Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
1.
Cell Mol Neurobiol ; 41(1): 191-198, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32239389

RESUMO

Transmissible neurodegenerative prion diseases are characterized by the conversion of the cellular prion protein (PrPC) to misfolded isoforms denoted as prions or PrPSc. Although the conversion can occur in the test tube containing recombinant prion protein or cell lysates, efficient prion formation depends on the integrity of intact cell functions. Since neurons are main targets for prion replication, we asked whether their most specialized function, i.e. synaptic plasticity, could be a factor by which PrPSc formation can be modulated.Immortalized gonadotropin-releasing hormone cells infected with the Rocky Mountain Laboratory prion strain were treated with L-type calcium channels (LTCCs) and NMDA receptors (NMDARs) stimulators or inhibitors. Western blotting was used to monitor the effects on PrPSc formation in relation to ERK signalling.Infected cells showed enhanced levels of phosphorylated ERK (pERK) compared with uninfected cells. Exposure of infected cells to the LTCC agonist Bay K8644 enhanced pERK and PrPSc levels. Although treatment with an LTCC blocker (nimodipine) or an NMDAR competitive antagonist (D-AP5) had no effects, their combination reduced both pERK and PrPSc levels. Treatment with the non-competitive NMDAR channel blocker MK-801 markedly reduced pERK and PrPSc levels.Our study shows that changes in LTCCs and NMDARs activities can modulate PrPSc formation through ERK signalling. During synaptic plasticity, while ERK signalling promotes long-term potentiation accompanied by expansion of post-synaptic lipid rafts, other NMDA receptor-depending signalling pathways, p38-JNK, have opposing effects. Our findings indicate that contrasting intracellular signals of synaptic plasticity can influence time-dependent prion conversion.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Príons/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Linhagem Celular , Maleato de Dizocilpina/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Modelos Biológicos , Nimodipina/farmacologia , Fosforilação/efeitos dos fármacos , Proteínas PrPSc/metabolismo
2.
Proc Natl Acad Sci U S A ; 113(3): E368-77, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26668381

RESUMO

An increased incidence in the sleep-disorder narcolepsy has been associated with the 2009-2010 pandemic of H1N1 influenza virus in China and with mass vaccination campaigns against influenza during the pandemic in Finland and Sweden. Pathogenetic mechanisms of narcolepsy have so far mainly focused on autoimmunity. We here tested an alternative working hypothesis involving a direct role of influenza virus infection in the pathogenesis of narcolepsy in susceptible subjects. We show that infection with H1N1 influenza virus in mice that lack B and T cells (Recombinant activating gene 1-deficient mice) can lead to narcoleptic-like sleep-wake fragmentation and sleep structure alterations. Interestingly, the infection targeted brainstem and hypothalamic neurons, including orexin/hypocretin-producing neurons that regulate sleep-wake stability and are affected in narcolepsy. Because changes occurred in the absence of adaptive autoimmune responses, the findings show that brain infections with H1N1 virus have the potential to cause per se narcoleptic-like sleep disruption.


Assuntos
Vírus da Influenza A Subtipo H1N1/fisiologia , Narcolepsia/fisiopatologia , Narcolepsia/virologia , Neurônios/fisiologia , Sono , Vigília , Animais , Antígenos Virais/imunologia , Eletroencefalografia , Proteínas de Homeodomínio/metabolismo , Hipotálamo/fisiopatologia , Hipotálamo/virologia , Imunidade Inata , Camundongos , Camundongos Endogâmicos C57BL , Modelos Neurológicos , Bulbo Olfatório/fisiopatologia , Bulbo Olfatório/virologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/fisiopatologia , Infecções por Orthomyxoviridae/virologia
3.
PLoS Pathog ; 12(2): e1005442, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26915097

RESUMO

Nitric oxide (NO) generated by inducible NO synthase (iNOS) is critical for defense against intracellular pathogens but may mediate inflammatory tissue damage. To elucidate the role of iNOS in neuroinflammation, infections with encephalitogenic Trypanosoma brucei parasites were compared in inos(-/-) and wild-type mice. Inos(-/-) mice showed enhanced brain invasion by parasites and T cells, and elevated protein permeability of cerebral vessels, but similar parasitemia levels. Trypanosome infection stimulated T cell- and TNF-mediated iNOS expression in perivascular macrophages. NO nitrosylated and inactivated pro-inflammatory molecules such as NF-κΒp65, and reduced TNF expression and signalling. iNOS-derived NO hampered both TNF- and T cell-mediated parasite brain invasion. In inos(-/-) mice, TNF stimulated MMP, including MMP9 activity that increased cerebral vessel permeability. Thus, iNOS-generated NO by perivascular macrophages, strategically located at sites of leukocyte brain penetration, can serve as a negative feed-back regulator that prevents unlimited influx of inflammatory cells by restoring the integrity of the blood-brain barrier.


Assuntos
Barreira Hematoencefálica/metabolismo , Encefalite/metabolismo , Macrófagos Peritoneais/metabolismo , Macrófagos/metabolismo , Óxido Nítrico/metabolismo , Animais , Citocinas/metabolismo , Camundongos Knockout , Óxido Nítrico Sintase Tipo II/metabolismo , Trypanosoma brucei brucei/metabolismo
4.
Nat Rev Neurosci ; 12(6): 345-57, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21587289

RESUMO

The nervous system is protected by barriers that restrict the invasion of pathogens. Nevertheless, mechanisms have evolved by which microbes can pass these barriers, enter and exit neurons and target various regions of the nervous system. In the brain, immune responses to pathogens are generally not robust, so microbes can hide and survive or, conversely, cause severe uncontrolled infections. Depending on their sites of entry and the regions that they target, microbes can cause diverse nervous system dysfunctions and even influence host behaviour to their own advantage. This Review discusses routes by which microbes can reach the nervous system and cause persistent or life-threatening infections.


Assuntos
Barreira Hematoencefálica/microbiologia , Encéfalo/microbiologia , Neurônios/microbiologia , Animais
5.
Neurobiol Dis ; 58: 116-22, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23742760

RESUMO

Cellular mechanisms play a role in conversion of the normal prion protein PrP(C) to the disease-associated protein PrP(Sc). The cells provide not only PrP(C), but also still largely undefined factors required for efficient prion replication. Previously, we have observed that interference with ERK and p38-JNK MAP kinase pathways has opposing effects on the formation of prions indicating that the process is regulated by a balance in intracellualar signaling pathways. In order to obtain a "flow-chart" of such pathways, we here studied the activation of MEK/ERK and mTORC1 downstream targets in relation to PrP(Sc) accumulation in GT1-1 cells infected with the RML or 22L prion strains. We show that inhibition of mTORC1 with rapamycin causes a reduction of PrP(Sc) accumulation at similar low levels as seen when the interaction between the translation initiation factors eIF4E and eIF4G downstream mTORC1 is inhibited using 4EGI-1. No effect is seen following the inhibition of molecules (S6K1 and Mnk1) that links MEK/ERK signaling to mTORC1-mediated control of translation. Instead, stimulation (high [KCl] or [serum]) or inhibition (MEK-inhibitor) of prion formation is associated with increased or decreased phosphorylation of the neuronal transcription factor Elk1, respectively. This study shows that prion formation can be modulated by translational initiating factors, and suggests that MEK/ERK signaling plays a role in the conversion of PrP(C) to PrP(Sc) via an Elk1-mediated transcriptional control. Altogether, our studies indicate that prion protein conversion is under the control of intracellular signals, which hypothetically, under certain conditions may elicit irreversible responses leading to progressive neurodegenerative diseases.


Assuntos
Proteínas de Transporte/metabolismo , Neurônios/metabolismo , Fosfoproteínas/metabolismo , Príons/metabolismo , Proteínas Elk-1 do Domínio ets/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Butadienos/farmacologia , Proteínas de Transporte/genética , Proteínas de Ciclo Celular , Linhagem Celular Transformada , Inibidores Enzimáticos/farmacologia , Fatores de Iniciação em Eucariotos , Histonas/metabolismo , Hipotálamo/citologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Complexos Multiproteicos/metabolismo , Nitrilas/farmacologia , Fosfoproteínas/genética , Cloreto de Potássio/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Transfecção , Proteínas Elk-1 do Domínio ets/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
6.
J Infect Dis ; 205(2): 320-32, 2012 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22116836

RESUMO

BACKGROUND: The penetration of T cells and trypanosomes into the brain parenchyma is a major pathogenetic event in African trypanosomiasis. METHODS: The role of innate immune responses in the penetration of T cells and Trypanosoma brucei brucei into the brain was studied in knockout mice by using double immunofluorescent staining and real-time polymerase chain reaction. RESULTS: We demonstrate that Toll-like receptor (TLR)-MyD88-mediated signaling is required for T-cell and parasite penetration into the brain and microglial activation, besides controlling parasitemia and antigen-specific T-cell activation. Among different TLR-deficient mice studied, TLR9 mediated parasitemia control and T-cell penetration into the brain. TLR-MyD88 signals increased levels of interferon (IFN) ß and tumor necrosis factor (TNF) α transcripts in the brains of infected mice and both TNF-α and IFN-α/ß, receptors promoted T-cell and trypanosoma infiltration into the brain parenchyma. Both resident and infiltrating inflammatory cells in the brain controlled parasite densities in a TLR2- and TLR9-MyD88-mediated manner. However, neither IFN-α/ß nor TNF-α contributed to parasite control in the brain. CONCLUSIONS: Our data indicate that innate immune TLR signals stimulate the expression of TNF-α and IFN-α/ß that initiate brain invasion of T cells and trypanosomes, and control T. brucei brucei load in the brain by molecules distinct from these.


Assuntos
Encéfalo/imunologia , Fator 88 de Diferenciação Mieloide/metabolismo , RNA Mensageiro/metabolismo , Trypanosoma brucei brucei , Tripanossomíase Africana/imunologia , Animais , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/parasitologia , Encéfalo/parasitologia , Encéfalo/patologia , Linfócitos T CD4-Positivos , Linfócitos T CD8-Positivos , Imunidade Inata , Interferon beta/metabolismo , Interferon gama/sangue , Interferon gama/imunologia , Camundongos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/deficiência , Fator 88 de Diferenciação Mieloide/genética , Carga Parasitária , Parasitemia/imunologia , Receptor de Interferon alfa e beta/imunologia , Transdução de Sinais , Receptor 2 Toll-Like/metabolismo , Receptor Toll-Like 9/metabolismo , Tripanossomíase Africana/metabolismo , Tripanossomíase Africana/parasitologia , Fator de Necrose Tumoral alfa/metabolismo
7.
FASEB J ; 23(2): 613-22, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18824519

RESUMO

Brain-derived neurotrophic factor, which activates the extracellular regulated kinase (ERK) pathway, increases formation of prions in scrapie-infected gonadotropin-releasing hormone (GT1-1) cells. This indicates that conversion of the cellular prion protein PrP(C) to its pathogenic isoform, PrP(Sc), can be regulated by physiological stimuli acting on specific signal transduction pathways. In the present study, we examined the involvement of different mitogen-activated protein (MAP) kinase cascades and the cAMP-PKA pathway in formation of proteinase K-resistant PrP(Sc) (rPrP(Sc)). Long-term depolarization of GT1-1 cells infected with the Rocky Mountain Laboratory strain of scrapie increased the formation of rPrP(Sc). This effect was associated to ERK activation and was blocked by the MAPK/ERK kinase (MEK) inhibitor U0126. Treatment with forskolin caused a similar increase in rPrP(Sc) formation that was prevented by the protein kinase A (PKA) inhibitor H89. Both depolarization and forskolin treatment were accompanied by increased phosphorylation of the S6 ribosomal protein, while phosphorylation of histone H3 occurred only after forskolin treatment. Inhibitors of p38- and c-Jun NH(2)-terminal kinase (JNK) promoted the formation of rPrP(Sc), in contrast to the clearance of rPrP(Sc) produced by inhibitors of the ERK pathway. Thus, the ERK and the p38-JNK MAP kinase pathways appear to exert opposing effects on rPrP(Sc) formation, suggesting that balances between these intracellular signaling cascades may regulate replication of prions.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sistema de Sinalização das MAP Quinases , Príons/biossíntese , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Colforsina/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Cloreto de Potássio/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Fatores de Tempo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
8.
Brain Behav Immun ; 24(1): 138-52, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19765643

RESUMO

Aging is often accompanied by increased levels of inflammatory molecules in the organism, but age-related changes in the brain response to inflammatory challenges still require clarification. We here investigated in mice whether cytokine signaling and T-cell neuroinvasion undergo age-related changes. We first analyzed the expression of molecules involved in T-cell infiltration and cytokine signaling regulation in the septum and hippocampus of 2-3 months and 20- to 24-month-old mice at 4h after intracerebroventricular injections of tumor necrosis factor (TNF)-alpha or interferon-gammaversus saline injections. Transcripts of the chemokine CXCL9, intercellular adhesion molecule (ICAM)-1 and suppressor of cytokine signaling molecules (SOCS) 1 and 3 were increased in both age groups after cytokine injection; microglia-derived matrix metalloproteinase (MMP) 12 mRNA was induced in old mice also after control saline injections. Age-related changes in ICAM-1 protein expression and T-cell infiltration were then analyzed in mice of 3-4, 8-9 and 15-16 months at 48h after TNF-alpha injections. ICAM-1 immunoreactivity, and Western blotting in striatum, septum, hippocampus and hypothalamus showed progressive age-related enhancement of TNF-alpha-elicited ICAM-1 upregulation. Double immunofluorescence revealed ICAM-1 expression in microglia and astrocytic processes. CD3(+), CD4(+) and CD8(+) T-cells exhibited progressive age-related increases in brain parenchyma and choroid plexus after cytokine exposure. The findings indicate that the brain responses to inflammatory challenges are not only preserved with advancing age, but also include gradual amplification of ICAM-1 expression and T-cell recruitment. The data highlight molecular and cellular correlates of age-related increase of brain sensitivity to inflammatory stimuli, which could be involved in altered brain vulnerability during aging.


Assuntos
Envelhecimento/fisiologia , Encéfalo/crescimento & desenvolvimento , Encéfalo/imunologia , Citocinas/fisiologia , Inflamação/patologia , Transdução de Sinais/fisiologia , Linfócitos T/fisiologia , Animais , Western Blotting , Citocinas/administração & dosagem , Citocinas/farmacologia , Interpretação Estatística de Dados , Imuno-Histoquímica , Injeções Intraventriculares , Molécula 1 de Adesão Intercelular/biossíntese , Molécula 1 de Adesão Intercelular/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA/biossíntese , RNA/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Transcrição Gênica/fisiologia , Fator de Necrose Tumoral alfa/administração & dosagem , Fator de Necrose Tumoral alfa/farmacologia
9.
J Immunol ; 181(5): 3057-66, 2008 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-18713976

RESUMO

Cannabinoids affect diverse biological processes, including functions of the immune system. With respect to the immune system, anti-inflammatory and immunosuppressive effects of cannabinoids have been reported. Cannabinoids stimulate G protein-coupled cannabinoid receptors CB1 and CB2. These receptors are found primarily on neurons. However, they are also found on dendritic cells (DC), which are recognized for their critical role in initiating and maintaining immune responses. Therefore, DC are potential targets for cannabinoids. We report in this study that cannabinoids reduced the DC surface expression of MHC class II molecules as well as their capacity to stimulate T cells. In the nervous system, CB1 receptor signaling modulates K(+) and Ca(2+) channels. Interestingly, cannabinoid-treated DC also showed altered voltage-gated potassium (K(V)) channel function. We speculate that attenuation of K(V) channel function via CB1 receptor signaling in DC may represent one mechanism by which cannabinoids alter DC function.


Assuntos
Canabinoides/farmacologia , Células Dendríticas/efeitos dos fármacos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Linfócitos T/imunologia , Animais , Canais de Cálcio , Células Dendríticas/química , Células Dendríticas/imunologia , Eletrofisiologia , Antígenos de Histocompatibilidade Classe II , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia
10.
J Infect Dis ; 200(10): 1556-65, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19827943

RESUMO

BACKGROUND: Human African trypanosomiasis, caused by Trypanosoma brucei, involves an early hemolymphatic stage followed by a late encephalitic stage. METHODS: We studied the expression of chemokines with use of microarray and enzyme-linked immunosorbent assay in T. brucei brucei-infected mice and in patients with human African trypanosomiasis and examined their role in controlling brain accumulation of T cells and parasites. RESULTS: The messenger RNAs (mRNAs) encoding CXCR3 ligands CXCL9 and CXCL10 demonstrated the greatest increases among chemokines in brain specimens of infected mice, as determined by microarray. CXCL9 and CXCL10 mRNA accumulation was interferon (IFN)-gamma-dependent. Expression of CXCL10 was predominantly observed in astrocytes. Weight loss was registered in wild-type but not in CXCL10(-/-) and CXCR3(-/-) infected mice. Infected CXCL10(-/-) or CXCR3(-/-) mice demonstrated reduced accumulation of trypanosomes and T cells in the brain parenchyma but similar parasitemia levels, compared with wild-type mice. CXCL10 and IFN-gamma levels were increased in the cerebrospinal fluid of patients with late stage but not early stage human African trypanosomiasis. Levels of CXCL10 in patients with late stage human African trypanosomiasis were associated with somnolence, low body weight, and trypanosomes in the cerebrospinal fluid. CONCLUSION: IFN-gamma-dependent CXCL10 is critical for accumulation of T cells and trypanosomes in the brain during experimental African trypanosomiasis. Data suggest CXCL10 as a candidate marker for late stage human African trypanosomiasis.


Assuntos
Astrócitos/metabolismo , Quimiocina CXCL10/metabolismo , Trypanosoma brucei brucei/imunologia , Trypanosoma brucei gambiense/imunologia , Tripanossomíase Africana/metabolismo , Animais , Quimiocina CXCL10/líquido cefalorraquidiano , Quimiocina CXCL10/genética , Quimiocina CXCL9/imunologia , Humanos , Camundongos , Camundongos Knockout , RNA Mensageiro/metabolismo , Receptores CXCR3/imunologia , Tripanossomíase Africana/líquido cefalorraquidiano , Regulação para Cima
11.
Trends Neurosci ; 30(12): 645-52, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17988749

RESUMO

Novel findings on the effects of inflammatory molecules on neuronal circuits, and on molecular interactions between immunity and sleep, in health and disease, shed light on the pathogenesis of disorders of past (encephalitis lethargica) and present concern (human African trypanosomiasis and narcolepsy), which share alterations in sleep-wakefulness transitions. Although these three disorders differ in etiology, synaptic interactions with immune-response-derived molecules could play a pathogenetic role. Knowledge obtained on neural-immune interplay during senescence also has implications for age-related sleep dysregulation, which is common in the elderly population. Altogether, the data indicate that cell groups implicated in the regulation of sleep and wakefulness, circadian timing, and their interactions could be sensitive to synaptic effects of immune molecules.


Assuntos
Encéfalo/fisiologia , Neuroimunomodulação/fisiologia , Fases do Sono/imunologia , Transtornos do Sono-Vigília/imunologia , Vigília/fisiologia , Envelhecimento/imunologia , Encéfalo/imunologia , Encéfalo/fisiopatologia , Humanos , Narcolepsia/imunologia , Narcolepsia/fisiopatologia , Transtornos do Sono-Vigília/fisiopatologia , Tripanossomíase Africana/imunologia , Tripanossomíase Africana/fisiopatologia
12.
J Gen Virol ; 90(Pt 11): 2821-2828, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19605588

RESUMO

Prions are infectious agents resulting from the conversion of a normal cellular protein, PrP(C), to a misfolded species, PrP(Sc). Iatrogenic transmission of prions is known from surgical procedures involving stainless steel materials. Here, it was shown that stainless steel containing nickel and molybdenum binds PrP(Sc) more efficiently and transmits infection to cells in culture to a higher degree than if these elements are not present. Furthermore, both nickel and molybdenum alone adsorbed PrP(Sc), and nickel powder could be used to extract PrP(Sc) from dilute solutions, thus providing a simple approach to concentration of PrP(Sc). The fact that nickel and molybdenum in steel alloys increased the binding affinity, and bound infectivity, of PrP(Sc) is an important issue to consider in the manufacture of surgical instruments and abattoir tools.


Assuntos
Cirurgia Geral/instrumentação , Molibdênio/metabolismo , Níquel/metabolismo , Doenças Priônicas/transmissão , Príons/metabolismo , Aço Inoxidável , Adsorção , Animais , Humanos , Ligação Proteica
13.
Brain Behav Immun ; 23(6): 733-41, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19362585

RESUMO

Epidemiological studies suggest that early life infections may contribute to the development of psychiatric disorders characterized by cognitive deficits. Here, we studied the effects of a neonatal influenza A/WSN/33 virus infection on locomotor activity, working memory and emotional behavior in adult mice. In addition to wild type mice, immunodeficient (Tap1(-/-)) mice lacking functional CD8(+) T cells, were included in the study to model the potential influence of a genetic deficit relating to virus clearance. Three to four months after the infection, infected Tap1(-/-) mice, but not wild type mice, exhibited deficits in working memory as well as increased rearing activity and anxiety. In the medial prefrontal cortices of these infected Tap1(-/-) mice reduced levels of type III Nrg1 transcripts were observed supporting a role for neuregulin 1 signaling in neuronal circuits involved in working memory. Virus replication, distribution or clearance did not differ between the two genotypes. The lack of CD8(+) T cells, however, appeared to contribute to a more pronounced glia response in Tap1(-/-) than in wild type mice. Thus, the present study suggest that the risk of developing deficits in cognitive and emotional behavior following a CNS infection during brain development is influenced by genetic variation in genes involved in the immune response.


Assuntos
Animais Recém-Nascidos , Vírus da Influenza A , Memória de Curto Prazo/fisiologia , Proteínas do Tecido Nervoso/genética , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/psicologia , Membro 2 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Ansiedade/genética , Ansiedade/psicologia , Encéfalo/patologia , Encéfalo/virologia , Sinais (Psicologia) , Imuno-Histoquímica , Inflamação/genética , Inflamação/patologia , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/fisiologia , Neuregulina-1 , Infecções por Orthomyxoviridae/metabolismo , RNA/biossíntese , RNA/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
J Biol Rhythms ; 23(3): 220-31, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18487414

RESUMO

Biological rhythms, and especially the sleep/wake cycle, are frequently disrupted during senescence. This draws attention to the study of aging-related changes in the hypothalamic suprachiasmatic nucleus (SCN), the master circadian pacemaker. The authors here compared the SCN of young and old mice, analyzing presynaptic terminals, including the gamma-aminobutyric acid (GABA)ergic network, and molecules related to the regulation of GABA, the main neurotransmitter of SCN neurons. Transcripts of the alpha3 subunit of the GABAA receptor and the GABA-synthesizing enzyme glutamic acid decarboxylase isoform 67 (GAD67) were analyzed with real-time RT-PCR and GAD67 protein with Western blotting. These parameters did not show significant changes between the 2 age groups. Presynaptic terminals were identified in confocal microscopy with synaptophysin immunofluorescence, and the GABAergic subset of those terminals was revealed by the colocalization of GAD67 and synaptophysin. Quantitative analysis of labeled synaptic endings performed in 2 SCN subregions, where retinal afferents are known to be, respectively, very dense or very sparse, revealed marked aging-related changes. In both subregions, the evaluated parameters (the number of and the area covered by presynaptic terminals and by their GABAergic subset) were significantly decreased in old versus young mice. No significant differences were found between SCN tissue samples from animals sacrificed at different times of day, in either age group. Altogether, the data point out marked reduction in the synaptic network of the aging biological clock, which also affects GABAergic terminals. Such alterations could underlie aging-related SCN dysfunction, including low-amplitude output during senescence.


Assuntos
Envelhecimento/metabolismo , Terminações Pré-Sinápticas/metabolismo , Núcleo Supraquiasmático/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Sequência de Bases , Primers do DNA , Glutamato Descarboxilase/genética , Glutamato Descarboxilase/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , Receptores de GABA/genética , Receptores de GABA/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
J Biol Rhythms ; 23(2): 150-9, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18375864

RESUMO

The proinflammatory cytokine interferon (IFN-gamma) is an immunomodulatory molecule released by immune cells. It was originally described as an antiviral agent but can also affect functions in the nervous system including circadian activity of the principal mammalian circadian pacemaker, the suprachiasmatic nucleus. IFN-gamma and the synergistically acting cytokine tumor necrosis factor-alpha acutely decrease spontaneous excitatory postsynaptic activity and alter spiking activity in tissue preparations of the SCN. Because IFN-gamma can be released chronically during infections, the authors studied the long-term effects of IFN-gamma on SCN neurons by treating dispersed rat SCN cultures with IFN-gamma over a 4-week period. They analyzed the effect of the treatment on the spontaneous spiking pattern and rhythmic expression of the "clock gene," Period 1. They found that cytokine-treated cells exhibited a lower average spiking frequency and displayed a more irregular firing pattern when compared with controls. Furthermore, long-term treatment with IFN-gamma in cultures obtained from a transgenic Per1-luciferase rat significantly reduced the Per1-luc rhythm amplitude in individual SCN neurons. These results show that IFN-gamma can alter the electrical properties and circadian clock gene expression in SCN neurons. The authors hypothesize that IFN-gamma can modulate circadian output, which may be associated with sleep and rhythm disturbances observed in certain infections and in aging.


Assuntos
Potenciais de Ação/fisiologia , Regulação da Expressão Gênica , Interferon gama/metabolismo , Neurônios/fisiologia , Núcleo Supraquiasmático/citologia , Transativadores , Animais , Animais Geneticamente Modificados , Relógios Biológicos/fisiologia , Proteínas CLOCK , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Ritmo Circadiano/fisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Interferon gama/genética , Neurônios/citologia , Proteínas Circadianas Period , Ratos , Núcleo Supraquiasmático/metabolismo , Temperatura , Transativadores/genética , Transativadores/metabolismo , Ácido gama-Aminobutírico/metabolismo
16.
Brain Res Bull ; 145: 18-29, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-29870779

RESUMO

In this review we describe how Trypanosoma brucei brucei, a rodent pathogenic strain of African trypanosomes, can invade the nervous system, first by localization to the choroid plexus, the circumventricular organs (CVOs) and peripheral ganglia, which have fenestrated vessels, followed by crossing of the blood-brain barrier (BBB) into the white matter, hypothalamus, thalamus and basal ganglia. White blood cells (WBCs) pave the way for the trypanosome neuroinvasion. Experiments with immune deficient mice show that the invasion of WBCs is initiated by the toll-like receptor 9, followed by an augmentation phase that depends on the cytokine IFN-γ and the chemokine CXCL10. Nitric oxide (NO) derived from iNOS then prevents a break-down of the BBB and non-regulated passage of cells. This chain of events is relevant for design of better diagnostic tools to distinguish the different stages of the disease as well as for better understanding of the pathogenesis of the nervous system dysfunctions, which include circadian rhythm changes with sleep pattern disruption, pain syndromes, movement disorders and mental disturbances including dementia.


Assuntos
Tripanossomíase Africana/metabolismo , Tripanossomíase Africana/transmissão , África , Animais , Barreira Hematoencefálica/imunologia , Encéfalo/imunologia , Ritmo Circadiano , Modelos Animais de Doenças , Humanos , Camundongos , Sistema Nervoso , Óxido Nítrico Sintase/metabolismo , Parasitos/patogenicidade , Trypanosoma brucei brucei/metabolismo , Trypanosoma brucei brucei/patogenicidade , Tripanossomíase Africana/diagnóstico
17.
Am J Psychiatry ; 165(1): 59-65, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18056223

RESUMO

OBJECTIVE: Infections during early life have been suggested to play a role in the etiology of schizophrenia. Most studies have focused on fetal life; few have explored risk associated with infection during childhood. The results of these have been inconsistent. The present study aims to investigate whether there is an increased risk of schizophrenia and other nonaffective psychoses associated with viral or bacterial CNS infections during childhood and, if so, which specific agents are involved. METHOD: A national cohort consisting of 1.2 million children born between 1973 and 1985 was followed up by using Swedish national registers to retrieve data on hospital admissions for CNS infections at 0-12 years of age (bacterial: N=2,435, viral: N=6,550) as well as admissions for nonaffective psychotic illnesses from the 14th birthday (N=2,269). RESULTS: There was a slightly increased risk of nonaffective psychotic illness associated with viral CNS infections, as well as schizophrenia. There was no evidence of increased risk in relation to bacterial infections. When divided into specific agents, exposures to mumps virus or cytomegalovirus were associated with subsequent psychoses. CONCLUSIONS: Serious viral CNS infections during childhood appear to be associated with the later development of schizophrenia and nonaffective psychoses. The association with specific viruses suggests that the risk is related to infectious agents with a propensity to invade the brain parenchyma.


Assuntos
Infecções do Sistema Nervoso Central/epidemiologia , Transtornos Psicóticos/epidemiologia , Esquizofrenia/epidemiologia , Adolescente , Adulto , Fatores Etários , Idade de Início , Infecções Bacterianas do Sistema Nervoso Central/complicações , Infecções Bacterianas do Sistema Nervoso Central/epidemiologia , Infecções do Sistema Nervoso Central/complicações , Viroses do Sistema Nervoso Central/complicações , Viroses do Sistema Nervoso Central/epidemiologia , Criança , Estudos de Coortes , Comorbidade , Humanos , Estudos Longitudinais , Razão de Chances , Estudos Prospectivos , Transtornos Psicóticos/etiologia , Sistema de Registros/estatística & dados numéricos , Fatores de Risco , Esquizofrenia/etiologia , Estações do Ano , Fatores Sexuais , Suécia/epidemiologia , População Urbana/estatística & dados numéricos
18.
Acta Trop ; 106(1): 72-4, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18329619

RESUMO

The effect of treatment on relapses of Trypanosoma brucei (T. b.) brucei infections in mice in relation to passage of the parasites across the blood-brain barrier (BBB) as visualized by immunohistochemistry was studied. Three daily intraperitoneal injections of 20mg/kg suramin starting at 15 days post-infection (p.i.), when trypanosomes had begun to traverse the BBB, were curative, but not when starting at 21 days p.i. when parasite brain invasion was more pronounced. Relapses occurred in all mice after one or two daily injections of suramin starting at 15 days p.i., but they were delayed when treatment was supplemented with minocycline, which impedes penetration of T. b. brucei into the brain. This study supports the notion that suramin may be effective even when minor parasite neuroinvasion has appeared in African trypanosomiasis and it shows that minocyline can affect relapses of the disease.


