Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Malar J ; 16(1): 463, 2017 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-29137631

RESUMO

BACKGROUND: Cerebral malaria (CM) is a severe complication of Plasmodium falciparum infection associated with high mortality and neurocognitive impairment in survivors. New anti-malarials and host-based adjunctive therapy may improve clinical outcome in CM. Synthetic oleanane triterpenoid (SO) compounds have shown efficacy in the treatment of diseases where inflammation and oxidative stress contribute to pathogenesis. METHODS: A derivative of the SO 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid (CDDO), CDDO-ethyl amide (CDDO-EA) was investigated for the treatment of severe malaria in a pre-clinical model. CDDO-EA was evaluated in vivo as a monotherapy as well as adjunctive therapy with parenteral artesunate in the Plasmodium berghei strain ANKA experimental cerebral malaria (ECM) model. RESULTS: CDDO-EA alone improved outcome in ECM and, given as adjunctive therapy in combination with artesunate, it significantly improved outcome over artesunate alone (p = 0.009). Improved survival was associated with reduced inflammation, enhanced endothelial stability and blood-brain barrier integrity. Survival was improved even when administered late in the disease course after the onset of neurological symptoms. CONCLUSIONS: These results indicate that SO are a new class of immunomodulatory drugs and support further studies investigating this class of agents as potential adjunctive therapy for severe malaria.


Assuntos
Antimaláricos/farmacologia , Barreira Hematoencefálica/efeitos dos fármacos , Endotélio/efeitos dos fármacos , Inflamação/tratamento farmacológico , Longevidade/efeitos dos fármacos , Malária Cerebral/fisiopatologia , Ácido Oleanólico/análogos & derivados , Animais , Barreira Hematoencefálica/fisiopatologia , Endotélio/parasitologia , Feminino , Inflamação/parasitologia , Malária Cerebral/tratamento farmacológico , Masculino , Camundongos Endogâmicos C57BL , Ácido Oleanólico/farmacologia , Ratos
2.
Infect Immun ; 85(9)2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28674030

RESUMO

Artesunate remains the mainstay of treatment for cerebral malaria, but it is less effective in later stages of disease when the host inflammatory response and blood-brain barrier integrity dictate clinical outcomes. Nitric oxide (NO) is an important regulator of inflammation and microvascular integrity, and impaired NO bioactivity is associated with fatal outcomes in malaria. Endogenous NO bioactivity in mammals is largely mediated by S-nitrosothiols (SNOs). Based on these observations, we hypothesized that animals deficient in the SNO-metabolizing enzyme, S-nitrosoglutathione reductase (GSNOR), which exhibit enhanced S-nitrosylation, would have improved outcomes in a preclinical model of cerebral malaria. GSNOR knockout (KO) mice infected with Plasmodium berghei ANKA had significantly delayed mortality compared to WT animals (P < 0.0001), despite higher parasite burdens (P < 0.01), and displayed markedly enhanced survival versus the wild type (WT) when treated with the antimalarial drug artesunate (77% versus 38%; P < 0.001). Improved survival was associated with higher levels of protein-bound NO, decreased levels of CD4+ and CD8+ T cells in the brain, improved blood-brain barrier integrity, and improved coma scores, as well as higher levels of gamma interferon. GSNOR KO animals receiving WT bone marrow had significantly reduced survival following P. berghei ANKA infection compared to those receiving KO bone barrow (P < 0.001). Reciprocal transplants established that survival benefits of GSNOR deletion were attributable primarily to the T cell compartment. These data indicate a role for GSNOR in the host response to malaria infection and suggest that strategies to disrupt its activity will improve clinical outcomes by enhancing microvascular integrity and modulating T cell tissue tropism.


Assuntos
Álcool Desidrogenase/deficiência , Malária Cerebral/patologia , Plasmodium berghei/patogenicidade , Animais , Antimaláricos/administração & dosagem , Artemisininas/administração & dosagem , Artesunato , Modelos Animais de Doenças , Feminino , Malária Cerebral/tratamento farmacológico , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico/metabolismo , Análise de Sobrevida , Linfócitos T/imunologia , Resultado do Tratamento
3.
Mol Med ; 23: 24-33, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28182191

RESUMO

miR-155 has been shown to participate in host response to infection and neuro-inflammation via negative regulation of blood-brain-barrier (BBB) integrity and T cell function. We hypothesized that miR-155 may contribute to the pathogenesis of cerebral malaria (CM). To test this hypothesis, we used a genetic approach to modulate miR-155 expression in an experimental model of cerebral malaria (ECM). In addition, an engineered endothelialized microvessel system and serum samples from Ugandan children with CM were used to examine an anti-miR-155 as a potential adjunctive therapeutic for severe malaria. Despite higher parasitemia, survival was significantly improved in miR-155-/- mice vs. wild-type littermate mice in ECM. Improved survival was associated with preservation of BBB integrity and reduced endothelial activation, despite increased levels of pro-inflammatory cytokines. Pre-treatment with antagomir-155 reduced vascular leak induced by human CM sera in an ex vivo endothelial microvessel model. These data provide evidence supporting a mechanistic role for miR-155 in host response to malaria via regulation of endothelial activation, microvascular leak and BBB dysfunction in CM.

4.
Infect Immun ; 84(7): 2002-2011, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27091932

RESUMO

CD47 engagement by the macrophage signal regulatory protein alpha (SIRPα) inhibits phagocytic activity and protects red blood cells (RBCs) from erythrophagocytosis. The role of CD47-SIRPα in the innate immune response to Plasmodium falciparum infection is unknown. We hypothesized that disruption of SIRPα signaling may enhance macrophage uptake of malaria parasite-infected RBCs. To test this hypothesis, we examined in vivo clearance in CD47-deficient mice infected with Plasmodium berghei ANKA and in vitro phagocytosis of P. falciparum-infected RBCs by macrophages from SHP-1-deficient (Shp-1(-/-)) mice and NOD.NOR-Idd13.Prkdc(scid) (NS-Idd13) mice, as well as human macrophages, following disruption of CD47-SIRPα interactions with anti-SIRPα antibodies or recombinant SIRPα-Fc fusion protein. Compared to their wild-type counterparts, Cd47(-/-) mice displayed significantly lower parasitemia, decreased endothelial activation, and enhanced survival. Using macrophages from SHP-1-deficient mice or from NS-Idd13 mice, which express a SIRPα variant that does not bind human CD47, we showed that altered SIRPα signaling resulted in enhanced phagocytosis of P. falciparum-infected RBCs. Moreover, disrupting CD47-SIRPα engagement using anti-SIRPα antibodies or SIRPα-Fc fusion protein also increased phagocytosis of P. falciparum-infected RBCs. These results indicate an important role for CD47-SIRPα interactions in innate control of malaria and suggest novel targets for intervention.


Assuntos
Antígeno CD47/metabolismo , Macrófagos/fisiologia , Macrófagos/parasitologia , Malária Falciparum/imunologia , Malária Falciparum/metabolismo , Malária Falciparum/parasitologia , Plasmodium falciparum/imunologia , Receptores Imunológicos/metabolismo , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Eritrócitos/parasitologia , Interações Hospedeiro-Parasita , Imunidade Inata , Camundongos , Camundongos Knockout , Fagocitose/imunologia , Plasmodium falciparum/crescimento & desenvolvimento , Ligação Proteica , Transdução de Sinais
5.
Malar J ; 13: 279, 2014 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-25047113

RESUMO

BACKGROUND: Severe and fatal malaria are associated with dysregulated host inflammatory responses to infection. Chitinase 3-like 1 (CHI3L1) is a secreted glycoprotein implicated in regulating immune responses. Expression and function of CHI3L1 in malaria infection were investigated. METHODS: Plasma levels of CHI3L1 were quantified in a case-control study of Ugandan children presenting with Plasmodium falciparum malaria. CHI3L1 levels were compared in children with uncomplicated malaria (UM; n = 53), severe malarial anaemia (SMA; n = 59) and cerebral malaria (CM; n = 44) using the Kruskall Wallis-test, and evaluated for utility in predicting fatal (n = 23) versus non-fatal (n = 80) outcomes in severe disease using the Mann Whitney U test, receiver operating characteristic curves, and combinatorial analysis. Co-culture of P. falciparum with human peripheral blood mononuclear cells and the Plasmodium berghei ANKA experimental model of cerebral malaria were used to examine the role of CHI3L1 in severe malaria. RESULTS: In children presenting with falciparum malaria, CHI3L1 levels were increased in SMA and CM versus UM (p < 0.001). Among severe malaria cases, CHI3L1 levels at presentation predicted subsequent death (area under receiver operating characteristic curve 0.84 [95% CI 0.76-0.92]) and in combination with other host biomarkers, predicted mortality with high sensitivity (100% [85.7-100]) and specificity (81.3% [71.3-88.3]). Plasmodium falciparum stimulated CHI3L1 production by human peripheral blood mononuclear cells in vitro. CHI3L1 was increased in plasma and brain tissue in experimental cerebral malaria, but targeted Chi3l1 deletion did not alter cytokine production or survival in this model. CONCLUSIONS: These data suggest that plasma CHI3L1 measured at presentation correlates with malaria severity and predicts outcome in paediatric SMA and CM, but do not support a causal role for CHI3L1 in cerebral malaria pathobiology in the model tested.


Assuntos
Adipocinas/sangue , Lectinas/sangue , Malária Cerebral/sangue , Malária Falciparum/sangue , Adipocinas/biossíntese , Adipocinas/genética , Anemia/sangue , Anemia/etiologia , Animais , Área Sob a Curva , Biomarcadores , Química Encefálica , Estudos de Casos e Controles , Criança , Pré-Escolar , Proteína 1 Semelhante à Quitinase-3 , Feminino , Regulação da Expressão Gênica , Glicoproteínas/deficiência , Glicoproteínas/genética , Glicoproteínas/fisiologia , Interações Hospedeiro-Parasita , Humanos , Lactente , Lectinas/biossíntese , Lectinas/genética , Leucócitos Mononucleares/metabolismo , Malária/sangue , Malária/genética , Malária Cerebral/mortalidade , Malária Falciparum/complicações , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Plasmodium berghei , Plasmodium falciparum/fisiologia , Prognóstico , Estudos Prospectivos , Curva ROC , Estatísticas não Paramétricas , Células Th1/imunologia , Uganda/epidemiologia
6.
PLoS Pathog ; 10(3): e1003980, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24603727

RESUMO

Cerebral malaria (CM) is associated with a high mortality rate, and long-term neurocognitive impairment in approximately one third of survivors. Adjunctive therapies that modify the pathophysiological processes involved in CM may improve outcome over anti-malarial therapy alone. PPARγ agonists have been reported to have immunomodulatory effects in a variety of disease models. Here we report that adjunctive therapy with PPARγ agonists improved survival and long-term neurocognitive outcomes in the Plasmodium berghei ANKA experimental model of CM. Compared to anti-malarial therapy alone, PPARγ adjunctive therapy administered to mice at the onset of CM signs, was associated with reduced endothelial activation, and enhanced expression of the anti-oxidant enzymes SOD-1 and catalase and the neurotrophic factors brain derived neurotrophic factor (BDNF) and nerve growth factor (NGF) in the brains of infected mice. Two months following infection, mice that were treated with anti-malarials alone demonstrated cognitive dysfunction, while mice that received PPARγ adjunctive therapy were completely protected from neurocognitive impairment and from PbA-infection induced brain atrophy. In humans with P. falciparum malaria, PPARγ therapy was associated with reduced endothelial activation and with induction of neuroprotective pathways, such as BDNF. These findings provide insight into mechanisms conferring improved survival and preventing neurocognitive injury in CM, and support the evaluation of PPARγ agonists in human CM.


Assuntos
Antimaláricos/farmacologia , Encéfalo/efeitos dos fármacos , Malária Cerebral/complicações , PPAR gama/antagonistas & inibidores , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Fator Neurotrófico Derivado do Encéfalo/análise , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Malária Cerebral/metabolismo , Malária Cerebral/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fármacos Neuroprotetores/farmacologia , Ensaios Clínicos Controlados Aleatórios como Assunto , Reação em Cadeia da Polimerase em Tempo Real , Ribonuclease Pancreático/análise , Rosiglitazona , Tiazolidinedionas/farmacologia
7.
Infect Immun ; 82(1): 371-9, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24191300

RESUMO

The host immune response plays an important role in the onset and progression of cerebral malaria (CM). The complement system is an essential component of the innate immune response to malaria, and its activation generates the anaphylatoxin C5a. To test the hypothesis that C5a signaling contributes to the pathogenesis of CM, we investigated a causal role for the C5a receptors C5aR and C5L2 in a mouse model of experimental CM (ECM) induced by Plasmodium berghei ANKA infection, and using a case-control design, we examined levels of C5a in plasma samples from Ugandan children presenting with CM or uncomplicated malaria (UM). In the ECM model, C5aR(-/-) mice displayed significantly improved survival compared to their wild-type (WT) counterparts (P = 0.004), whereas C5L2(-/-) mice showed no difference in survival from WT mice. Improved survival in C5aR(-/-) mice was associated with reduced levels of the proinflammatory cytokines tumor necrosis factor (TNF) and gamma interferon (IFN-γ) and the chemokine, monocyte chemoattractant protein 1 (MCP-1) (CCL2). Furthermore, endothelial integrity was enhanced, as demonstrated by increased levels of angiopoietin-1, decreased levels of angiopoietin-2 and soluble ICAM-1, and decreased Evans blue extravasation into brain parenchyma. In the case-control study, the median levels of C5a at presentation were significantly higher in children with CM versus those in children with UM (43.7 versus 22.4 ng/ml; P < 0.001). These findings demonstrate that C5a is dysregulated in human CM and contributes to the pathogenesis of ECM via C5aR-dependent inflammation and endothelial dysfunction.


Assuntos
Complemento C5a/imunologia , Malária Cerebral/imunologia , Receptores de Quimiocinas/imunologia , Receptores de Complemento/imunologia , Animais , Estudos de Casos e Controles , Criança , Pré-Escolar , Complemento C5a/deficiência , Modelos Animais de Doenças , Feminino , Humanos , Lactente , Inflamação/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptor da Anafilatoxina C5a , Receptores de Complemento/deficiência , Receptores de Concanavalina A
8.
PLoS One ; 6(11): e27714, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22110737

RESUMO

The host immune response contributes to the onset and progression of severe malaria syndromes, such as cerebral malaria. Adjunctive immunomodulatory strategies for severe malaria may improve clinical outcome beyond that achievable with artemisinin-based therapy alone. Here, we report that prophylaxis with inhaled nitric oxide significantly reduced systemic inflammation (lower TNF, IFNγ and MCP-1 in peripheral blood) and endothelial activation (decreased sICAM-1 and vWF, and increased angiopoeitin-1 levels in peripheral blood) in an experimental cerebral malaria model. Mice that received inhaled nitric oxide starting prior to infection had reduced parasitized erythrocyte accumulation in the brain, decreased brain expression of ICAM-1, and preserved vascular integrity compared to control mice.Inhaled nitric oxide administered in combination with artesunate, starting as late as 5.5 days post-infection, improved survival over treatment with artesunate alone (70% survival in the artesunate only vs. 100% survival in the artesunate plus iNO group, p = 0.03). These data support the clinical investigation of inhaled nitric oxide as a novel adjunctive therapy in patients with severe malaria.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/parasitologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Malária Cerebral/tratamento farmacológico , Óxido Nítrico/administração & dosagem , Óxido Nítrico/farmacologia , Administração por Inalação , Animais , Artemisininas/farmacologia , Artesunato , Vasos Sanguíneos/efeitos dos fármacos , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Células Endoteliais/metabolismo , Células Endoteliais/parasitologia , Regulação da Expressão Gênica/efeitos dos fármacos , Inflamação/tratamento farmacológico , Molécula 1 de Adesão Intercelular/metabolismo , Malária Cerebral/metabolismo , Malária Cerebral/patologia , Malária Cerebral/fisiopatologia , Masculino , Camundongos , Óxido Nítrico/uso terapêutico , Plasmodium berghei/patogenicidade , Análise de Sobrevida , Fatores de Tempo
9.
Am J Trop Med Hyg ; 83(1): 69-74, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20595480

RESUMO

Severe malaria represents a clinical spectrum of disease. We propose that innate immune inflammatory responses to malaria play key roles in the pathogenesis and clinical outcomes of distinct severe malaria syndromes. To investigate this hypothesis, mice deficient in IRAK4, central to Toll-like receptor (TLR)-mediated signaling, were studied in two experimental models of malaria: Plasmodium berghei (PbA) and Plasmodium chabaudi (PccAS). Irak4(-/-)mice had decreased pro-inflammatory cytokine production during infection in both models. However, animals were relatively protected from PbA-associated symptoms compared with wild-type mice, whereas Irak4(-/-) animals were more susceptible to PccAS-associated disease. These results show that IRAK4-mediated innate immune inflammatory responses play critical roles in divergent clinical outcomes in murine malaria models. As such, integrated approaches, using more than one model, are required to fully understand the parasite/host interactions that characterize severe malaria, and more importantly, to fully assess the effect of adjunctive therapies targeting innate immune responses to malaria.


Assuntos
Imunidade Inata/imunologia , Quinases Associadas a Receptores de Interleucina-1/imunologia , Malária Falciparum/imunologia , Malária/imunologia , Receptores Toll-Like/fisiologia , Animais , Modelos Animais de Doenças , Humanos , Quinases Associadas a Receptores de Interleucina-1/genética , Quinases Associadas a Receptores de Interleucina-1/fisiologia , Camundongos
10.
J Inflamm (Lond) ; 7: 30, 2010 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-20565784

RESUMO

Although Fas (CD95) is recognized as a death receptor that induces apoptosis, recent studies indicate that the Fas/FasL system can induce pro-inflammatory cytokine production by macrophages independent of conventional caspase-mediated apoptotic signaling. The precise mechanism(s) by which Fas activates macrophage inflammation is unknown. We hypothesized that Fas stimulates rapid release of high mobility group box 1 (HMGB1) that acts in an autocrine and/or paracrine manner to stimulate pro-inflammatory cytokine production via a Toll-like receptor-4 (TLR4)/Interleukin-1 receptor associated kinase-4 (IRAK4)-dependent mechanism. Following Fas activation, HMGB1 was released within 1 hr from viable RAW267.4 cells and primary murine peritoneal macrophages. HMGB1 release was more rapid following Fas activation compared to LPS stimulation. Neutralization of HMGB1 with an inhibitory anti-HMGB1 monoclonal antibody strongly inhibited Fas-induced production of tumor necrosis factor (TNF) and macrophage inflammatory protein-2 (MIP-2). Both Fas-induced HMGB1 release and associated pro-inflammatory cytokine production were significantly decreased from Tlr4-/- and Irak4-/- macrophages, but not Tlr2-/- macrophages. These findings reveal a novel mechanism underlying Fas-mediated pro-inflammatory physiological responses in macrophages. We conclude that Fas activation induces rapid, TLR4/IRAK4-dependent release of HMGB1 that contributes to Fas-mediated pro-inflammatory cytokine production by viable macrophages.

11.
Am J Trop Med Hyg ; 80(5): 718-22, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19407112

RESUMO

Research relating to host inflammatory processes during malaria infection has focused on Toll-like receptors, membrane-bound receptors implicated in innate sensing, and phagocytosis of parasitized erythrocytes by host cells. This is the first study to examine the role of Nod proteins, members of the Nod-like receptor (NLR) family of cytoplasmic proteins involved in pathogen recognition, in a murine model of cerebral malaria (Plasmodium berghei ANKA, PbA). Here, we find that nod1nod2(-/-) mice infected with PbA show no difference in survival or parasitemia compared with wild-type infected animals. However, cytokine levels, notably those associated with NLR activation including interleukin (IL)1-beta, KC, and MCP-1, and proteins linked to malaria pathogenesis, such as interferon-gamma (IFN-gamma), were decreased in the nod-1nod2(-/-) animals. We therefore demonstrate for the first time that Nod proteins are activated in response to parasites, and they play a role in regulating host inflammatory responses during malaria infection.


Assuntos
Malária Cerebral/imunologia , Proteína Adaptadora de Sinalização NOD1/genética , Proteína Adaptadora de Sinalização NOD2/genética , Plasmodium berghei , Animais , Modelos Animais de Doenças , Inflamação , Malária Cerebral/mortalidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Adaptadora de Sinalização NOD1/metabolismo , Proteína Adaptadora de Sinalização NOD2/metabolismo , Parasitemia
12.
J Infect Dis ; 199(10): 1536-45, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19392627

RESUMO

For severe malarial syndromes such as cerebral malaria, adverse clinical outcomes are often mediated by the immune system rather than caused by the parasite directly. However, few therapeutic agents have been developed to modulate the host's immunopathological responses to infection. Here, we report that the peroxisome proliferator-activated receptor gamma (PPARgamma) agonist rosiglitazone modulated the host response to malaria by enhancing phagocytic clearance of malaria-parasitized erythrocytes and by decreasing inflammatory responses to infection via inhibition of Plasmodium falciparum glycosylphosphatidylinositol-induced activation of the mitogen-activated protein kinase (MAPK) and nuclear factor-kappaB (NF-kappaB) signaling pathways. We found that, in the Plasmodium berghei strain ANKA experimental model of cerebral malaria, rosiglitazone modified the inflammatory response to malarial infection and improved the survival rate even when treatment was initiated as late as day 5 after infection. Furthermore, rosiglitazone reduced the parasitemia in a CD36-dependent manner in the Plasmodium chabaudi chabaudi hyperparasitemia model. These data suggest that PPARgamma agonists represent a novel class of host immunomodulatory drugs that may be useful for treatment of severe malaria syndromes.


Assuntos
Imunidade Inata/efeitos dos fármacos , Malária Cerebral/imunologia , Malária Falciparum/imunologia , Plasmodium falciparum/imunologia , Tiazolidinedionas/farmacologia , Animais , Antígenos CD36/análise , Citometria de Fluxo , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Malária Falciparum/sangue , Camundongos , Fagocitose , Plasmodium falciparum/efeitos dos fármacos , Rosiglitazona , Transdução de Sinais , Sobreviventes
13.
J Immunol ; 178(6): 3954-61, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17339496

RESUMO

CD36 is a scavenger receptor that has been implicated in malaria pathogenesis as well as innate defense against blood-stage infection. Inflammatory responses to Plasmodium falciparum GPI (pfGPI) anchors are believed to play an important role in innate immune response to malaria. We investigated the role of CD36 in pfGPI-induced MAPK activation and proinflammatory cytokine secretion. Furthermore, we explored the role of this receptor in an experimental model of acute malaria in vivo. We demonstrate that ERK1/2, JNK, p38, and c-Jun became phosphorylated in pfGPI-stimulated macrophages. In contrast, pfGPI-induced phosphorylation of JNK, ERK1/2, and c-Jun was reduced in Cd36(-/-) macrophages and Cd36(-/-) macrophages secreted significantly less TNF-alpha in response to pfGPI than their wild-type counterparts. In addition, we demonstrate a role for CD36 in innate immune response to malaria in vivo. Compared with wild-type mice, Cd36(-/-) mice experienced more severe and fatal malaria when challenged with Plasmodium chabaudi chabaudi AS. Cd36(-/-) mice displayed a combined defect in cytokine induction and parasite clearance with a dysregulated cytokine response to infection, earlier peak parasitemias, higher parasite densities, and higher mortality rates than wild-type mice. These results provide direct evidence that pfGPI induces TNF-alpha secretion in a CD36-dependent manner and support a role for CD36 in modulating host cytokine response and innate control of acute blood-stage malaria infection in vivo.


Assuntos
Antígenos CD36/genética , Glicosilfosfatidilinositóis/imunologia , Imunidade Inata , Macrófagos/imunologia , Malária Falciparum/imunologia , Plasmodium falciparum/imunologia , Doença Aguda , Animais , Antígenos CD36/imunologia , Modelos Animais de Doenças , Humanos , Imunidade Inata/genética , Mediadores da Inflamação/imunologia , Sistema de Sinalização das MAP Quinases/genética , Sistema de Sinalização das MAP Quinases/imunologia , Malária Falciparum/genética , Camundongos , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/imunologia , Parasitemia/genética , Parasitemia/imunologia , Plasmodium chabaudi/imunologia , Fator de Necrose Tumoral alfa
14.
J Immunol ; 177(9): 6344-52, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17056565

RESUMO

Host inflammatory responses to Plasmodium falciparum GPI (pfGPI) anchors are believed to play an important role in the pathophysiology of severe malaria. However, relatively little is known about the signal transduction pathways involved in pfGPI-stimulated inflammatory response and its potential contribution to severe malaria syndromes. In this study, we investigated the role of MAPK activation in pfGPI-induced cytokine secretion and examined the role of selected MAPKs in a model of cerebral malaria in vivo. We demonstrate that ERK1/2, JNK, p38, c-Jun, and activating transcription factor-2 became phosphorylated in pfGPI-stimulated macrophages. A JNK inhibitor (1,9-pyrazoloanthrone) inhibited pfGPI-induced phosphorylation of JNK, c-Jun, and activating transcription factor-2 and significantly decreased pfGPI-induced TNF-alpha secretion. pfGPI-stimulated JNK and c-Jun phosphorylation was absent in Jnk2(-/-) macrophages but unchanged in Jnk1(-/-) and Jnk3(-/-) macrophages compared with wild-type macrophages. Jnk2(-/-) macrophages secreted significantly less TNF-alpha in response to pfGPI than macrophages from Jnk1(-/-), Jnk3(-/-), and wild-type counterparts. Furthermore, we demonstrate a role for JNK2 in mediating inflammatory responses and severe malaria in vivo. In contrast to wild-type or Jnk1(-/-) mice, Jnk2(-/-) mice had lower levels of TNF-alpha in vivo and exhibited significantly higher survival rates when challenged with Plasmodium berghei ANKA. These results provide direct evidence that pfGPI induces TNF-alpha secretion through activation of MAPK pathways, including JNK2. These results suggest that JNK2 is a potential target for therapeutic interventions in severe malaria.


Assuntos
Glicosilfosfatidilinositóis/imunologia , Malária Cerebral/prevenção & controle , Malária Falciparum/prevenção & controle , Proteína Quinase 9 Ativada por Mitógeno/fisiologia , Plasmodium falciparum/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Ativação Enzimática , Glicosilfosfatidilinositóis/farmacologia , Malária Cerebral/enzimologia , Malária Cerebral/imunologia , Malária Falciparum/enzimologia , Malária Falciparum/imunologia , Camundongos , Camundongos Mutantes , Proteína Quinase 9 Ativada por Mitógeno/efeitos dos fármacos , Proteína Quinase 9 Ativada por Mitógeno/genética , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/efeitos dos fármacos , Proteínas Quinases/metabolismo
15.
Surgery ; 132(2): 360-4, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12219035

RESUMO

BACKGROUND: Patients sustaining major trauma are at risk of developing organ dysfunction. We have previously shown that resuscitated hemorrhagic shock primes for increased lung injury in response to lippolysaccharide (LPS), in part by preventing upregulation of the counterinflammatory cytokine IL-10. Because the mitogen-activated protein kinase (MAPK) family is known to participate in LPS signaling, we hypothesized that altered upstream signaling through these kinases might contribute to impaired LPS-simulated IL-10 release after shock and resuscitation. METHODS: Rats were bled to a mean arterial pressure of 40 mm Hg and maintained for 1 hour, then resuscitated. Alveolar macrophages were retrieved at the end of resuscitation and exposed to LPS (0.5 microg/mL). Western blotting for p38, extracellular-regulated protein kinase, and c-Jun NH2-terminal kinase was performed on whole cell lysates. In some studies, the alveolar macrophages were preincubated with the p38 inhibitor or the extracellular-regulated protein kinase inhibitor before LPS stimulation. IL-10 levels were measured by enzyme-linked immunosorbent assay. RESULTS: LPS caused an early activation in all members of the MAPK family, whereas antecedent shock both delayed and attenuated the LPS induction. To discern whether this reduction in LPS-stimulated MAPK activation after shock might contribute to reduced IL-10, specific inhibitors were used. Inhibition of p38 MAPK completely inhibited LPS-induced IL-10 production, whereas blockade of extracellular-regulated protein kinase pathway had no effect. CONCLUSIONS: Shock resuscitation impairs LPS-induced activation of the members of the MAPK family. For the critical counterinflammatory cytokine IL-10, inhibition of p38 activation appears to contribute to the reduced levels of this cytokine in response to LPS. This study provides in vitro evidence for altered signaling through p38 MAPK, as a mechanism leading to failed upregulation of a counterinflammatory cytokine, and thus the propagation of an unrestrained proinflammatory state. Restoration of normal signaling may represent an effective strategy to reverse this effect.


Assuntos
Proteínas Quinases Ativadas por Mitógeno/metabolismo , Choque Hemorrágico/metabolismo , Animais , Ativação Enzimática/efeitos dos fármacos , Interleucina-10/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/enzimologia , Masculino , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/metabolismo , Regulação para Cima/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...