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.
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
3.
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.

4.
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
5.
Prion ; 10(3): 165-81, 2016 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-27220820

RESUMO

Bovine spongiform encephalopathy (BSE) created a global European crisis in the 1980s and 90s, with very serious health and economic implications. Classical BSE now appears to be under control, to a great extent as a result of a global research effort that identified the sources of prions in meat and bone meal (MBM) and developed new animal-testing tools that guided policy. Priority ( www.prionpriority.eu ) was a European Union (EU) Framework Program 7 (FP7)-funded project through which 21 European research institutions and small and medium enterprises (SMEs) joined efforts between 2009 and 2014, to conduct coordinated basic and applied research on prions and prion diseases. At the end of the project, the Priority consortium drafted a position paper ( www.prionpriority.eu/Priority position paper) with its main conclusions. In the present opinion paper, we summarize these conclusions. With respect to the issue of re-introducing ruminant protein into the feed-chain, our opinion is that sustaining an absolute ban on feeding ruminant protein to ruminants is essential. In particular, the spread and impact of non-classical forms of scrapie and BSE in ruminants is not fully understood and the risks cannot be estimated. Atypical prion agents will probably continue to represent the dominant form of prion diseases in the near future in Europe. Atypical L-type BSE has clear zoonotic potential, as demonstrated in experimental models. Similarly, there are now data indicating that the atypical scrapie agent can cross various species barriers. More epidemiological data from large cohorts are necessary to reach any conclusion on the impact of its transmissibility on public health. Re-evaluations of safety precautions may become necessary depending on the outcome of these studies. Intensified searching for molecular determinants of the species barrier is recommended, since this barrier is key for important policy areas and risk assessment. Understanding the structural basis for strains and the basis for adaptation of a strain to a new host will require continued fundamental research, also needed to understand mechanisms of prion transmission, replication and how they cause nervous system dysfunction and death. Early detection of prion infection, ideally at a preclinical stage, also remains crucial for development of effective treatment strategies.


Assuntos
Cadeia Alimentar , Doenças Priônicas/epidemiologia , Doenças Priônicas/prevenção & controle , Príons/análise , Ração Animal/efeitos adversos , Animais , Bovinos , Diagnóstico Precoce , Encefalopatia Espongiforme Bovina/diagnóstico , Encefalopatia Espongiforme Bovina/epidemiologia , Encefalopatia Espongiforme Bovina/prevenção & controle , Encefalopatia Espongiforme Bovina/transmissão , Europa (Continente)/epidemiologia , Humanos , Doenças Priônicas/diagnóstico , Doenças Priônicas/transmissão , Príons/isolamento & purificação , Príons/metabolismo , Príons/patogenicidade , Scrapie/diagnóstico , Scrapie/epidemiologia , Scrapie/prevenção & controle , Scrapie/transmissão
6.
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
7.
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
8.
Trends Neurosci ; 37(6): 325-33, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24780507

RESUMO

One hundred years ago, Edwin E. Goldmann discovered the blood-brain barrier (BBB) using trypan dyes. These dyes were developed and named by Paul Ehrlich during his search for drugs to kill African trypanosomes (extracellular parasites that cause sleeping sickness) while sparing host cells. For Ehrlich, this was the first strategy based on the 'chemotherapy' concept he had introduced. The discovery of the BBB revealed, however, the difficulties in drug delivery to the brain. Mechanisms by which parasites enter, dwell, and exit the brain currently provide novel views on cell trafficking across the BBB. These mechanisms also highlight the role of pericytes and endocytosis regulation in BBB functioning and in disrupted BBB gating, which may be involved in the pathogenesis of neurodegeneration.


Assuntos
Compostos Azo/farmacologia , Barreira Hematoencefálica/fisiologia , Movimento Celular/fisiologia , Corantes/farmacologia , Tripanossomicidas/farmacologia , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/parasitologia , Barreira Hematoencefálica/fisiopatologia , Encéfalo/efeitos dos fármacos , Encéfalo/parasitologia , Encéfalo/fisiologia , Encéfalo/fisiopatologia , História do Século XIX , História do Século XX , Humanos , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/fisiopatologia , Neurologia/história , Tripanossomíase/tratamento farmacológico , Tripanossomíase/fisiopatologia
9.
J Cell Biol ; 204(3): 423-41, 2014 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-24493590

RESUMO

Mammalian prions refold host glycosylphosphatidylinositol-anchored PrP(C) into ß-sheet-rich PrP(Sc). PrP(Sc) is rapidly truncated into a C-terminal PrP27-30 core that is stable for days in endolysosomes. The nature of cell-associated prions, their attachment to membranes and rafts, and their subcellular locations are poorly understood; live prion visualization has not previously been achieved. A key obstacle has been the inaccessibility of PrP27-30 epitopes. We overcame this hurdle by focusing on nascent full-length PrP(Sc) rather than on its truncated PrP27-30 product. We show that N-terminal PrP(Sc) epitopes are exposed in their physiological context and visualize, for the first time, PrP(Sc) in living cells. PrP(Sc) resides for hours in unexpected cell-surface, slow moving strings and webs, sheltered from endocytosis. Prion strings observed by light and scanning electron microscopy were thin, micrometer-long structures. They were firmly cell associated, resisted phosphatidylinositol-specific phospholipase C, aligned with raft markers, fluoresced with thioflavin, and were rapidly abolished by anti-prion glycans. Prion strings and webs are the first demonstration of membrane-anchored PrP(Sc) amyloids.


Assuntos
Amiloide/metabolismo , Imageamento Tridimensional , Microdomínios da Membrana/metabolismo , Proteínas PrPSc/metabolismo , Actinas/metabolismo , Amiloide/química , Amiloide/ultraestrutura , Animais , Anticorpos/metabolismo , Benzotiazóis , Sobrevivência Celular , Endocitose , Hipocampo/metabolismo , Camundongos , Modelos Biológicos , Fosfoinositídeo Fosfolipase C/metabolismo , Polissacarídeos/metabolismo , Proteínas PrPSc/química , Ligação Proteica , Desnaturação Proteica , Coloração e Rotulagem , Tiazóis/metabolismo
10.
Pathog Dis ; 70(1): 70-4, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23929591

RESUMO

Peroxiredoxins contribute to protection of some bacteria against reactive oxygen intermediates (ROIs) and reactive nitrogen intermediates (RNIs). Listeria monocytogenes, a facultative intracellular bacterial pathogen, interacts with ROIs and RNIs during infection. In this study, we investigated the involvement of the 2-Cys peroxiredoxin (Prx) homologue in L. monocytogenes in the protection against ROIs and RNIs and in virulence through the construction of an in-frame prx deletion mutant. The Δprx mutant had increased sensitivity to hydrogen peroxide and cumene hydroperoxide compared to the wild-type strain. The mutant also exhibited an increased susceptibility to the nitric oxide-generating compound S-nitroso-N-acetylpenicillamine (SNAP) and 3-morpholinosydnonimine hydrochloride (SIN-1), a peroxynitrite donor. Furthermore, a diminished virulence of the Δprx mutant relative to the wild-type was observed in C57BL/6 mice, but not in inducible nitric oxide synthase-deficient mice. The results suggest that Prx protects L. monocytogenes against oxidative and nitrosative stress in vitro and in vivo and that the prx-encoded polypeptide thereby is involved in L. monocytogenes virulence.


Assuntos
Listeria monocytogenes/genética , Listeria monocytogenes/metabolismo , Estresse Oxidativo/genética , Peroxirredoxinas/genética , Peroxirredoxinas/metabolismo , Virulência/genética , Animais , Peróxido de Hidrogênio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico/genética , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , S-Nitroso-N-Acetilpenicilamina/metabolismo
11.
Handb Clin Neurol ; 114: 11-22, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23829898

RESUMO

Invasion of the central nervous system (CNS) is a most devastating complication of a parasitic infection. Several physical and immunological barriers provide obstacles to such an invasion. In this broad overview focus is given to the physical barriers to neuroinvasion of parasites provided at the portal of entry of the parasites, i.e., the skin and epithelial cells of the gastrointestinal tract, and between the blood and the brain parenchyma, i.e., the blood-brain barrier (BBB). A description is given on how human pathogenic parasites can reach the CNS via the bloodstream either as free-living or extracellular parasites, by embolization of eggs, or within red or white blood cells when adapted to intracellular life. Molecular mechanisms are discussed by which parasites can interact with or pass across the BBB. The possible targeting of the circumventricular organs by parasites, as well as the parasites' direct entry to the brain from the nasal cavity through the olfactory nerve pathway, is also highlighted. Finally, examples are given which illustrate different mechanisms by which parasites can cause dysfunction or damage in the CNS related to toxic effects of parasite-derived molecules or to immune responses to the infection.


Assuntos
Infecções Parasitárias do Sistema Nervoso Central/parasitologia , Interações Hospedeiro-Patógeno , Parasitos/patogenicidade , Animais , Transporte Biológico , Barreira Hematoencefálica/parasitologia , Barreira Hematoencefálica/fisiologia , Infecções Parasitárias do Sistema Nervoso Central/imunologia , Infecções Parasitárias do Sistema Nervoso Central/patologia , Endotélio Vascular/parasitologia , Endotélio Vascular/fisiologia , Humanos
12.
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
13.
PLoS Negl Trop Dis ; 7(2): e2088, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23469311

RESUMO

BACKGROUND: Post-therapeutic follow-up is essential to confirm cure and to detect early treatment failures in patients affected by sleeping sickness (HAT). Current methods, based on finding of parasites in blood and cerebrospinal fluid (CSF) and counting of white blood cells (WBC) in CSF, are imperfect. New markers for treatment outcome evaluation are needed. We hypothesized that alternative CSF markers, able to diagnose the meningo-encephalitic stage of the disease, could also be useful for the evaluation of treatment outcome. METHODOLOGY/PRINCIPAL FINDINGS: Cerebrospinal fluid from patients affected by Trypanosoma brucei gambiense HAT and followed for two years after treatment was investigated. The population comprised stage 2 (S2) patients either cured or experiencing treatment failure during the follow-up. IgM, neopterin, B2MG, MMP-9, ICAM-1, VCAM-1, CXCL10 and CXCL13 were first screened on a small number of HAT patients (n = 97). Neopterin and CXCL13 showed the highest accuracy in discriminating between S2 cured and S2 relapsed patients (AUC 99% and 94%, respectively). When verified on a larger cohort (n = 242), neopterin resulted to be the most efficient predictor of outcome. High levels of this molecule before treatment were already associated with an increased risk of treatment failure. At six months after treatment, neopterin discriminated between cured and relapsed S2 patients with 87% specificity and 92% sensitivity, showing a higher accuracy than white blood cell numbers. CONCLUSIONS/SIGNIFICANCE: In the present study, neopterin was highlighted as a useful marker for the evaluation of the post-therapeutic outcome in patients suffering from sleeping sickness. Detectable levels of this marker in the CSF have the potential to shorten the follow-up for HAT patients to six months after the end of the treatment.


Assuntos
Biomarcadores/líquido cefalorraquidiano , Monitoramento de Medicamentos/métodos , Neopterina/líquido cefalorraquidiano , Trypanosoma brucei gambiense/patogenicidade , Tripanossomíase Africana/tratamento farmacológico , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento , Adulto Jovem
14.
Clin Transl Med ; 2(1): 1, 2013 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-23369533

RESUMO

Accurate stage determination is crucial in the choice of treatment for patients suffering from sleeping sickness, also known as human African trypanosomiasis (HAT). Current staging methods, based on the counting of white blood cells (WBC) and the detection of parasites in the cerebrospinal fluid (CSF) have limited accuracy. We hypothesized that immune mediators reliable for staging T. b. gambiense HAT could also be used to stratify T. b. rhodesiense patients, the less common form of HAT.A population comprising 85 T. b. rhodesiense patients, 14 stage 1 (S1) and 71 stage 2 (S2) enrolled in Malawi and Uganda, was investigated. The CSF levels of IgM, MMP-9, CXCL13, CXCL10, ICAM-1, VCAM-1, neopterin and B2MG were measured and their staging performances evaluated using receiver operating characteristic (ROC) analyses.IgM, MMP-9 and CXCL13 were the most accurate markers for stage determination (partial AUC 88%, 86% and 85%, respectively). The combination in panels of three molecules comprising CXCL13-CXCL10-MMP-9 or CXCL13-CXCL10-IgM significantly increased their staging ability to partial AUC 94% (p value < 0.01).The present study highlighted new potential markers for stage determination of T. b. rhodesiense patients. Further investigations are needed to better evaluate these molecules, alone or in panels, as alternatives to WBC to make reliable choice of treatment.

15.
PLoS One ; 7(7): e40909, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22815865

RESUMO

BACKGROUND: Sleeping sickness, or human African trypanosomiasis (HAT), is a protozoan disease that affects rural communities in sub-Saharan Africa. Determination of the disease stage, essential for correct treatment, represents a key issue in the management of patients. In the present study we evaluated the potential of CXCL10, CXCL13, ICAM-1, VCAM-1, MMP-9, B2MG, neopterin and IgM to complement current methods for staging Trypanosoma brucei gambiense patients. METHODS AND FINDINGS: Five hundred and twelve T. b. gambiense HAT patients originated from Angola, Chad and the Democratic Republic of the Congo (D.R.C.). Their classification as stage 2 (S2) was based on the number of white blood cells (WBC) (>5/µL) or presence of parasites in the cerebrospinal fluid (CSF). The CSF concentration of the eight markers was first measured on a training cohort encompassing 100 patients (44 S1 and 56 S2). IgM and neopterin were the best in discriminating between the two stages of disease with 86.4% and 84.1% specificity respectively, at 100% sensitivity. When a validation cohort (412 patients) was tested, neopterin (14.3 nmol/L) correctly classified 88% of S1 and S2 patients, confirming its high staging power. On this second cohort, neopterin also predicted both the presence of parasites, and of neurological signs, with the same ability as IgM and WBC, the current reference for staging. CONCLUSIONS: This study has demonstrated that neopterin is an excellent biomarker for staging T. b. gambiense HAT patients. A rapid diagnostic test for detecting this metabolite in CSF could help in more accurate stage determination.


Assuntos
Meningoencefalite/líquido cefalorraquidiano , Meningoencefalite/parasitologia , Neopterina/líquido cefalorraquidiano , Trypanosoma brucei gambiense/fisiologia , Tripanossomíase Africana/líquido cefalorraquidiano , Tripanossomíase Africana/parasitologia , Adulto , Biomarcadores/líquido cefalorraquidiano , Estudos de Coortes , Feminino , Humanos , Imunoglobulina M/líquido cefalorraquidiano , Contagem de Leucócitos , Masculino , Reprodutibilidade dos Testes , Tripanossomíase Africana/sangue
16.
Virulence ; 3(2): 202-12, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22460639

RESUMO

The blood-brain barrier (BBB) is a structural and functional barrier that protects the central nervous system (CNS) from invasion by blood-borne pathogens including parasites. However, some intracellular and extracellular parasites can traverse the BBB during the course of infection and cause neurological disturbances and/or damage which are at times fatal. The means by which parasites cross the BBB and how the immune system controls the parasites within the brain are still unclear. In this review we present the current understanding of the processes utilized by two human neuropathogenic parasites, Trypanosoma brucei spp and Toxoplasma gondii, to go across the BBB and consequences of CNS invasion. We also describe briefly other parasites that can invade the brain and how they interact with or circumvent the BBB. The roles played by parasite-derived and host-derived molecules during parasitic and white blood cell invasion of the brain are discussed.


Assuntos
Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/parasitologia , Infecções Protozoárias do Sistema Nervoso Central/patologia , Infecções Protozoárias do Sistema Nervoso Central/parasitologia , Interações Hospedeiro-Patógeno , Toxoplasma/patogenicidade , Trypanosoma brucei brucei/patogenicidade , Humanos , Toxoplasma/imunologia , Trypanosoma brucei brucei/imunologia
17.
PLoS Negl Trop Dis ; 6(2): e1525, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22348168

RESUMO

BACKGROUND: Human African trypanosomiasis (HAT) or sleeping sickness leads to a complex neuropsychiatric syndrome with characteristic sleep alterations. Current division into a first, hemolymphatic stage and second, meningoencephalitic stage is primarily based on the detection of white blood cells and/or trypanosomes in the cerebrospinal fluid. The validity of this criterion is, however, debated, and novel laboratory biomarkers are under study. Objective clinical HAT evaluation and monitoring is therefore needed. Polysomnography has effectively documented sleep-wake disturbances during HAT, but could be difficult to apply as routine technology in field work. The non-invasive, cost-effective technique of actigraphy has been widely validated as a tool for the ambulatory evaluation of sleep disturbances. In this pilot study, actigraphy was applied to the clinical assessment of HAT patients. METHODS/PRINCIPAL FINDINGS: Actigraphy was recorded in patients infected by Trypanosoma brucei gambiense, and age- and sex-matched control subjects. Simultaneous nocturnal polysomnography was also performed in the patients. Nine patients, including one child, were analyzed at admission and two of them also during specific treatment. Parameters, analyzed with user-friendly software, included sleep time evaluated from rest-activity signals, rest-activity rhythm waveform and characteristics. The findings showed sleep-wake alterations of various degrees of severity, which in some patients did not parallel white blood cell counts in the cerebrospinal fluid. Actigraphic recording also showed improvement of the analyzed parameters after treatment initiation. Nocturnal polysomnography showed alterations of sleep time closely corresponding to those derived from actigraphy. CONCLUSIONS/SIGNIFICANCE: The data indicate that actigraphy can be an interesting tool for HAT evaluation, providing valuable clinical information through simple technology, well suited also for long-term follow-up. Actigraphy could therefore objectively contribute to the clinical assessment of HAT patients. This method could be incorporated into a clinical scoring system adapted to HAT to be used in the evaluation of novel treatments and laboratory biomarkers.


Assuntos
Actigrafia/métodos , Tripanossomíase Africana/diagnóstico , Tripanossomíase Africana/fisiopatologia , Adulto , Biomarcadores , Pré-Escolar , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Trypanosoma brucei gambiense/isolamento & purificação
18.
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
19.
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
20.
Am J Trop Med Hyg ; 82(6): 983-90, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20519589

RESUMO

Human African trypanosomiasis (HAT), caused by infection with sub-species of Trypanosoma brucei (T. b.), manifests as a hemolymphatic stage followed by an encephalitic stage. The distinction of the two stages needs improvement as drugs used for the late stage are highly toxic. Transcripts encoding 16 secreted proteins differentially expressed in the brains of mice at late stage T. b. brucei infection when the early stage drug suramin is no longer effective and different to immunoglobulins, chemokines, and cytokines, were selected by microarray analysis. Lipocalin 2 and secretory leukocyte peptidase inhibitor (SLPI) mRNA showed the highest differential expression in mice. These transcripts were also upregulated in brains from infected rats. Lipocalin 2 was increased in cerebrospinal fluid (CSF) from rats during late stage T. b. brucei infection. Protein levels of lipocalin 2, SLPI, and the chemokine CXCL10 were found increased in CSF from Trypanosoma brucei gambiense and Trypanosoma brucei rhodesiense late stage HAT compared to early stage.


Assuntos
Tripanossomíase Africana/líquido cefalorraquidiano , Tripanossomíase Africana/diagnóstico , Animais , Biomarcadores/líquido cefalorraquidiano , Biomarcadores/metabolismo , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Regulação da Expressão Gênica , Humanos , Lipocalinas/genética , Lipocalinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Ratos , Ratos Sprague-Dawley , Inibidor Secretado de Peptidases Leucocitárias/genética , Inibidor Secretado de Peptidases Leucocitárias/metabolismo , Organismos Livres de Patógenos Específicos , Tripanossomíase Africana/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...