is an attempt to summarize the key points made at the workshop. It is hoped that these lessons will enhance the planning and design of future efforts in this important field of research.


Assuntos
Lesões Encefálicas/terapia , Ensaios Clínicos como Assunto/métodos , Humanos
6.
J Appl Physiol (1985) ; 95(2): 460-8, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12598490

RESUMO

Viral infections induce excess non-rapid eye movement sleep (NREMS) in mice. Growth hormone-releasing hormone receptor (GHRH receptor) was previously identified as a candidate gene responsible for NREMS responses to influenza challenge in mice. The dwarf lit/lit mouse with a nonfunctional GHRH receptor was used to assess the role of the GHRH receptor in viral-induced NREMS. After influenza A virus infection the duration and intensity [electroencephalogram (EEG) delta power] of NREMS increased in heterozygous mice with the normal phenotype, whereas NREMS and EEG delta power decreased in homozygous lit/lit mice. Lit/lit mice developed a pathological state with EEG slow waves and enhanced muscle tone. Other influenza-induced responses (decreases in rapid eye movement sleep, changes in the EEG high-frequency bands during the various stages of vigilance, hypothermia, and decreased motor activity) did not differ between the heterozygous and lit/lit mice. GH replacement failed to normalize the NREMS responses in the lit/lit mice after influenza inoculation. Decreases in NREMS paralleled hypothermia in the lit/lit mice. Lung virus levels were similar in the two mouse strains. Lit/lit mice had a higher death rate after influenza challenge than the heterozygotes. In conclusion, GHRH signaling is involved in the NREMS response to influenza infection.


Assuntos
Eletroencefalografia , Vírus da Influenza A , Infecções por Orthomyxoviridae/fisiopatologia , Receptores de Neuropeptídeos/deficiência , Receptores de Hormônios Reguladores de Hormônio Hipofisário/deficiência , Sono , Animais , Temperatura Corporal , Encéfalo/virologia , Nanismo/genética , Nanismo/metabolismo , Hormônio do Crescimento/administração & dosagem , Heterozigoto , Homozigoto , Vírus da Influenza A/isolamento & purificação , Bombas de Infusão , Fator de Crescimento Insulin-Like I/metabolismo , Pulmão/patologia , Pulmão/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Orthomyxoviridae/mortalidade , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/virologia , Mutação Puntual , Receptores de Neuropeptídeos/genética , Receptores de Hormônios Reguladores de Hormônio Hipofisário/genética , Sono/efeitos dos fármacos , Sono REM , Carga Viral
7.
J Appl Physiol (1985) ; 97(1): 17-28, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15220315

RESUMO

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.


Assuntos
Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/fisiologia , Infecções por Orthomyxoviridae/fisiopatologia , Orthomyxoviridae , Sono/genética , Sono/fisiologia , 2',5'-Oligoadenilato Sintetase/biossíntese , 2',5'-Oligoadenilato Sintetase/genética , Animais , Tronco Encefálico/enzimologia , Tronco Encefálico/metabolismo , Eletroencefalografia , Regulação da Expressão Gênica , Vírus da Influenza A/genética , Interleucina-1/biossíntese , Interleucina-1/genética , Pulmão/enzimologia , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo I , Óxido Nítrico Sintase Tipo II , Infecções por Orthomyxoviridae/enzimologia , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sono REM/genética , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética
8.
Brain Res ; 973(2): 214-22, 2003 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-12738065

RESUMO

Nitric oxide (NO) affects almost every physiological process, including the regulation of sleep. There is strong evidence that NO plays an important role in rapid eye movement sleep (REMS) regulation. To further investigate the role of NO in sleep, we characterized spontaneous sleep in mice with targeted disruptions (knockout; KO) in the neuronal nitric oxide synthase (nNOS) or inducible (i)NOS genes. REMS in nNOS KO mice was substantially lower than that of their control mice. In contrast, the iNOS KO mice had significantly more REMS than their controls. Inducible NOS KO mice also had less non-REMS (NREMS) during the dark period. Results suggest that nNOS and iNOS play opposite roles in REMS regulation.


Assuntos
Camundongos Knockout , Óxido Nítrico Sintase/fisiologia , Sono REM/fisiologia , Animais , Linhagem Celular , Eletroencefalografia/métodos , Eletromiografia/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo I , Óxido Nítrico Sintase Tipo II , Periodicidade , Sono/genética , Sono/fisiologia , Sono REM/genética , Células-Tronco , Fatores de Tempo
9.
Life Sci ; 74(20): 2563-76, 2004 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-15010266

RESUMO

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.


Assuntos
Reação de Fase Aguda , Interferon gama/metabolismo , RNA de Cadeia Dupla/metabolismo , Infecções Respiratórias/imunologia , Traqueia/virologia , Animais , Temperatura Corporal , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica , Pulmão/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora/fisiologia , Poli I-C/administração & dosagem , Poli I-C/metabolismo , RNA de Cadeia Dupla/administração & dosagem , RNA Viral , Fatores de Tempo
10.
Med Hypotheses ; 75(2): 204-13, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20236772

RESUMO

Neurodegenerative diseases are a horrendous burden for their victims, their families, and society as a whole. For half a century scientists have pursued the hypothesis that these diseases involve a chronic viral infection in the brain. However, efforts to consistently detect a specific virus in brains of patients with such diseases as Alzheimer's or multiple sclerosis have generally failed. Neuropathologists have become increasingly aware that most patients with neurodegenerative diseases demonstrate marked deterioration of the brain olfactory bulb in addition to brain targets that define the specific disease. In fact, the loss of the sense of smell may precede overt neurological symptoms by many years. This realization that the olfactory bulb is a common target in neurodegenerative diseases suggests the possibility that microbes and/or toxins in inhaled air may play a role in their pathogenesis. With regard to inhaled viruses, neuropathologists have focused on those viruses that infect and kill neurons. However, a recent study shows that a respiratory virus with no neurotropic properties can rapidly invade the mouse olfactory bulb from the nasal cavity. Available data suggest that this strain of influenza is passively transported to the bulb via the olfactory nerves (mechanism unknown), and is taken up by glial cells in the outer layers of the bulb. The infected glial cells appear to be activated by the virus, secrete proinflammatory cytokines, and block further spread of virus within the brain. At the time that influenza symptoms become apparent (15 h post-infection), but not prior to symptom onset (10 h post-infection), proinflammatory cytokine-expressing neurons are increased in olfactory cortical pathways and hypothalamus as well as in the olfactory bulb. The mice go on to die of pneumonitis with severe acute phase and respiratory disease symptoms but no classical neurological symptoms. While much remains to be learned about this intranasal influenza-brain invasion model, it suggests the hypothesis that common viruses encountered in our daily life may initiate neuroinflammation via olfactory neural networks. The numerous viruses that we inhale during a lifetime might cause the death of only a few neurons per infection, but this minor damage would accumulate over time and contribute to age-related brain shrinkage and/or neurodegenerative diseases. Elderly individuals with a strong innate inflammatory system, or ongoing systemic inflammation (or both), might be most susceptible to these outcomes. The evidence for the hypothesis that common respiratory viruses may contribute to neurodegenerative processes is developed in the accompanying article.


Assuntos
Encéfalo/virologia , Degeneração Neural , Neurônios/virologia , Administração Intranasal , Idoso , Animais , Córtex Cerebral , Citocinas , Humanos , Influenza Humana , Camundongos , Modelos Biológicos , Doenças Neurodegenerativas , Bulbo Olfatório/virologia , Nervo Olfatório , Risco , Vírus
11.
J Neuroimmunol ; 211(1-2): 73-83, 2009 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-19410300

RESUMO

Influenza virus invades the olfactory bulb (OB) and enhances cytokine mRNAs therein at the time of illness onset. Here we show that viral antigen immunoreactivity co-localized with glial markers in the OB but could not be detected in other brain areas. Interleukin 1beta- and tumor necrosis factor alpha-immunoreactivity co-localized with neuronal markers in olfactory and central autonomic systems, and the number of cytokine-immunoreactive neurons increased at the time of illness onset [15 h post-inoculation (PI)] but not before (10 h PI). These results suggest that the OB virus influences the brain cytokines and therefore the onset of illness.


Assuntos
Sistema Nervoso Autônomo/imunologia , Citocinas/imunologia , Bulbo Olfatório/imunologia , Infecções por Orthomyxoviridae/imunologia , Animais , Sistema Nervoso Autônomo/metabolismo , Sistema Nervoso Autônomo/virologia , Citocinas/metabolismo , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Vírus da Influenza A , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Neuroglia/imunologia , Neuroglia/metabolismo , Neuroglia/virologia , Neurônios/imunologia , Neurônios/metabolismo , Neurônios/virologia , Bulbo Olfatório/metabolismo , Bulbo Olfatório/virologia , Infecções por Orthomyxoviridae/metabolismo
12.
Brain Behav Immun ; 21(3): 311-22, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17098395

RESUMO

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.


Assuntos
Interferon Tipo I/metabolismo , Infecções por Orthomyxoviridae/imunologia , Orthomyxoviridae/imunologia , Receptores de Interferon/metabolismo , Fases do Sono/imunologia , Análise de Variância , Animais , Regulação da Temperatura Corporal/imunologia , Interferon Tipo I/imunologia , Masculino , Camundongos , Camundongos Knockout , Receptores de Interferon/genética , Receptores de Interferon/imunologia
13.
Brain Behav Immun ; 21(1): 60-7, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15951155

RESUMO

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.


Assuntos
Corticosterona/sangue , Citocinas/metabolismo , Hipotálamo/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Pulmão/imunologia , Infecções por Orthomyxoviridae/imunologia , Receptores de Neuropeptídeos/fisiologia , Receptores de Hormônios Reguladores de Hormônio Hipofisário/fisiologia , Análise de Variância , Animais , Ritmo Circadiano/imunologia , Corticosterona/imunologia , Citocinas/imunologia , Perfilação da Expressão Gênica , Hormônio Liberador de Hormônio do Crescimento/deficiência , Hipotálamo/metabolismo , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Orthomyxoviridae/sangue , Infecções por Orthomyxoviridae/complicações , Infecções por Orthomyxoviridae/virologia , RNA Mensageiro/análise , Sono/imunologia , Transtornos do Sono-Vigília/etiologia , Transtornos do Sono-Vigília/fisiopatologia , Regulação para Cima
14.
J Neurovirol ; 13(5): 399-409, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17994424

RESUMO

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.


Assuntos
Vírus da Influenza A Subtipo H1N1/genética , Bulbo Olfatório/virologia , Infecções por Orthomyxoviridae/virologia , Animais , Temperatura Corporal , Citocinas/genética , Vírus da Influenza A Subtipo H1N1/patogenicidade , Vírus da Influenza A Subtipo H1N1/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , RNA Viral/genética , RNA Viral/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Brain Behav Immun ; 20(3): 290-9, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16243480

RESUMO

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.


Assuntos
Temperatura Corporal/imunologia , Receptores de Interferon/fisiologia , Infecções Respiratórias/imunologia , Fases do Sono/imunologia , Viroses/imunologia , Análise de Variância , Animais , Interferon gama/imunologia , Masculino , Camundongos , Camundongos Knockout , Mutação , RNA de Cadeia Dupla/imunologia , Receptores de Interferon/deficiência
17.
J Allergy Clin Immunol ; 116(6): 1188-98, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16337444

RESUMO

Sleep is a fundamental physiologic process with unknown functions. It is divided into 2 distinct states: non-rapid-eye-movement sleep and rapid-eye-movement sleep. After acute infection with nonneurotropic agents, there are stereotypic changes in non-rapid-eye-movement sleep, particularly increased time spent in slow-wave sleep, and often a reduction of time spent in rapid-eye-movement sleep. It is now recognized that both infection-associated sleep and spontaneous sleep are regulated, in part, by immune mediators called cytokines. This review provides brief tutorials on the elements of the innate immune system that detect infection, how sleep is characterized in the laboratory, issues regarding the interpretation of sleep effects on immune function, the interaction of sleep with circadian rhythms and stress, and some of the microbial products, cytokines, and neuropeptides associated with sleep regulation. We also summarize our current understanding of the role of sleep in host defense and asthma exacerbation.


Assuntos
Imunidade Inata , Sono/imunologia , Reação de Fase Aguda , Asma/etiologia , Asma/fisiopatologia , Citocinas/fisiologia , Humanos , Imunocompetência , Privação do Sono/imunologia
18.
J Trauma ; 54(5 Suppl): S100-5, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12768110

RESUMO

BACKGROUND: This report summarizes recent workshop discussions on animal models for hemorrhage and resuscitation research, and it is supplemented with relevant current literature. The emphasis is on models for casualty care on the battlefield. RESULTS: Current animal models of hemorrhage and resuscitation vary substantially from one laboratory to another, and are not based on clinical experience. Hemorrhage and resuscitation protocols are arbitrarily determined, and there is no consensus on predictive endpoints. The use of anesthetics in most animal models obscures crucial hemodynamic responses. Environmental variables that influence outcomes by modulating the stress state of the animal are not controlled. A standardized predictive preclinical animal model that addresses these issues is needed. CONCLUSIONS: Recently conscious animal models have been developed that can minimize anesthesia artifacts, and prognostic endpoints have been defined in the clinic. The time may be right to define a predictive animal model for hemorrhage and resuscitation research that will allow new trauma therapies to advance.


Assuntos
Modelos Animais de Doenças , Hemorragia/terapia , Pesquisa/normas , Ressuscitação/normas , Guerra , Animais , Humanos
19.
Brain Behav Immun ; 17 Suppl 1: S41-7, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12615185

RESUMO

Sleep remains an important enigma in neurobiology; it has a robust adaptive value yet its function remains elusive. Changes in sleep are hallmarks of the acute phase response to infectious challenge. The molecular regulation of these responses involves a cytokine cascade within brain, including interleukin-1 and tumor necrosis factor, and several other substances such as growth hormone releasing hormone, prolactin, nitric oxide and nuclear factor kappaB. These substances are also involved in the regulation of normal spontaneous sleep. Fatigue and sleep disturbances are common in cancer patients and in those receiving cytokine therapy. Regardless, the role of sleep in cancer is relatively uninvestigated.


Assuntos
Imunidade/fisiologia , Neuroimunomodulação/fisiologia , Sono/imunologia , Animais , Humanos , Infecções/imunologia , Neoplasias/imunologia
20.
Brain Behav Immun ; 18(4): 390-8, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15157956

RESUMO

It is well established that cytokines such as tumor necrosis factor-alpha (TNFalpha) and interleukin-1beta (IL-1beta) are involved in physiological sleep regulation, yet their downstream somnogenic mechanisms remain largely uninvestigated. Nitric oxide (NO) is an effector molecule for some TNFalpha actions. Neuronal nitric oxide synthase (nNOS) and inducible nitric oxide synthase (iNOS) gene knockout (KO) mice sleep differently than their respective controls. In this study, we tested the hypothesis that NO mediates TNFalpha-induced sleep using iNOS and nNOS KO mice and their corresponding wild-type controls. Systemic administration of TNFalpha increased non-rapid eye movement sleep (NREMS) in the two control strains and in the iNOS KO mice during the first 4 h post-injection but failed to increase NREMS in nNOS KO mice. Rapid eye movement sleep (REMS) was suppressed by TNFalpha in nNOS controls but not in the other strains examined. The results suggest that TNFalpha affects sleep, in part, through nNOS.


Assuntos
Óxido Nítrico Sintase/fisiologia , Sono/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Encéfalo/fisiologia , Eletroencefalografia , Regulação da Expressão Gênica , Hipotálamo/metabolismo , Injeções Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase/deficiência , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , Óxido Nítrico Sintase Tipo III , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Fases do Sono/fisiologia , Especificidade da Espécie , Fator de Necrose Tumoral alfa/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa