Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Cell Death Differ ; 31(3): 280-291, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38383887

RESUMO

Detection of cytosolic nucleic acids by pattern recognition receptors, including STING and RIG-I, leads to the activation of multiple signalling pathways that culminate in the production of type I interferons (IFNs) which are vital for host survival during virus infection. In addition to protective immune modulatory functions, type I IFNs are also associated with autoimmune diseases. Hence, it is important to elucidate the mechanisms that govern their expression. In this study, we identified a critical regulatory function of the DUSP4 phosphatase in innate immune signalling. We found that DUSP4 regulates the activation of TBK1 and ERK1/2 in a signalling complex containing DUSP4, TBK1, ERK1/2 and IRF3 to regulate the production of type I IFNs. Mice deficient in DUSP4 were more resistant to infections by both RNA and DNA viruses but more susceptible to malaria parasites. Therefore, our study establishes DUSP4 as a regulator of nucleic acid sensor signalling and sheds light on an important facet of the type I IFN regulatory system.


Assuntos
Interferon Tipo I , Proteínas de Membrana , Proteínas Tirosina Fosfatases , Receptores de Superfície Celular , Proteínas Roundabout , Viroses , Animais , Camundongos , Imunidade Inata , Interferon Tipo I/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Viroses/imunologia , Viroses/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Roundabout/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Receptores de Superfície Celular/metabolismo
2.
Mem Inst Oswaldo Cruz ; 117: e210287, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35730803

RESUMO

BACKGROUND: It has been demonstrated that proteins expressed by liver-stage Plasmodium parasites can inhibit the translocation of transcription factors to the nucleus of different cells. This process would hinder the expression of immune genes, such as the CCL20 chemokine. OBJECTIVE: Since CCR6 is the only cognate receptor for CCL20, we investigated the importance of this chemokine-receptor axis against rodent malaria. METHODS: CCR6-deficient (KO) and wild-type (WT) C57BL/6 mice were challenged with Plasmodium berghei (Pb) NK65 sporozoites or infected red blood cells (iRBCs). Liver parasitic cDNA, parasitemia and serum cytokine concentrations were respectively evaluated through reverse transcription-polymerase chain reaction (RT-PCR), staining thin-blood smears with Giemsa solution, and enzyme-linked immunosorbent assay (ELISA). FINDINGS: Although the sporozoite challenges yielded similar liver parasitic cDNA and parasitemia, KO mice presented a prolonged survival than WT mice. After iRBC challenges, KO mice kept displaying higher survival rates as well as a decreased IL-12 p70 concentration in the serum than WT mice. CONCLUSION: Our data suggest that malaria triggered by PbNK65 liver- or blood-stage forms elicit a pro-inflammatory environment that culminates with a decreased survival of infected C57BL/6 mice.


Assuntos
Malária , Plasmodium berghei , Animais , DNA Complementar , Malária/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Parasitemia/parasitologia , Receptores CCR6
3.
Front Cell Infect Microbiol ; 12: 899581, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35677654

RESUMO

Malaria-associated acute respiratory distress syndrome (MA-ARDS) is increasingly gaining recognition as a severe malaria complication because of poor prognostic outcomes, high lethality rate, and limited therapeutic interventions. Unfortunately, invasive clinical studies are challenging to conduct and yields insufficient mechanistic insights. These limitations have led to the development of suitable MA-ARDS experimental mouse models. In patients and mice, MA-ARDS is characterized by edematous lung, along with marked infiltration of inflammatory cells and damage of the alveolar-capillary barriers. Although, the pathogenic pathways have yet to be fully understood, the use of different experimental mouse models is fundamental in the identification of mediators of pulmonary vascular damage. In this review, we discuss the current knowledge on endothelial activation, leukocyte recruitment, leukocyte induced-endothelial dysfunction, and other important findings, to better understand the pathogenesis pathways leading to endothelial pulmonary barrier lesions and increased vascular permeability. We also discuss how the advances in imaging techniques can contribute to a better understanding of the lung lesions induced during MA-ARDS, and how it could aid to monitor MA-ARDS severity.


Assuntos
Malária , Síndrome do Desconforto Respiratório , Animais , Modelos Animais de Doenças , Humanos , Pulmão/patologia , Malária/patologia , Camundongos , Camundongos Endogâmicos C57BL , Plasmodium berghei/fisiologia , Síndrome do Desconforto Respiratório/etiologia
4.
Mem. Inst. Oswaldo Cruz ; 117: e210287, 2022. graf
Artigo em Inglês | LILACS-Express | LILACS | ID: biblio-1386359

RESUMO

BACKGROUND It has been demonstrated that proteins expressed by liver-stage Plasmodium parasites can inhibit the translocation of transcription factors to the nucleus of different cells. This process would hinder the expression of immune genes, such as the CCL20 chemokine. OBJECTIVE Since CCR6 is the only cognate receptor for CCL20, we investigated the importance of this chemokine-receptor axis against rodent malaria. METHODS CCR6-deficient (KO) and wild-type (WT) C57BL/6 mice were challenged with Plasmodium berghei (Pb) NK65 sporozoites or infected red blood cells (iRBCs). Liver parasitic cDNA, parasitemia and serum cytokine concentrations were respectively evaluated through reverse transcription-polymerase chain reaction (RT-PCR), staining thin-blood smears with Giemsa solution, and enzyme-linked immunosorbent assay (ELISA). FINDINGS Although the sporozoite challenges yielded similar liver parasitic cDNA and parasitemia, KO mice presented a prolonged survival than WT mice. After iRBC challenges, KO mice kept displaying higher survival rates as well as a decreased IL-12 p70 concentration in the serum than WT mice. CONCLUSION Our data suggest that malaria triggered by PbNK65 liver- or blood-stage forms elicit a pro-inflammatory environment that culminates with a decreased survival of infected C57BL/6 mice.

5.
Infect Immun ; 89(10): e0002421, 2021 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-34251290

RESUMO

Malaria-associated acute respiratory distress syndrome (MA-ARDS) is a severe complication of malaria that occurs despite effective antimalarial treatment. Currently, noninvasive imaging procedures such as chest X-rays are used to assess edema in established MA-ARDS, but earlier detection methods are needed to reduce morbidity and mortality. The early stages of MA-ARDS are characterized by the infiltration of leukocytes, in particular monocytes/macrophages; thus, monitoring of immune infiltrates may provide a useful indicator of early pathology. In this study, Plasmodium berghei ANKA-infected C57BL/6 mice, a rodent model of MA-ARDS, were longitudinally imaged using the 18-kDa translocator protein (TSPO) imaging agent [18F]FEPPA as a marker of macrophage accumulation during the development of pathology and in response to combined artesunate and chloroquine diphosphate (ART+CQ) therapy. [18F]FEPPA uptake was compared to blood parasitemia levels and to levels of pulmonary immune cell infiltrates by using flow cytometry. Infected animals showed rapid increases in lung retention of [18F]FEPPA, correlating well with increases in blood parasitemia and pulmonary accumulation of interstitial inflammatory macrophages and major histocompatibility complex class II (MHC-II)-positive alveolar macrophages. Treatment with ART+CQ abrogated this increase in parasitemia and significantly reduced both lung uptake of [18F]FEPPA and levels of macrophage infiltrates. We conclude that retention of [18F]FEPPA in the lungs is well correlated with changes in blood parasitemia and levels of lung-associated macrophages during disease progression and in response to ART+CQ therapy. With further development, TSPO biomarkers may have the potential to accurately assess the early onset of MA-ARDS.


Assuntos
Biomarcadores/metabolismo , Pulmão/metabolismo , Malária/metabolismo , Pneumonia/metabolismo , Animais , Modelos Animais de Doenças , Leucócitos/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/metabolismo , Plasmodium berghei/patogenicidade , Tomografia por Emissão de Pósitrons/métodos , Síndrome do Desconforto Respiratório/metabolismo
7.
Nat Commun ; 10(1): 4241, 2019 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-31534124

RESUMO

Malaria-associated acute respiratory distress syndrome (ARDS) and acute lung injury (ALI) are life-threatening manifestations of severe malaria infections. The pathogenic mechanisms that lead to respiratory complications, such as vascular leakage, remain unclear. Here, we confirm that depleting CD8+T cells with anti-CD8ß antibodies in C57BL/6 mice infected with P. berghei ANKA (PbA) prevent pulmonary vascular leakage. When we transfer activated parasite-specific CD8+T cells into PbA-infected TCRß-/- mice (devoid of all T-cell populations), pulmonary vascular leakage recapitulates. Additionally, we demonstrate that PbA-infected erythrocyte accumulation leads to lung endothelial cell cross-presentation of parasite antigen to CD8+T cells in an IFNγ-dependent manner. In conclusion, pulmonary vascular damage in ALI is a consequence of IFNγ-activated lung endothelial cells capturing, processing, and cross-presenting malaria parasite antigen to specific CD8+T cells induced during infection. The mechanistic understanding of the immunopathogenesis in malaria-associated ARDS and ALI provide the basis for development of adjunct treatments.


Assuntos
Lesão Pulmonar Aguda/patologia , Linfócitos T CD8-Positivos/imunologia , Apresentação Cruzada/imunologia , Interferon gama/imunologia , Malária/imunologia , Síndrome do Desconforto Respiratório/patologia , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/parasitologia , Animais , Modelos Animais de Doenças , Células Endoteliais/imunologia , Feminino , Pulmão/parasitologia , Pulmão/patologia , Malária/tratamento farmacológico , Malária/parasitologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Plasmodium berghei/imunologia , Edema Pulmonar/parasitologia , Edema Pulmonar/patologia , Síndrome do Desconforto Respiratório/imunologia , Síndrome do Desconforto Respiratório/parasitologia
8.
Nat Commun ; 9(1): 3905, 2018 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-30254309

RESUMO

Co-infection with Plasmodium and chikungunya virus (CHIKV) has been reported in humans, but the impact of co-infection on pathogenesis remains unclear. Here, we show that prior exposure to Plasmodium suppresses CHIKV-associated pathologies in mice. Mechanistically, Plasmodium infection induces IFNγ, which reduces viraemia of a subsequent CHIKV infection and suppresses tissue viral load and joint inflammation. Conversely, concomitant infection with both pathogens limits the peak of joint inflammation with no effect on CHIKV viraemia. Reduced peak joint inflammation is regulated by elevated apoptosis of CD4+ T-cells in the lymph nodes and disrupted CXCR3-mediated CD4+ T-cell migration that abolishes their infiltration into the joints. Virus clearance from tissues is delayed in both infection scenarios, and is associated with a disruption of B cell affinity-maturation in the spleen that reduces CHIKV-neutralizing antibody production.


Assuntos
Febre de Chikungunya/imunologia , Vírus Chikungunya/imunologia , Coinfecção/imunologia , Malária/imunologia , Plasmodium/imunologia , Animais , Apoptose/imunologia , Artrite/genética , Artrite/imunologia , Artrite/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Febre de Chikungunya/virologia , Vírus Chikungunya/fisiologia , Coinfecção/parasitologia , Coinfecção/virologia , Feminino , Humanos , Interferon gama/genética , Interferon gama/imunologia , Interferon gama/metabolismo , Malária/metabolismo , Malária/parasitologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Plasmodium/fisiologia , Carga Viral/imunologia , Viremia/imunologia , Viremia/virologia
9.
Biomaterials ; 152: 77-85, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29111495

RESUMO

The study of blood brain barrier (BBB) functions is important for neurological disorder research. However, the lack of suitable tools and methods has hampered the progress of this field. Herein, we present a hybrid nanodot strategy, termed AIE-Gd dots, comprising of a fluorogen with aggregation-induced emission (AIE) characteristics as the core to provide bright and stable fluorescence for optical imaging, and gadolinium (Gd) for accurate quantification of vascular leakage via inductively-coupled plasma mass spectrometry (ICP-MS). In this report, we demonstrate that AIE-Gd dots enable direct visualization of brain vascular networks under resting condition, and that they form localized punctate aggregates and accumulate in the brain tissue during experimental cerebral malaria, indicative of hemorrhage and BBB malfunction. With its superior detection sensitivity and multimodality, we hereby propose that AIE-Gd dots can serve as a better alternative to Evans blue for visualization and quantification of changes in brain barrier functions.


Assuntos
Barreira Hematoencefálica/metabolismo , Encéfalo/diagnóstico por imagem , Complexos de Coordenação/química , Gadolínio/química , Nanopartículas Metálicas/química , Imagem Óptica/métodos , Animais , Encéfalo/irrigação sanguínea , Quelantes/química , Feminino , Corantes Fluorescentes/química , Luz , Malária Cerebral/diagnóstico por imagem , Camundongos Endogâmicos C57BL , Tamanho da Partícula , Pele/irrigação sanguínea , Pele/diagnóstico por imagem , Propriedades de Superfície , Distribuição Tecidual
10.
EMBO Mol Med ; 10(1): 121-138, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29113976

RESUMO

Arboviral diseases have risen significantly over the last 40 years, increasing the risk of co-infection with other endemic disease such as malaria. However, nothing is known about the impact arboviruses have on the host response toward heterologous pathogens during co-infection. Here, we investigate the effects of Chikungunya virus (CHIKV) co-infection on the susceptibility and severity of malaria infection. Using the Plasmodium berghei ANKA (PbA) experimental cerebral malaria (ECM) model, we show that concurrent co-infection induced the most prominent changes in ECM manifestation. Concurrent co-infection protected mice from ECM mortality without affecting parasite development in the blood. This protection was mediated by the alteration of parasite-specific CD8+ T-cell trafficking through an IFNγ-mediated mechanism. Co-infection with CHIKV induced higher splenic IFNγ levels that lead to high local levels of CXCL9 and CXCL10. This induced retention of CXCR3-expressing pathogenic CD8+ T cells in the spleen and prevented their migration to the brain. This then averts all downstream pathogenic events such as parasite sequestration in the brain and disruption of blood-brain barrier that prevents ECM-induced mortality in co-infected mice.


Assuntos
Encéfalo/patologia , Linfócitos T CD8-Positivos/patologia , Febre de Chikungunya/patologia , Vírus Chikungunya/fisiologia , Coinfecção/patologia , Malária Cerebral/patologia , Plasmodium berghei/fisiologia , Animais , Encéfalo/parasitologia , Encéfalo/virologia , Linfócitos T CD8-Positivos/parasitologia , Linfócitos T CD8-Positivos/virologia , Movimento Celular , Febre de Chikungunya/parasitologia , Febre de Chikungunya/virologia , Coinfecção/parasitologia , Coinfecção/virologia , Feminino , Malária Cerebral/parasitologia , Malária Cerebral/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuropatologia , Fatores de Proteção
11.
Cell Microbiol ; 20(5): e12819, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29281764

RESUMO

Host immune response has a key role in controlling the progression of malaria infection. In the well-established murine model of experimental cerebral malaria (ECM) with Plasmodium berghei ANKA infection, proinflammatory Th1 and CD8+ T cell response are essential for disease development. Interferon regulatory factor 1 (IRF1) is a transcription factor that promotes Th1 responses, and its absence was previously shown to protect from ECM death. Yet the exact mechanism of protection remains unknown. Here we demonstrated that IRF1-deficient mice (IRF1 knockout) were protected from ECM death despite displaying early neurological signs. Resistance to ECM death was a result of reduced parasite sequestration and pathogenic CD8+ T cells in the brain. Further analysis revealed that IRF1 deficiency suppress interferon-γ production and delayed CD8+ T cell proliferation. CXCR3 expression was found to be decreased in pathogenic CD8+ T cells, which limited their migration to the brain. In addition, reduced expression of adhesion molecules by brain endothelial cells hampered leucocyte retention in the brain. Taken together, these factors limited sequestration of pathogenic CD8+ T cells and consequently its ability to induce extensive damage to the blood-brain barrier.


Assuntos
Fator Regulador 1 de Interferon/genética , Malária Cerebral/genética , Plasmodium berghei/patogenicidade , Receptores CXCR3/genética , Animais , Encéfalo/microbiologia , Encéfalo/patologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/microbiologia , Movimento Celular/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Malária Cerebral/imunologia , Malária Cerebral/microbiologia , Camundongos , Camundongos Knockout
12.
Cell Microbiol ; 19(11)2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28664674

RESUMO

Artemisinin-based antimalarials, such as artesunate (ART), alone or in combination, are the mainstay of the therapy against malaria caused by Plasmodium falciparum. However, the emergence and spread of artemisinin resistance threatens the future success of its global malaria eradication. Although much of the reported artemisinin resistance can be attributed to mutations intrinsic to the parasite, a significant proportion of treatment failures are thought to be due to other factors such as the host's immune system. Exactly how the immune system participates in the clearance and elimination of malaria parasites during ART treatment is unknown. Here, we show that a developing primary immune response, involving both B and CD4+ T cells, is necessary for the complete elimination but not initial clearance, of Plasmodium yoelii YM parasites in mice treated with ART. Our study uncovers a dynamic interplay between ART and host adaptive immunity in Plasmodium sp. elimination.


Assuntos
Antimaláricos/uso terapêutico , Artemisininas/uso terapêutico , Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Malária/tratamento farmacológico , Plasmodium yoelii/efeitos dos fármacos , Plasmodium yoelii/imunologia , Imunidade Adaptativa/imunologia , Animais , Artesunato , Modelos Animais de Doenças , Resistência a Medicamentos , Feminino , Malária/imunologia , Malária/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Recidiva
13.
Infect Immun ; 85(6)2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28396319

RESUMO

The resolution of malaria infection is dependent on a balance between proinflammatory and regulatory immune responses. While early effector T cell responses are required for limiting parasitemia, these responses need to be switched off by regulatory mechanisms in a timely manner to avoid immune-mediated tissue damage. Interleukin-10 receptor (IL-10R) signaling is considered to be a vital component of regulatory responses, although its role in host resistance to severe immune pathology during acute malaria infections is not fully understood. In this study, we have determined the contribution of IL-10R signaling to the regulation of immune responses during Plasmodium berghei ANKA-induced experimental cerebral malaria (ECM). We show that antibody-mediated blockade of the IL-10R during P. berghei ANKA infection in ECM-resistant BALB/c mice leads to amplified T cell activation, higher serum gamma interferon (IFN-γ) concentrations, enhanced intravascular accumulation of both parasitized red blood cells and CD8+ T cells to the brain, and an increased incidence of ECM. Importantly, the pathogenic effects of IL-10R blockade during P. berghei ANKA infection were reversible by depletion of T cells and neutralization of IFN-γ. Our findings underscore the importance of IL-10R signaling in preventing T-cell- and cytokine-mediated pathology during potentially lethal malaria infections.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Interferon gama/sangue , Malária Cerebral/imunologia , Plasmodium berghei/imunologia , Receptores de Interleucina-10/imunologia , Animais , Anticorpos Bloqueadores/administração & dosagem , Anticorpos Neutralizantes/administração & dosagem , Encéfalo/patologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Eritrócitos/efeitos dos fármacos , Eritrócitos/parasitologia , Feminino , Fígado/patologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Parasitemia/imunologia , Receptores de Interleucina-10/antagonistas & inibidores , Transdução de Sinais
14.
Antimicrob Agents Chemother ; 60(11): 6859-6866, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27600050

RESUMO

Ex vivo assay systems provide a powerful approach to studying human malaria parasite biology and to testing antimalarials. For rodent malaria parasites, short-term in vitro culture and ex vivo antimalarial susceptibility assays are relatively cumbersome, relying on in vivo passage for synchronization, since ring-stage parasites are an essential starting material. Here, we describe a new approach based on the enrichment of ring-stage Plasmodium berghei, P. yoelii, and P. vinckei vinckei using a single-step Percoll gradient. Importantly, we demonstrate that the enriched ring-stage parasites develop synchronously regardless of the parasite strain or species used. Using a flow cytometry assay with Hoechst and ethidium or MitoTracker dye, we show that parasite development is easily and rapidly monitored. Finally, we demonstrate that this approach can be used to screen antimalarial drugs.


Assuntos
Antimaláricos/farmacologia , Avaliação Pré-Clínica de Medicamentos/métodos , Malária/parasitologia , Plasmodium/efeitos dos fármacos , Plasmodium/fisiologia , Animais , Modelos Animais de Doenças , Feminino , Citometria de Fluxo/métodos , Malária/tratamento farmacológico , Masculino , Camundongos Endogâmicos C57BL , Testes de Sensibilidade Microbiana , Plasmodium/patogenicidade , Plasmodium berghei/efeitos dos fármacos , Plasmodium berghei/patogenicidade , Plasmodium berghei/fisiologia
15.
Cell Rep ; 16(6): 1749-1761, 2016 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-27477286

RESUMO

Tissue macrophages exhibit diverse functions, ranging from the maintenance of tissue homeostasis, including clearance of senescent erythrocytes and cell debris, to modulation of inflammation and immunity. Their contribution to the control of blood-stage malaria remains unclear. Here, we show that in the absence of tissue-resident CD169(+) macrophages, Plasmodium berghei ANKA (PbA) infection results in significantly increased parasite sequestration, leading to vascular occlusion and leakage and augmented tissue deposition of the malarial pigment hemozoin. This leads to widespread tissue damage culminating in multiple organ inflammation. Thus, the capacity of CD169(+) macrophages to contain the parasite burden and its sequestration into different tissues and to limit infection-induced inflammation is crucial to mitigating Plasmodium infection and pathogenesis.


Assuntos
Macrófagos/parasitologia , Malária/imunologia , Plasmodium berghei/parasitologia , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico/genética , Animais , Eritrócitos/parasitologia , Hemeproteínas/metabolismo , Macrófagos/patologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
16.
Nat Protoc ; 10(12): 2016-26, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26562622

RESUMO

We have recently demonstrated that brain endothelial cells cross-present parasite antigen during mouse experimental cerebral malaria (ECM). Here we describe a 2-d protocol to detect cross-presentation by isolating the brain microvessels and incubating them with a reporter cell line that expresses lacZ upon detection of the relevant peptide-major histocompatibility complex. After X-gal staining, a typical positive result consists of hundreds of blue spots, compared with fewer than 20 spots from a naive brain. The assay is generalizable to other disease contexts by using reporter cells that express appropriate specific T cell receptors. Also described is the protocol for culturing endothelial cells from brain microvessels isolated from naive mice. After 7-10 d, an in vitro cross-presentation assay can be performed by adding interferon-γ, antigen (e.g., Plasmodium berghei-infected red blood cells) and reporter cells in sequence over 3 d. This is useful for comparing different antigen forms or for probing the effects of various interventions.


Assuntos
Apresentação de Antígeno , Encéfalo/imunologia , Células Endoteliais/imunologia , Microvasos/imunologia , Animais , Encéfalo/irrigação sanguínea , Encéfalo/citologia , Técnicas de Cultura de Células , Linhagem Celular , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/citologia , Complexo Principal de Histocompatibilidade , Malária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Microvasos/citologia , Plasmodium berghei/imunologia , Receptores de Antígenos de Linfócitos T/imunologia
17.
Infect Immun ; 83(10): 3781-92, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26169268

RESUMO

Malaria remains a world-threatening disease largely because of the lack of a long-lasting and fully effective vaccine. MAEBL is a type 1 transmembrane molecule with a chimeric cysteine-rich ectodomain homologous to regions of the Duffy binding-like erythrocyte binding protein and apical membrane antigen 1 (AMA1) antigens. Although MAEBL does not appear to be essential for the survival of blood-stage forms, ectodomains M1 and M2, homologous to AMA1, seem to be involved in parasite attachment to erythrocytes, especially M2. MAEBL is necessary for sporozoite infection of mosquito salivary glands and is expressed in liver stages. Here, the Plasmodium yoelii MAEBL-M2 domain was expressed in a prokaryotic vector. C57BL/6J mice were immunized with doses of P. yoelii recombinant protein rPyM2-MAEBL. High levels of antibodies, with balanced IgG1 and IgG2c subclasses, were achieved. rPyM2-MAEBL antisera were capable of recognizing the native antigen. Anti-MAEBL antibodies recognized different MAEBL fragments expressed in CHO cells, showing stronger IgM and IgG responses to the M2 domain and repeat region, respectively. After a challenge with P. yoelii YM (lethal strain)-infected erythrocytes (IE), up to 90% of the immunized animals survived and a reduction of parasitemia was observed. Moreover, splenocytes harvested from immunized animals proliferated in a dose-dependent manner in the presence of rPyM2-MAEBL. Protection was highly dependent on CD4(+), but not CD8(+), T cells toward Th1. rPyM2-MAEBL antisera were also able to significantly inhibit parasite development, as observed in ex vivo P. yoelii erythrocyte invasion assays. Collectively, these findings support the use of MAEBL as a vaccine candidate and open perspectives to understand the mechanisms involved in protection.


Assuntos
Vacinas Antimaláricas/imunologia , Malária/prevenção & controle , Plasmodium yoelii/imunologia , Proteínas de Protozoários/química , Proteínas de Protozoários/imunologia , Animais , Anticorpos Antiprotozoários/imunologia , Eritrócitos/parasitologia , Feminino , Humanos , Imunização , Malária/imunologia , Malária/mortalidade , Malária/parasitologia , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/química , Vacinas Antimaláricas/genética , Masculino , Merozoítos/química , Merozoítos/crescimento & desenvolvimento , Merozoítos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Plasmodium yoelii/química , Plasmodium yoelii/genética , Plasmodium yoelii/crescimento & desenvolvimento , Estrutura Terciária de Proteína , Proteínas de Protozoários/administração & dosagem , Proteínas de Protozoários/genética , Esporozoítos/química , Esporozoítos/crescimento & desenvolvimento , Esporozoítos/imunologia
18.
Semin Immunopathol ; 37(3): 221-31, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25772948

RESUMO

Cerebral malaria (CM) is one the major complications occurring during malaria infection. The mechanisms leading to this syndrome are still not completely understood. Although it is clear that parasite sequestration is the key initiation factor, the downstream pathological processes are still highly debated. The experimental cerebral malaria (ECM) model, in which susceptible mice are infected with Plasmodium berghei ANKA, has led to the identification of CD8(+) T cells as the major mediator of ECM death. In this review, we discuss the recent advances and future developments in the understanding of the role of CD8(+) T cells in CM.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Interações Hospedeiro-Parasita/imunologia , Malária Cerebral/imunologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Movimento Celular , Citotoxicidade Imunológica , Modelos Animais de Doenças , Epitopos de Linfócito T/química , Epitopos de Linfócito T/imunologia , Matriz Extracelular/imunologia , Humanos , Imunomodulação , Malária Cerebral/parasitologia , Fenótipo , Plasmodium/imunologia
19.
Blood ; 125(8): 1314-24, 2015 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-25414440

RESUMO

Plasmodium vivax merozoites only invade reticulocytes, a minor though heterogeneous population of red blood cell precursors that can be graded by levels of transferrin receptor (CD71) expression. The development of a protocol that allows sorting reticulocytes into defined developmental stages and a robust ex vivo P vivax invasion assay has made it possible for the first time to investigate the fine-scale invasion preference of P vivax merozoites. Surprisingly, it was the immature reticulocytes (CD71(+)) that are generally restricted to the bone marrow that were preferentially invaded, whereas older reticulocytes (CD71(-)), principally found in the peripheral blood, were rarely invaded. Invasion assays based on the CD71(+) reticulocyte fraction revealed substantial postinvasion modification. Thus, 3 to 6 hours after invasion, the initially biomechanically rigid CD71(+) reticulocytes convert into a highly deformable CD71(-) infected red blood cell devoid of host reticular matter, a process that normally spans 24 hours for uninfected reticulocytes. Concurrent with these changes, clathrin pits disappear by 3 hours postinvasion, replaced by distinctive caveolae nanostructures. These 2 hitherto unsuspected features of P vivax invasion, a narrow preference for immature reticulocytes and a rapid remodeling of the host cell, provide important insights pertinent to the pathobiology of the P vivax infection.


Assuntos
Antígenos CD/metabolismo , Plasmodium vivax/crescimento & desenvolvimento , Receptores da Transferrina/metabolismo , Reticulócitos/fisiologia , Reticulócitos/parasitologia , Tropismo/fisiologia , Fenômenos Biomecânicos , Células Cultivadas , Deformação Eritrocítica , Humanos , Malária Vivax/sangue , Malária Vivax/parasitologia , Reticulócitos/metabolismo
20.
Eur J Immunol ; 45(1): 130-41, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25319247

RESUMO

Type I IFN signaling suppresses splenic T helper 1 (Th1) responses during blood-stage Plasmodium berghei ANKA (PbA) infection in mice, and is crucial for mediating tissue accumulation of parasites and fatal cerebral symptoms via mechanisms that remain to be fully characterized. Interferon regulatory factor 7 (IRF7) is considered to be a master regulator of type I IFN responses. Here, we assessed IRF7 for its roles during lethal PbA infection and nonlethal Plasmodium chabaudi chabaudi AS (PcAS) infection as two distinct models of blood-stage malaria. We found that IRF7 was not essential for tissue accumulation of parasites, cerebral symptoms, or brain pathology. Using timed administration of anti-IFNAR1 mAb, we show that late IFNAR1 signaling promotes fatal disease via IRF7-independent mechanisms. Despite this, IRF7 significantly impaired early splenic Th1 responses and limited control of parasitemia during PbA infection.  Finally, IRF7 also suppressed antiparasitic immunity and Th1 responses during nonlethal PcAS infection. Together, our data support a model in which IRF7 suppresses antiparasitic immunity in the spleen, while IFNAR1-mediated, but IRF7-independent, signaling contributes to pathology in the brain during experimental blood-stage malaria.


Assuntos
Encéfalo/imunologia , Fator Regulador 7 de Interferon/imunologia , Malária Cerebral/imunologia , Receptor de Interferon alfa e beta/imunologia , Baço/imunologia , Células Th1/imunologia , Animais , Anticorpos Monoclonais/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/parasitologia , Suscetibilidade a Doenças , Eritrócitos/parasitologia , Feminino , Regulação da Expressão Gênica , Interações Hospedeiro-Parasita , Fator Regulador 7 de Interferon/genética , Malária Cerebral/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Plasmodium berghei/imunologia , Plasmodium chabaudi/imunologia , Receptor de Interferon alfa e beta/antagonistas & inibidores , Receptor de Interferon alfa e beta/genética , Transdução de Sinais , Baço/efeitos dos fármacos , Baço/parasitologia , Células Th1/parasitologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...