Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Malar J ; 20(1): 9, 2021 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-33407502

RESUMEN

BACKGROUND: Plasmodium falciparum causes the majority of malaria cases worldwide and children in sub-Saharan Africa are the most vulnerable group affected. Non-sterile clinical immunity that protects from symptoms develops slowly and is relatively short-lived. Moreover, current malaria vaccine candidates fail to induce durable high-level protection in endemic settings, possibly due to the immunomodulatory effects of the malaria parasite itself. Because dendritic cells play a crucial role in initiating immune responses, the aim of this study was to better understand the impact of cumulative malaria exposure as well as concurrent P. falciparum infection on dendritic cell phenotype and function. METHODS: In this cross-sectional study, the phenotype and function of dendritic cells freshly isolated from peripheral blood samples of Malian adults with a lifelong history of malaria exposure who were either uninfected (n = 27) or asymptomatically infected with P. falciparum (n = 8) was assessed. Additionally, plasma cytokine and chemokine levels were measured in these adults and in Malian children (n = 19) with acute symptomatic malaria. RESULTS: With the exception of lower plasmacytoid dendritic cell frequencies in asymptomatically infected Malian adults, peripheral blood dendritic cell subset frequencies and HLA-DR surface expression did not differ by infection status. Peripheral blood myeloid dendritic cells of uninfected Malian adults responded to in vitro stimulation with P. falciparum blood-stage parasites by up-regulating the costimulatory molecules HLA-DR, CD80, CD86 and CD40 and secreting IL-10, CXCL9 and CXCL10. In contrast, myeloid dendritic cells of asymptomatically infected Malian adults exhibited no significant responses above the uninfected red blood cell control. IL-10 and CXCL9 plasma levels were elevated in both asymptomatic adults and children with acute malaria. CONCLUSIONS: The findings of this study indicate that myeloid dendritic cells of uninfected adults with a lifelong history of malaria exposure are able to up-regulate co-stimulatory molecules and produce cytokines. Whether mDCs of malaria-exposed individuals are efficient antigen-presenting cells capable of mounting an appropriate immune response remains to be determined. The data also highlights IL-10 and CXCL9 as important factors in both asymptomatic and acute malaria and add to the understanding of asymptomatic P. falciparum infections in malaria-endemic areas.


Asunto(s)
Citocinas/sangre , Células Dendríticas/parasitología , Malaria Falciparum/sangre , Adulto , Infecciones Asintomáticas , Quimiocinas/sangre , Niño , Preescolar , Estudios Transversales , Eritrocitos/parasitología , Femenino , Humanos , Malaria/sangre , Masculino , Malí , Persona de Mediana Edad , Fenotipo , Plasmodium falciparum/fisiología
2.
Proc Natl Acad Sci U S A ; 114(49): E10568-E10577, 2017 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-29162686

RESUMEN

Dendritic cells (DCs) are activated by pathogens to initiate and shape immune responses. We found that the activation of DCs by Plasmodium falciparum, the main causative agent of human malaria, induces a highly unusual phenotype by which DCs up-regulate costimulatory molecules and secretion of chemokines, but not of cytokines typical of inflammatory responses (IL-1ß, IL-6, IL-10, TNF). Similar results were obtained with DCs obtained from malaria-naïve US donors and malaria-experienced donors from Mali. Contact-dependent cross-talk between the main DC subsets, plasmacytoid and myeloid DCs (mDCs) was necessary for increased chemokine and IFN-α secretion in response to the parasite. Despite the absence of inflammatory cytokine secretion, mDCs incubated with P. falciparum-infected erythrocytes activated antigen-specific naïve CD4+ T cells to proliferate and secrete Th1-like cytokines. This unexpected response of human mDCs to P. falciparum exhibited a transcriptional program distinct from a classical LPS response, pointing to unique P. falciparum-induced activation pathways that may explain the uncharacteristic immune response to malaria.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Eritrocitos/parasitología , Interacciones Huésped-Parásitos , Activación de Linfocitos , Plasmodium falciparum/metabolismo , Antígenos CD/genética , Antígenos CD/inmunología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/efectos de los fármacos , Quimiocina CCL2/genética , Quimiocina CCL2/inmunología , Quimiocina CCL5/genética , Quimiocina CCL5/inmunología , Quimiocina CXCL10/genética , Quimiocina CXCL10/inmunología , Quimiocina CXCL9/genética , Quimiocina CXCL9/inmunología , Técnicas de Cocultivo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/parasitología , Regulación de la Expresión Génica , Antígenos HLA-DR/genética , Antígenos HLA-DR/inmunología , Humanos , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Lipopolisacáridos/farmacología , Malaria Falciparum/genética , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Malí , Plasmodium falciparum/crecimiento & desarrollo , Transducción de Señal , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
3.
Immunohorizons ; 3(11): 511-518, 2019 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-31690559

RESUMEN

Malaria is a highly inflammatory disease caused by the protozoan parasite Plasmodium During the blood stage of infection, patients exhibit fever with high levels of inflammatory cytokines in their blood. However, when cells of the immune system are incubated with the parasite in vitro, their cytokine response is low. In particular, human primary dendritic cells (DCs) respond to Plasmodium falciparum-infected erythrocytes by upregulating maturation markers and chemokines but lack a substantial cytokine response. Because oxidative stress is a trigger of inflammatory cytokines in malaria and synergizes with P. falciparum to induce IL-1ß secretion by macrophages, we assessed whether oxidative stress has an impact on DC maturation and function in response to P. falciparum Using xanthine oxidase, a reactive oxygen species- (ROS) producing enzyme that is increased during malaria, we observed that exposure to extracellular ROS potentiated DC maturation in response to the parasite. Xanthine oxidase-derived ROS increased parasite-induced cytokine secretion and CD80 surface expression in DCs. This enhanced maturation phenotype boosted the DCs' ability to prime autologous naive CD4+ T cells, resulting in higher T cell proliferation in vitro. Xanthine oxidase-derived ROS did not have an effect on the cytokines produced by primed T cells. We propose that oxidative stress during malaria contributes to the inflammatory response by enhancing the magnitude of DC and CD4+ T cell responses without changing the quality.


Asunto(s)
Citocinas/inmunología , Células Dendríticas/inmunología , Eritrocitos/parasitología , Malaria Falciparum/inmunología , Estrés Oxidativo , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/parasitología , Humanos , Activación de Linfocitos , Plasmodium falciparum
4.
EMBO Mol Med ; 11(8): e9903, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31265218

RESUMEN

Malaria is a highly inflammatory disease caused by Plasmodium infection of host erythrocytes. However, the parasite does not induce inflammatory cytokine responses in macrophages in vitro and the source of inflammation in patients remains unclear. Here, we identify oxidative stress, which is common in malaria, as an effective trigger of the inflammatory activation of macrophages. We observed that extracellular reactive oxygen species (ROS) produced by xanthine oxidase (XO), an enzyme upregulated during malaria, induce a strong inflammatory cytokine response in primary human monocyte-derived macrophages. In malaria patients, elevated plasma XO activity correlates with high levels of inflammatory cytokines and with the development of cerebral malaria. We found that incubation of macrophages with plasma from these patients can induce a XO-dependent inflammatory cytokine response, identifying a host factor as a trigger for inflammation in malaria. XO-produced ROS also increase the synthesis of pro-IL-1ß, while the parasite activates caspase-1, providing the two necessary signals for the activation of the NLRP3 inflammasome. We propose that XO-produced ROS are a key factor for the trigger of inflammation during malaria.


Asunto(s)
Inflamación/enzimología , Macrófagos/enzimología , Malaria Cerebral/enzimología , Malaria Falciparum/enzimología , Estrés Oxidativo , Plasmodium falciparum/patogenicidad , Especies Reactivas de Oxígeno/metabolismo , Xantina Oxidasa/metabolismo , Caspasa 1/metabolismo , Células Cultivadas , Citocinas/metabolismo , Interacciones Huésped-Patógeno , Humanos , Inflamación/sangre , Inflamación/parasitología , Mediadores de Inflamación/metabolismo , Activación de Macrófagos , Macrófagos/parasitología , Malaria Cerebral/sangre , Malaria Cerebral/parasitología , Malaria Falciparum/sangre , Malaria Falciparum/parasitología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Transducción de Señal
5.
Cell Host Microbe ; 19(2): 194-203, 2016 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-26867178

RESUMEN

Plasmodium species, the parasitic agents of malaria, invade erythrocytes to reproduce, resulting in erythrocyte loss. However, a greater loss is caused by the elimination of uninfected erythrocytes, sometimes long after infection has been cleared. Using a mouse model, we found that Plasmodium infection induces the generation of anti-self antibodies that bind to the surface of uninfected erythrocytes from infected, but not uninfected, mice. These antibodies recognize phosphatidylserine, which is exposed on the surface of a fraction of uninfected erythrocytes during malaria. We find that phosphatidylserine-exposing erythrocytes are reticulocytes expressing high levels of CD47, a "do-not-eat-me" signal, but the binding of anti-phosphatidylserine antibodies mediates their phagocytosis, contributing to anemia. In human patients with late postmalarial anemia, we found a strong inverse correlation between the levels of anti-phosphatidylserine antibodies and plasma hemoglobin, suggesting a similar role in humans. Inhibition of this pathway may be exploited for treating malarial anemia.


Asunto(s)
Anemia/etiología , Anticuerpos Antiprotozoarios/inmunología , Eritrocitos/inmunología , Malaria Falciparum/complicaciones , Fosfatidilserinas/inmunología , Plasmodium falciparum/fisiología , Animales , Eritrocitos/parasitología , Femenino , Humanos , Malaria Falciparum/inmunología , Masculino , Ratones , Fagocitosis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA