Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
1.
Trends Parasitol ; 38(6): 450-461, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35227615

RESUMO

The blood-brain barrier (BBB) efficiently protects the central nervous system (CNS) from infectious insults. Yet, the apicomplexan parasite Toxoplasma gondii has a remarkable capability to establish latent cerebral infection in humans and other vertebrates. In addition to the proposed mechanisms for access to the brain parenchyma, recent findings highlight a paramount role played by the BBB in restricting parasite passage and minimizing parasite loads in the brain. Consistent with clinically asymptomatic primary infections in humans, mounting evidence indicates that the original colonization of the brain by T. gondii encompasses previously unappreciated, nondisruptive translocation processes that precede the onset of parasite-limiting immune responses.


Assuntos
Parasitos , Toxoplasma , Animais , Barreira Hematoencefálica/parasitologia , Encéfalo/parasitologia , Humanos , Imunidade
2.
Elife ; 102021 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-34877929

RESUMO

The cellular barriers of the central nervous system proficiently protect the brain parenchyma from infectious insults. Yet, the single-celled parasite Toxoplasma gondii commonly causes latent cerebral infection in humans and other vertebrates. Here, we addressed the role of the cerebral vasculature in the passage of T. gondii to the brain parenchyma. Shortly after inoculation in mice, parasites mainly localized to cortical capillaries, in preference over post-capillary venules, cortical arterioles or meningeal and choroidal vessels. Early invasion to the parenchyma (days 1-5) occurred in absence of a measurable increase in blood-brain barrier (BBB) permeability, perivascular leukocyte cuffs or hemorrhage. However, sparse focalized permeability elevations were detected adjacently to replicative parasite foci. Further, T. gondii triggered inflammatory responses in cortical microvessels and endothelium. Pro- and anti-inflammatory treatments of mice with LPS and hydrocortisone, respectively, impacted BBB permeability and parasite loads in the brain parenchyma. Finally, pharmacological inhibition or Cre/loxP conditional knockout of endothelial focal adhesion kinase (FAK), a BBB intercellular junction regulator, facilitated parasite translocation to the brain parenchyma. The data reveal that the initial passage of T. gondii to the central nervous system occurs principally across cortical capillaries. The integrity of the microvascular BBB restricts parasite transit, which conversely is exacerbated by the inflammatory response.


Assuntos
Barreira Hematoencefálica/parasitologia , Capilares/fisiologia , Toxoplasma/fisiologia , Toxoplasmose Cerebral/parasitologia , Animais , Encéfalo/parasitologia , Feminino , Masculino , Camundongos , Carga Parasitária , Permeabilidade
3.
PLoS Negl Trop Dis ; 15(11): e0009892, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34762691

RESUMO

Human African trypanosomiasis (HAT), also known as sleeping sickness, is a major cause of mortality and morbidity in sub-Saharan Africa. We hypothesised that recent findings of neurological features and parasite brain infiltration occurring at much earlier stages in HAT than previously thought could be explained by early activation of host genetic programmes controlling CNS disease. Accordingly, a transcriptomal analysis was performed on brain tissue at 0, 7, 14, 21 and 28dpi from the HAT CD1/GVR35 mouse model. Up to 21dpi, most parasites are restricted to the blood and lymphatic system. Thereafter the trypanosomes enter the brain initiating the encephalitic stage. Analysis of ten different time point Comparison pairings, revealed a dynamic transcriptome comprising four message populations. All 7dpi Comparisons had by far more differentially expressed genes compared to all others. Prior to invasion of the parenchyma, by 7dpi, ~2,000 genes were up-regulated, denoted [7dpi↑] in contrast to a down regulated population [7dpi↓] also numbering ~2,000. However, by 14dpi both patterns had returned to around the pre-infected levels. The third, [28dpi↑] featured over three hundred transcripts which had increased modestly up to14dpi, thereafter were significantly up-regulated and peaked at 28dpi. The fourth, a minor population, [7dpi↑-28dpi↑], had similar elevated levels at 7dpi and 28dpi. KEGG and GO enrichment analysis predicted a diverse phenotype by 7dpi with changes to innate and adaptive immunity, a Type I interferon response, neurotransmission, synaptic plasticity, pleiotropic signalling, circadian activity and vascular permeability without disruption of the blood brain barrier. This key observation is consistent with recent rodent model neuroinvasion studies and clinical reports of Stage 1 HAT patients exhibiting CNS symptoms. Together, these findings challenge the strict Stage1/Stage2 phenotypic demarcation in HAT and show that that significant neurological, and immune changes can be detected prior to the onset of CNS disease.


Assuntos
Encéfalo/parasitologia , Doenças do Sistema Nervoso Central/genética , Doenças do Sistema Nervoso Central/imunologia , Trypanosoma brucei brucei/fisiologia , Tripanossomíase Africana/genética , Tripanossomíase Africana/imunologia , Animais , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/parasitologia , Encéfalo/imunologia , Doenças do Sistema Nervoso Central/parasitologia , Modelos Animais de Doenças , Progressão da Doença , Feminino , Perfilação da Expressão Gênica , Humanos , Camundongos , Fenótipo , Análise Serial de Proteínas , Transcrição Gênica , Tripanossomíase Africana/parasitologia
4.
PLoS One ; 16(10): e0258199, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34610039

RESUMO

The Apicomplexa protozoan Toxoplasma gondii is a mandatory intracellular parasite and the causative agent of toxoplasmosis. This illness is of medical importance due to its high prevalence worldwide and may cause neurological alterations in immunocompromised persons. In chronically infected immunocompetent individuals, this parasite forms tissue cysts mainly in the brain. In addition, T. gondii infection has been related to mental illnesses such as schizophrenia, bipolar disorder, depression, obsessive-compulsive disorder, as well as mood, personality, and other behavioral changes. In the present study, we evaluated the kinetics of behavioral alterations in a model of chronic infection, assessing anxiety, depression and exploratory behavior, and their relationship with neuroinflammation and parasite cysts in brain tissue areas, blood-brain-barrier (BBB) integrity, and cytokine status in the brain and serum. Adult female C57BL/6 mice were infected by gavage with 5 cysts of the ME-49 type II T. gondii strain, and analyzed as independent groups at 30, 60 and 90 days postinfection (dpi). Anxiety, depressive-like behavior, and hyperactivity were detected in the early (30 dpi) and long-term (60 and 90 dpi) chronic T. gondii infection, in a direct association with the presence of parasite cysts and neuroinflammation, independently of the brain tissue areas, and linked to BBB disruption. These behavioral alterations paralleled the upregulation of expression of tumor necrosis factor (TNF) and CC-chemokines (CCL2/MCP-1, CCL3/MIP-1α, CCL4/MIP-1ß and CCL5/RANTES) in the brain tissue. In addition, increased levels of interferon-gamma (IFNγ), TNF and CCL2/MCP-1 were detected in the peripheral blood, at 30 and 60 dpi. Our data suggest that the persistence of parasite cysts induces sustained neuroinflammation, and BBB disruption, thus allowing leakage of cytokines of circulating plasma into the brain tissue. Therefore, all these factors may contribute to behavioral changes (anxiety, depressive-like behavior, and hyperactivity) in chronic T. gondii infection.


Assuntos
Comportamento Animal , Barreira Hematoencefálica/patologia , Barreira Hematoencefálica/parasitologia , Inflamação/parasitologia , Toxoplasma/fisiologia , Toxoplasmose Cerebral/parasitologia , Animais , Ansiedade/complicações , Ansiedade/fisiopatologia , Edema Encefálico/complicações , Edema Encefálico/fisiopatologia , Doença Crônica , Citocinas/metabolismo , Depressão/complicações , Depressão/fisiopatologia , Feminino , Inflamação/fisiopatologia , Locomoção , Camundongos Endogâmicos C57BL , Força Muscular , Parasitos/fisiologia , Fatores de Tempo , Toxoplasmose Cerebral/fisiopatologia , Regulação para Cima
5.
Cell Mol Life Sci ; 78(12): 5197-5212, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34023934

RESUMO

Multiple cellular processes, such as immune responses and cancer cell metastasis, crucially depend on interconvertible migration modes. However, knowledge is scarce on how infectious agents impact the processes of cell adhesion and migration at restrictive biological barriers. In extracellular matrix, dendritic cells (DCs) infected by the obligate intracellular protozoan Toxoplasma gondii undergo mesenchymal-to-amoeboid transition (MAT) for rapid integrin-independent migration. Here, in a cellular model of the blood-brain barrier, we report that parasitised DCs adhere to polarised endothelium and shift to integrin-dependent motility, accompanied by elevated transendothelial migration (TEM). Upon contact with endothelium, parasitised DCs dramatically reduced velocities and adhered under both static and shear stress conditions, thereby obliterating the infection-induced amoeboid motility displayed in collagen matrix. The motility of adherent parasitised DCs on endothelial monolayers was restored by blockade of ß1 and ß2 integrins or ICAM-1, which conversely reduced motility on collagen-coated surfaces. Moreover, parasitised DCs exhibited enhanced translocation across highly polarised primary murine brain endothelial cell monolayers. Blockade of ß1, ß2 integrins, ICAM-1 and PECAM-1 reduced TEM frequencies. Finally, gene silencing of the pan-integrin-cytoskeleton linker talin (Tln1) or of ß1 integrin (Itgb1) in primary DCs resulted in increased motility on endothelium and decreased TEM. Adding to the paradigms of leukocyte diapedesis, the findings provide novel insights in how an intracellular pathogen impacts the migratory plasticity of leukocytes in response to the cellular environment, to promote infection-related dissemination.


Assuntos
Barreira Hematoencefálica/parasitologia , Movimento Celular , Células Dendríticas/parasitologia , Endotélio Vascular/parasitologia , Integrinas/metabolismo , Toxoplasma/fisiologia , Toxoplasmose/parasitologia , Animais , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/metabolismo , Adesão Celular , Células Dendríticas/metabolismo , Endotélio Vascular/imunologia , Endotélio Vascular/metabolismo , Feminino , Interações Hospedeiro-Parasita , Integrinas/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Toxoplasmose/imunologia , Toxoplasmose/metabolismo , Toxoplasmose/patologia
6.
Biochim Biophys Acta Gen Subj ; 1865(3): 129813, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33321150

RESUMO

BACKGROUND: Malaria is a parasitic disease that compromises the human host. Currently, control of the Plasmodium falciparum burden is centered on artemisinin-based combination therapies. However, decreased sensitivity to artemisinin and derivatives has been reported, therefore it is important to identify new therapeutic strategies. METHOD: We used human erythrocytes infected with P. falciparum and experimental cerebral malaria (ECM) animal model to assess the potential antimalarial effect of eugenol, a component of clove bud essential oil. RESULTS: Plasmodium falciparum cultures treated with increasing concentrations of eugenol reduced parasitemia in a dose-dependent manner, with IC50 of 532.42 ± 29.55 µM. This effect seems to be irreversible and maintained even in the presence of high parasitemia. The prominent effect of eugenol was detected in the evolution from schizont to ring forms, inducing important morphological changes, indicating a disruption in the development of the erythrocytic cycle. Aberrant structural modification was observed by electron microscopy, showing the separation of the two nuclear membrane leaflets as well as other subcellular membranes, such as from the digestive vacuole. Importantly, in vivo studies using ECM revealed a reduction in blood parasitemia and cerebral edema when mice were treated for 6 consecutive days upon infection. CONCLUSIONS: These data suggest a potential effect of eugenol against Plasmodium sp. with an impact on cerebral malaria. GENERAL SIGNIFICANCE: Our results provide a rational basis for the use of eugenol in therapeutic strategies to the treatment of malaria.


Assuntos
Antimaláricos/farmacologia , Edema Encefálico/tratamento farmacológico , Eugenol/farmacologia , Estágios do Ciclo de Vida/efeitos dos fármacos , Malária Cerebral/tratamento farmacológico , Malária Falciparum/tratamento farmacológico , Plasmodium falciparum/efeitos dos fármacos , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/parasitologia , Edema Encefálico/parasitologia , Modelos Animais de Doenças , Eritrócitos/efeitos dos fármacos , Eritrócitos/parasitologia , Humanos , Concentração Inibidora 50 , Estágios do Ciclo de Vida/fisiologia , Malária Cerebral/parasitologia , Malária Falciparum/parasitologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Plasmodium berghei/efeitos dos fármacos , Plasmodium berghei/crescimento & desenvolvimento , Plasmodium berghei/parasitologia , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium falciparum/patogenicidade
7.
PLoS Pathog ; 16(1): e1008261, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31999807

RESUMO

Disruption of blood-brain barrier (BBB) function is a key feature of cerebral malaria. Increased barrier permeability occurs due to disassembly of tight and adherens junctions between endothelial cells, yet the mechanisms governing junction disassembly and vascular permeability during cerebral malaria remain poorly characterized. We found that EphA2 is a principal receptor tyrosine kinase mediating BBB breakdown during Plasmodium infection. Upregulated on brain microvascular endothelial cells in response to inflammatory cytokines, EphA2 is required for the loss of junction proteins on mouse and human brain microvascular endothelial cells. Furthermore, EphA2 is necessary for CD8+ T cell brain infiltration and subsequent BBB breakdown in a mouse model of cerebral malaria. Blocking EphA2 protects against BBB breakdown highlighting EphA2 as a potential therapeutic target for cerebral malaria.


Assuntos
Barreira Hematoencefálica/parasitologia , Malária Cerebral/parasitologia , Receptor EphA2/metabolismo , Adolescente , Animais , Barreira Hematoencefálica/metabolismo , Criança , Pré-Escolar , Estudos Transversais , Feminino , Humanos , Lactente , Malária Cerebral/genética , Malária Cerebral/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Plasmodium falciparum/fisiologia , Receptor EphA2/genética
8.
Immunology ; 159(2): 193-204, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31631339

RESUMO

Excessive inflammatory immune responses during infections with Plasmodium parasites are responsible for severe complications such as cerebral malaria (CM) that can be studied experimentally in mice. Dendritic cells (DCs) activate cytotoxic CD8+ T-cells and initiate immune responses against the parasites. Batf3-/- mice lack a DC subset, which efficiently induces strong CD8 T-cell responses by cross-presentation of exogenous antigens. Here we show that Batf3-/- mice infected with Plasmodium berghei ANKA (PbA) were protected from experimental CM (ECM), characterized by a stable blood-brain barrier (BBB) and significantly less infiltrated peripheral immune cells in the brain. Importantly, the absence of ECM in Batf3-/- mice correlated with attenuated responses of cytotoxic T-cells, as their parasite-specific lytic activity as well as the production of interferon gamma and granzyme B were significantly decreased. Remarkably, spleens of ECM-protected Batf3-/- mice had elevated levels of regulatory immune cells and interleukin 10. Thus, protection from ECM in PbA-infected Batf3-/- mice was associated with the absence of strong CD8+ T-cell activity and induction of immunoregulatory mediators and cells.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/deficiência , Encéfalo/imunologia , Células Dendríticas/imunologia , Malária Cerebral/prevenção & controle , Plasmodium berghei/patogenicidade , Proteínas Repressoras/deficiência , Linfócitos T Citotóxicos/imunologia , Animais , Fatores de Transcrição de Zíper de Leucina Básica/genética , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/parasitologia , Encéfalo/metabolismo , Encéfalo/parasitologia , Células Cultivadas , Células Dendríticas/metabolismo , Células Dendríticas/parasitologia , Modelos Animais de Doenças , Feminino , Granzimas/imunologia , Granzimas/metabolismo , Interações Hospedeiro-Parasita , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-10/imunologia , Interleucina-10/metabolismo , Malária Cerebral/imunologia , Malária Cerebral/metabolismo , Malária Cerebral/parasitologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Plasmodium berghei/imunologia , Proteínas Repressoras/genética , Baço/imunologia , Baço/metabolismo , Baço/parasitologia , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Citotóxicos/parasitologia
9.
J Pediatric Infect Dis Soc ; 9(5): 580-586, 2020 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-31808816

RESUMO

BACKGROUND: Our prior study findings suggest that Plasmodium falciparum is the cause of disease in both malaria retinopathy-positive (RP) and most retinopathy-negative (RN) cerebral malaria (CM), and that absence of retinopathy and decreased disease severity in RN CM may be due to shorter duration of illness, lower parasite biomass, and decreased var gene expression in RN compared to RP CM. In the present study, we assessed the pathophysiology of RP and RN CM. METHODS: We compared markers of systemic and central nervous system inflammation, oxidative stress, neuronal injury, systemic endothelial activation, angiogenesis, and platelet activation in Ugandan children with RP (n = 167) or RN (n = 87) CM. RESULTS: RP children had higher plasma C-reactive protein (P = .013), ferritin and erythropoietin (both P < .001) levels, an elevated cerebrospinal fluid (CSF):plasma albumin ratio (P < .001), and higher CSF tau protein levels (P = .049) than RN children. Levels of plasma and CSF proinflammatory and anti-inflammatory cytokines and oxidative stress markers did not differ between RP and RN children. RN children had higher plasma levels of endothelin 1 (P = .003), platelet-derived growth factor (P = .012), and platelet factor 4 (P = .034). CONCLUSIONS: RP and RN CM may represent different phases of CM. RN CM may be driven by early vasospasm and platelet activation, whereas the more advanced RP CM is associated with greater inflammation, increased erythropoietic drive, blood-brain barrier breakdown, and neuronal injury, each of which may contribute to greater disease severity.


Assuntos
Barreira Hematoencefálica/parasitologia , Inflamação/parasitologia , Malária Cerebral/complicações , Malária Falciparum/complicações , Doenças Retinianas/parasitologia , Barreira Hematoencefálica/patologia , Criança , Pré-Escolar , Células Endoteliais , Feminino , Humanos , Lactente , Malária Cerebral/patologia , Malária Falciparum/patologia , Masculino , Neovascularização Patológica/parasitologia , Neurônios/patologia , Oftalmoscopia/métodos , Estresse Oxidativo , Plasmodium falciparum , Ativação Plaquetária , Doenças Retinianas/diagnóstico , Proteínas tau/líquido cefalorraquidiano
10.
Nano Lett ; 19(12): 8887-8895, 2019 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-31671939

RESUMO

Cerebral malaria is a lethal complication of malaria infection characterized by central nervous system dysfunction and is often not effectively treated by antimalarial combination therapies. It has been shown that the sequestration of the parasite-infected red blood cells that interact with cerebral vessel endothelial cells and the damage of the blood-brain barrier (BBB) play critical roles in the pathogenesis. In this study, we developed a ferritin nanozyme (Fenozyme) composed of recombinant human ferritin (HFn) protein shells that specifically target BBB endothelial cells (BBB ECs) and the inner Fe3O4 nanozyme core that exhibits reactive oxygen species-scavenging catalase-like activity. In the experimental cerebral malaria (ECM) mouse model, administration of the Fenozyme, but not HFn, markedly ameliorated the damage of BBB induced by the parasite and improved the survival rate of infected mice significantly. Further investigations found that Fenozyme, as well as HFn, was able to polarize the macrophages in the liver to the M1 phenotype and promote the elimination of malaria in the blood. Thus, the catalase-like activity of the Fenozyme is required for its therapeutic effect in the mouse model. Moreover, the Fenozyme significantly alleviated the brain inflammation and memory impairment in ECM mice that had been treated with artemether, indicating that combining Fenozyme with an antimalarial drug is a novel strategy for the treatment of cerebral malaria.


Assuntos
Barreira Hematoencefálica/metabolismo , Células Endoteliais/metabolismo , Ferritinas/farmacologia , Malária Cerebral/prevenção & controle , Plasmodium berghei/metabolismo , Animais , Barreira Hematoencefálica/parasitologia , Barreira Hematoencefálica/patologia , Modelos Animais de Doenças , Células Endoteliais/parasitologia , Células Endoteliais/patologia , Ferritinas/genética , Humanos , Inflamação/metabolismo , Inflamação/parasitologia , Inflamação/patologia , Inflamação/prevenção & controle , Fígado/metabolismo , Fígado/parasitologia , Fígado/patologia , Macrófagos/metabolismo , Macrófagos/parasitologia , Macrófagos/patologia , Malária Cerebral/metabolismo , Malária Cerebral/patologia , Camundongos , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia
12.
Trends Parasitol ; 35(7): 516-528, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31147271

RESUMO

Cerebral malaria is a life-threatening complication of malaria caused by the parasite Plasmodium falciparum. The growing problem of drug resistance and the dearth of new antiparasitic drugs are a serious threat to the antimalaria treatment regimes. Studies on humans and the murine model have implicated the disruption of the blood-brain barrier (BBB) in the lethal course of the disease. Therefore, efforts to alleviate the BBB dysfunction could serve as an adjunct therapy. Here, we review the mechanisms associated with the disruption of the BBB. In addition, we discuss the current, still limited, knowledge on the contribution of different cell types, microparticles, and the kynurenine pathway in the regulation of BBB dysfunction, and how these molecules could be used as potential new therapeutic targets.


Assuntos
Barreira Hematoencefálica/parasitologia , Malária Cerebral/fisiopatologia , Malária Cerebral/terapia , Animais , Antimaláricos/uso terapêutico , Barreira Hematoencefálica/citologia , Barreira Hematoencefálica/fisiopatologia , Micropartículas Derivadas de Células/metabolismo , Modelos Animais de Doenças , Desenvolvimento de Medicamentos , Humanos , Cinurenina/metabolismo
13.
Cell Microbiol ; 21(11): e13070, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31219666

RESUMO

Toxoplasma gondii (T. gondii) is a parasitic protist that can infect nearly all nucleated cell types and tissues of warm-blooded vertebrate hosts. T. gondii utilises a unique form of gliding motility to cross cellular barriers, enter tissues, and penetrate host cells, thus enhancing spread within an infected host. However, T. gondii also disseminates by hijacking the migratory abilities of infected leukocytes. Traditionally, this process has been viewed as a route to cross biological barriers such as the blood-brain barrier. Here, we review recent findings that challenge this view by showing that infection of monocytes downregulates the program of transendothelial migration. Instead, infection by T. gondii enhances Rho-dependent interstitial migration of monocytes and macrophages, which enhances dissemination within tissues. Collectively, the available evidence indicates that T. gondii parasites use multiple means to disseminate within the host, including enhanced motility in tissues and translocation across biological barriers.


Assuntos
Infecções do Sistema Nervoso Central/parasitologia , Leucócitos/parasitologia , Macrófagos/parasitologia , Monócitos/parasitologia , Toxoplasma/patogenicidade , Toxoplasmose/parasitologia , Animais , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/parasitologia , Movimento Celular , Infecções do Sistema Nervoso Central/imunologia , Interações Hospedeiro-Patógeno , Humanos , Integrinas/metabolismo , Leucócitos/metabolismo , Toxoplasma/genética , Toxoplasma/metabolismo , Toxoplasmose/imunologia , Toxoplasmose/metabolismo , Toxoplasmose/patologia , Migração Transendotelial e Transepitelial
14.
Cell Microbiol ; 21(9): e13048, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31099453

RESUMO

The apicomplexan parasite Toxoplasma gondii invades tissues and traverses non-permissive biological barriers in infected humans and other vertebrates. Following ingestion, the parasite penetrates the intestinal wall and disseminates to immune-privileged sites such as the brain parenchyma, after crossing the blood-brain barrier. In the present study, we have established a protocol for high-purification of primary mouse brain endothelial cells to generate stably polarised monolayers that allowed assessment of cellular barrier traversal by T. gondii. We report that T. gondii tachyzoites translocate across polarised monolayers of mouse brain endothelial cells and human intestinal Caco2 cells without significantly perturbing barrier impermeability and with minimal change in transcellular electrical resistance. In contrast, challenge with parasite lysate or LPS increased barrier permeability by destabilising intercellular tight junctions (TJs) and accentuated transmigration of T. gondii. Conversely, reduced phosphorylation of the TJ-regulator focal adhesion kinase (FAK) was observed dose-dependently upon challenge of monolayers with live T. gondii but not with parasite lysate or LPS. Pharmacological inhibition of FAK phosphorylation reversibly altered barrier integrity and facilitated T. gondii translocation. Finally, gene silencing of FAK by shRNA facilitated transmigration of T. gondii across epithelial and endothelial monolayers. Jointly, the data demonstrate that T. gondii infection transiently alters the TJ stability through FAK dysregulation to facilitate transmigration. This work identifies the implication of the TJ regulator FAK in the transmigration of T. gondii across polarised cellular monolayers and provides novel insights in how microbes overcome the restrictiveness of biological barriers.


Assuntos
Barreira Hematoencefálica/parasitologia , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Toxoplasma/patogenicidade , Animais , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/fisiopatologia , Encéfalo/parasitologia , Células CACO-2 , Polaridade Celular/fisiologia , Células Endoteliais/parasitologia , Proteína-Tirosina Quinases de Adesão Focal/antagonistas & inibidores , Proteína-Tirosina Quinases de Adesão Focal/genética , Inativação Gênica , Interações Hospedeiro-Patógeno , Humanos , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , RNA Interferente Pequeno , Junções Íntimas/metabolismo , Junções Íntimas/parasitologia , Virulência/efeitos dos fármacos , Virulência/imunologia
15.
PLoS One ; 14(3): e0213244, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30845271

RESUMO

The 14-3-3 proteins are cerebrospinal fluid (CSF) markers of neuronal damage during infectious meningitis and Creutzfeldt-Jakob disease. Little is known about dynamic changes in the individual isoforms in response to parasitic eosinophilic meningitis. The purposes of this study were to determine the 14-3-3 protein isoform patterns, examine the kinetics and correlate the severity of blood brain barrier (BBB) damage with the expressions of these markers in mice with eosinophilic meningitis. Mice were orally infected with 50 A. cantonensis L3 via an oro-gastric tube and sacrificed every week for 3 consecutive weeks after infection. The Evans blue method and BBB junctional protein expressions were used to measure changes in the BBB. Hematoxylin and eosin staining was used to analyze pathological changes in the mice brains following 1-3 weeks of infection with A. cantonensis. The levels of 14-3-3 protein isoforms in serum/CSF and brain homogenates were analyzed by Western blot, and immunohistochemistry (IHC) was used to explore the different isoform distributions of 14-3-3 proteins and changes in BBB junctional proteins in the mice brain meninges. Dexamethasone was injected intraperitoneally from the seventh day post infection (dpi) until the end of the study (21 dpi) to study the changes in BBB junctional proteins. The amounts of Evans blue, tight junction and 14-3-3 protein isoforms in the different groups of mice were compared using the nonparametric Kruskal-Wallis test. There were significant increases in 14-3-3 protein isoforms ß and γ in the CSF in the second and third weeks after infection compared to the controls and first week of infection, which were correlated with the severity of BBB damage in brain histology, and Evans blue extravasation. Using IHC to assess the distribution of 14-3-3 protein isoforms and changes in BBB junctional proteins in the mice brain meninges, the expressions of isoforms ß, γ, ε, and θ and junctional proteins occludin and claudin-5 in the brain meninges increased over a 3-week period after infection compared to the controls and 1 week after infection. The administration of dexamethasone decreased the expressions of BBB junctional proteins occludin and claudin-5 in the mice brain meninges. Our findings support that 14-3-3 proteins ß and γ can potentially be used as a CSF marker of neuronal damage in parasitic eosinophilic meningitis caused by A. cantonensis.


Assuntos
Proteínas 14-3-3/metabolismo , Angiostrongylus cantonensis/patogenicidade , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Eosinofilia/metabolismo , Meningite/metabolismo , Infecções por Strongylida/metabolismo , Animais , Biomarcadores/sangue , Biomarcadores/líquido cefalorraquidiano , Barreira Hematoencefálica/parasitologia , Encéfalo/parasitologia , Modelos Animais de Doenças , Eosinofilia/epidemiologia , Eosinofilia/parasitologia , Feminino , Incidência , Masculino , Meningite/epidemiologia , Meningite/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Isoformas de Proteínas , Infecções por Strongylida/complicações , Infecções por Strongylida/parasitologia
16.
Front Immunol ; 10: 39, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30740102

RESUMO

Trypanosomiasis has been recognized as a scourge in sub-Saharan Africa for centuries. The disease, caused by protozoan parasites of the Trypanosoma genus, is a major cause of mortality and morbidity in animals and man. Human African trypanosomiasis (HAT), or sleeping sickness, results from infections with T. brucei (b.) gambiense or T. b. rhodesiense with T. b. gambiense accounting for over 95% of infections. Historically there have been major epidemics of the infection, followed by periods of relative disease control. As a result of concerted disease surveillance and treatment programmes, implemented over the last two decades, there has been a significant reduction in the number of cases of human disease reported. However, the recent identification of asymptomatic disease carriers gives cause for some concern. The parasites evade the host immune system by switching their surface coat, comprised of variable surface glycoprotein (VSG). In addition, they have evolved a variety of strategies, including the production of serum resistance associated protein (SRA) and T. b. gambiense-specific glycoprotein (TgsGP) to counter host defense molecules. Infection with either disease variant results in an early haemolymphatic-stage followed by a late encephalitic-stage when the parasites migrate into the CNS. The clinical features of HAT are diverse and non-specific with early-stage symptoms common to several infections endemic within sub-Saharan Africa which may result in a delayed or mistaken diagnosis. Migration of the parasites into the CNS marks the onset of late-stage disease. Diverse neurological manifestations can develop accompanied by a neuroinflammatory response, comprised of astrocyte activation, and inflammatory cell infiltration. However, the transition between the early and late-stage is insidious and accurate disease staging, although crucial to optimize chemotherapy, remains problematic with neurological symptoms and neuroinflammatory changes recorded in early-stage infections. Further research is required to develop better diagnostic and staging techniques as well as safer more efficacious drug regimens. Clearer information is also required concerning disease pathogenesis, specifically regarding asymptomatic carriers and the mechanisms employed by the trypanosomes to facilitate progression to the CNS and precipitate late-stage disease. Without progress in these areas it may prove difficult to maintain current control over this historically episodic disease.


Assuntos
Doenças Negligenciadas/diagnóstico , Doenças Negligenciadas/epidemiologia , Trypanosoma brucei gambiense/patogenicidade , Trypanosoma brucei rhodesiense/patogenicidade , Tripanossomíase Africana/diagnóstico , Tripanossomíase Africana/epidemiologia , Animais , Antiprotozoários/administração & dosagem , Antiprotozoários/uso terapêutico , Barreira Hematoencefálica/parasitologia , Encéfalo/parasitologia , Diagnóstico Tardio , Humanos , Incidência , Doenças Negligenciadas/tratamento farmacológico , Doenças Negligenciadas/prevenção & controle , Pentamidina/administração & dosagem , Pentamidina/uso terapêutico , Índice de Gravidade de Doença , Suramina/administração & dosagem , Suramina/uso terapêutico , Resultado do Tratamento , Tripanossomíase Africana/tratamento farmacológico , Tripanossomíase Africana/prevenção & controle
17.
J Neurosci Res ; 97(2): 137-148, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30315659

RESUMO

Neurocysticercosis (NCC) is a helminth infection affecting the central nervous system caused by the larval stage (cysticercus) of Taenia solium. Since vascular alteration and blood-brain barrier (BBB) disruption contribute to NCC pathology, it is postulated that angiogenesis could contribute to the pathology of this disease. This study used a rat model for NCC and evaluated the expression of two angiogenic factors called vascular endothelial growth factor (VEGF-A) and fibroblast growth factor (FGF2). Also, two markers for BBB disruption, the endothelial barrier antigen and immunoglobulin G, were evaluated using immunohistochemical and immunofluorescence techniques. Brain vasculature changes, BBB disruption, and overexpression of angiogenesis markers surrounding viable cysts were observed. Both VEGF-A and FGF2 were overexpressed in the tissue surrounding the cysticerci, and VEGF-A was overexpressed in astrocytes. Vessels showed decreased immunoreactivity to endothelial barrier antigen marker and an extensive staining for IgG was found in the tissues surrounding the cysts. Additionally, an endothelial cell tube formation assay using human umbilical vein endothelial cells showed that excretory and secretory antigens of T. solium cysticerci induce the formation of these tubes. This in vitro model supports the hypothesis that angiogenesis in NCC might be caused by the parasite itself, as opposed to the host inflammatory responses alone. In conclusion, brain vasculature changes, BBB disruption, and overexpression of angiogenesis markers surrounding viable cysts were observed. This study also demonstrates that cysticerci excretory-secretory processes alone can stimulate angiogenesis.


Assuntos
Barreira Hematoencefálica/fisiopatologia , Fatores de Crescimento de Fibroblastos/metabolismo , Neovascularização Patológica/metabolismo , Neurocisticercose/fisiopatologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Vasos Sanguíneos/parasitologia , Vasos Sanguíneos/patologia , Barreira Hematoencefálica/parasitologia , Barreira Hematoencefálica/patologia , Encéfalo/parasitologia , Células Endoteliais/metabolismo , Células Endoteliais/parasitologia , Células Endoteliais/patologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Imunoglobulina G/metabolismo , Neovascularização Patológica/parasitologia , Neurocisticercose/parasitologia , Ratos , Ratos Sprague-Dawley , Taenia solium
18.
Parasitology ; 146(3): 284-298, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30246668

RESUMO

Human fascioliasis is a worldwide, pathogenic food-borne trematodiasis. Impressive clinical pictures comprising puzzling polymorphisms, manifestation multifocality, disease evolution changes, sequelae and mortality, have been reported in patients presenting with neurological, meningeal, neuropsychic and ocular disorders caused at distance by flukes infecting the liver. Proteomic and mass spectrometry analyses of the Fasciola hepatica excretome/secretome identified numerous, several new, plasminogen-binding proteins enhancing plasmin generation. This may underlie blood-brain barrier leakage whether by many simultaneously migrating, small-sized juvenile flukes in the acute phase, or by breakage of encapsulating formations triggered by single worm tracks in the chronic phase. Blood-brain barrier leakages may subsequently occur due to a fibrinolytic system-dependent mechanism involving plasmin-dependent generation of the proinflammatory peptide bradykinin and activation of bradykinin B2 receptors, after different plasminogen-binding protein agglomeration waves. Interactions between diverse parasitic situations and non-imbalancing fibrinolysis system alterations are for the first time proposed that explain the complexity, heterogeneity and timely variations of neurological disorders. Additionally, inflammation and dilation of blood vessels may be due to contact system-dependent generation bradykinin. This baseline allows for search of indicators to detect neurological risk in fascioliasis patients and experimental work on antifibrinolytic treatments or B2 receptor antagonists for preventing blood-brain barrier leakage.


Assuntos
Barreira Hematoencefálica/parasitologia , Proteínas de Transporte/genética , Fasciola hepatica/fisiologia , Fasciolíase/fisiopatologia , Proteínas de Helminto/genética , Doença Aguda , Animais , Transporte Biológico , Proteínas de Transporte/metabolismo , Doença Crônica , Fasciola hepatica/genética , Fasciolíase/parasitologia , Proteínas de Helminto/metabolismo , Humanos , Espectrometria de Massas , Proteômica
19.
FASEB J ; 33(2): 2058-2071, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30226810

RESUMO

Vascular pathology is central to malaria pathogenesis and associated with severity of disease. We have previously documented shedding of the cerebral endothelial glycocalyx in experimental malaria and hypothesized that this action is implicated in the pathogenesis of cerebral malaria (CM). Quantification and characterization of the intraluminal vascular glycocalyx are technically challenging. Here, we used ferritin labeling, computerized image analysis, and biochemical characterization by using in vivo biotinylation and pull down. Image analysis divided mice with CM and uncomplicated malaria and uninfected control mice into 3 non-overlapping groups. Biochemical assessment of the luminal surface revealed malaria-induced alterations in all components of the glycocalyx in CM. This loss was mirrored in increases of the same components in peripheral blood samples. Corticosteroid treatment protected against CM, reduced inflammation, and prevented glycocalyx loss. Adjunctive antithrombin-3 also prevented glycocalyx loss and significantly reduced CM-associated mortality, as well as reduced local inflammation and prevented blood-brain barrier leakage. In contrast, inhibition of matrix metalloproteases with batimastat had limited effects on the glycocalyx and disease progression. Thus, glycocalyx loss may be associated with malaria pathogenesis and could be targeted by adjunctive treatment.-Hempel, C., Sporring, J., Kurtzhals, J. A. L. Experimental cerebral malaria is associated with profound loss of both glycan and protein components of the endothelial glycocalyx.


Assuntos
Endotélio Vascular/metabolismo , Glicocálix/metabolismo , Malária Cerebral/metabolismo , Plasmodium berghei/metabolismo , Plasmodium chabaudi/metabolismo , Polissacarídeos/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/parasitologia , Barreira Hematoencefálica/patologia , Endotélio Vascular/parasitologia , Endotélio Vascular/patologia , Feminino , Glicocálix/patologia , Malária Cerebral/parasitologia , Malária Cerebral/patologia , Camundongos
20.
ACS Chem Neurosci ; 10(1): 658-666, 2019 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-30346711

RESUMO

Central nervous system (CNS) infections caused by free-living amoebae such as Acanthamoeba species and Naegleria fowleri are rare but fatal. A major challenge in the treatment against the infections caused by these amoebae is the discovery of novel compounds that can effectively cross the blood-brain barrier to penetrate the CNS. It is logical to test clinically approved drugs against CNS diseases for their potential antiamoebic effects since they are known for effective blood-brain barrier penetration and affect eukaryotic cell targets. The antiamoebic effects of clinically available drugs for seizures targeting gamma-amino butyric acid (GABA) receptor and ion channels were tested against Acanthamoeba castellanii belonging to the T4 genotype and N. fowleri. Three such drugs, namely, diazepam (Valium), phenobarbitone (Luminal), phenytoin (Dilantin), and their silver nanoparticles (AgNPs) were evaluated against both trophozoites and cysts stage. Drugs alone and drug conjugated silver nanoparticles were tested for amoebicidal, cysticidal, and host-cell cytotoxicity assays. Nanoparticles were synthesized by sodium borohydride reduction of silver nitrate with drugs as capping agents. Drug conjugated nanoconjugates were characterized by ultraviolet-visible (UV-vis) and Fourier transform infrared (FT-IR) spectroscopies and atomic force microscopy (AFM). In vitro moebicidal assay showed potent amoebicidal effects for diazepam, phenobarbitone, and phenytoin-conjugated AgNPs as compared to drugs alone against A. castellanii and N. fowleri. Furthermore, both drugs and drug conjugated AgNPs showed compelling cysticidal effects. Drugs conjugations with silver nanoparticles enhanced their antiacanthamoebic activity. Interestingly, amoeba-mediated host-cell cytotoxicity was also significantly reduced by drugs alone as well as their nanoconjugates. Since, these drugs are being used to target CNS diseases, their evaluation against brain-eating amoebae seems feasible due to advantages such as permeability of the blood-brain barrier, established pharmacokinetics and dynamics, and United States Food and Drug Administration (FDA) approval. Given the limited availability of effective drugs against brain-eating amoebae, the clinically available drugs tested here present potential for further in vivo studies.


Assuntos
Acanthamoeba castellanii/parasitologia , Amebicidas/farmacologia , Doenças do Sistema Nervoso Central/tratamento farmacológico , Naegleria fowleri/efeitos dos fármacos , Acanthamoeba castellanii/efeitos dos fármacos , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/parasitologia , Doenças do Sistema Nervoso Central/parasitologia , Humanos , Nanopartículas Metálicas/parasitologia , Nanoconjugados , Espectroscopia de Infravermelho com Transformada de Fourier/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...