Assuntos
Antiprotozoários/uso terapêutico , Minociclina/uso terapêutico , Suramina/uso terapêutico , Tripanossomíase Africana/tratamento farmacológico , Animais , Encéfalo/parasitologia , Quimioterapia Combinada , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Prevenção Secundária , Suramina/administração & dosagem , Fatores de Tempo , Trypanosoma brucei brucei/efeitos dos fármacos
19.
Front Immunol ; 9: 2877, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30619260

RESUMO

Circumventricular organs (CVOs), neural structures located around the third and fourth ventricles, harbor, similarly to the choroid plexus, vessels devoid of a blood-brain barrier (BBB). This enables them to sense immune-stimulatory molecules in the blood circulation, but may also increase chances of exposure to microbes. In spite of this, attacks to CVOs by microbes are rarely described. It is here highlighted that CVOs and choroid plexus can be infected by pathogens circulating in the bloodstream, providing a route for brain penetration, as shown by infections with the parasites Trypanosoma brucei. Immune responses elicited by pathogens or systemic infections in the choroid plexus and CVOs are briefly outlined. From the choroid plexus trypanosomes can seed into the ventricles and initiate accelerated infiltration of T cells and parasites in periventricular areas. The highly motile trypanosomes may also enter the brain parenchyma from the median eminence, a CVO located at the base of the third ventricle, by crossing the border into the BBB-protected hypothalamic arcuate nuclei. A gate may, thus, be provided for trypanosomes to move into brain areas connected to networks of regulation of circadian rhythms and sleep-wakefulness, to which other CVOs are also connected. Functional imbalances in these networks characterize human African trypanosomiasis, also called sleeping sickness. They are distinct from the sickness response to bacterial infections, but can occur in common neuropsychiatric diseases. Altogether the findings lead to the question: does the neglect in reporting microbe attacks to CVOs reflect lack of awareness in investigations or of gate-opening capability by microbes?


Assuntos
Encéfalo/imunologia , Órgãos Circunventriculares/imunologia , Sistema Nervoso/imunologia , Trypanosoma brucei brucei/imunologia , Tripanossomíase Africana/imunologia , Animais , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/parasitologia , Encéfalo/parasitologia , Plexo Corióideo/imunologia , Plexo Corióideo/parasitologia , Órgãos Circunventriculares/parasitologia , Humanos , Modelos Neurológicos , Sistema Nervoso/parasitologia , Parasitos/imunologia , Parasitos/fisiologia , Trypanosoma brucei brucei/fisiologia , Tripanossomíase Africana/parasitologia
20.
Front Neurol ; 9: 82, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29527187

RESUMO

The global public health concern is heightened over the increasing number of emerging viruses, i.e., newly discovered or previously known that have expanded into new geographical zones. These viruses challenge the health-care systems in sub-Saharan Africa (SSA) countries from which several of them have originated and been transmitted by insects worldwide. Some of these viruses are neuroinvasive, but have been relatively neglected by neuroscientists. They may provide experiments by nature to give a time window for exposure to a new virus within sizeable, previously non-infected human populations, which, for instance, enables studies on potential long-term or late-onset effects on the developing nervous system. Here, we briefly summarize studies on the developing brain by West Nile, Zika, and Chikungunya viruses, which are mosquito-borne and have spread worldwide out of SSA. They can all be neuroinvasive, but their effects vary from malformations caused by prenatal infections to cognitive disturbances following perinatal or later infections. We also highlight Ebola virus, which can leave surviving children with psychiatric disturbances and cause persistent infections in the non-human primate brain. Greater awareness within the neuroscience community is needed to emphasize the menace evoked by these emerging viruses to the developing brain. In particular, frontline neuroscience research should include neuropediatric follow-up studies in the field on long-term or late-onset cognitive and behavior disturbances or neuropsychiatric disorders. Studies on pathogenetic mechanisms for viral-induced perturbations of brain maturation should be extended to the vulnerable periods when neurocircuit formations are at peaks during infancy and early childhood.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa