Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 170
Filtrar
1.
Mol Biochem Parasitol ; 253: 111540, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36509228

RESUMEN

Recently, there is a paucity of studies focus on the characteristics of myeloid cells which expressed γδTCR. The aim of this study was to observe the properties of γδTCR-expressing myeloid cells in the spleen of C57BL/6 mice infected by P. yoelii nigeriensis NSM. Haematoxylin-eosin (HE) staining was used to observe pathological changes in the spleens from infected mice. The differentially expressed genes (DEGs) between the infection and control groups were analyzed by RNA sequencing (RNA -seq). Flow cytometry (FCM) was used to evaluate the frequency of γδTCR+ cells and the characteristics of γδTCR+ cells in P. yoelii nigeriensis NSM-infected mice. Obvious infiltration of inflammatory were observed in the spleens from infected C57BL/6 mouse. The proportions of γδTCR+ cells and CD11b+ γδTCR+ cells from infected group were higher than that from normal group. CD11b+ γδTCR+ cells expressed high levels of activated-mediated genes and inflammatory-mediated genes. The heterogeneous pathway activities among CD11b+ γδTCR+ cells from normal and infected group were characterized. The oxidative phosphorylation, respiratory electron transport chain and leukocyte activation involved in immune response pathways were up-regulated, while the alpha-beta T cell activation and myeloid leukocyte migration pathways were down-regulated in infected mice. Importantly, Ly6c2 was higher expressed in CD11b+ γδTCR+ cells than Ly6g. Consistent with it, flow cytometry results revealed that a subset of Ly6C+ cells was higher than Ly6G+ cells in the spleen. Taken together, our data suggest the existence of a population of γδTCR-expressing myeloid cells and they might be multifunctional cells, which play a role in couse of Plasmodium infection.


Asunto(s)
Malaria , Células Mieloides , Plasmodium yoelii , Receptores de Antígenos de Linfocitos T gamma-delta , Animales , Ratones , Citometría de Flujo , Ratones Endogámicos C57BL , Células Mieloides/metabolismo , Plasmodium yoelii/fisiología
2.
Parasitol Int ; 88: 102542, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35063657

RESUMEN

Increased levels of several human ubiquitin ligases, including ring finger protein 123 (RNF123), in red blood cells with Plasmodium falciparum infection, have been reported. RNF123 is an E3 ubiquitin ligase that is highly expressed in erythroid cells. However, the function of the RNF123 gene and the relationship between the RNF123 gene and malarial parasite has not been clarified in vivo. In this study, we generated RNF123-deficient mice using the CRISPR/Cas9 system, and analyzed malaria susceptibility and erythrocyte morphology. The levels of parasitemia 5 days post-infection and mortality 21 days post-infection with the lethal type of rodent malaria (Plasmodium yoelii 17XL) in RNF123-deficient mice was significantly lower than that in wild-type mice. In contrast, red blood cell morphology in RNF123-deficient mice was almost normal. These results suggest that erythrocytic RNF123 plays a role in susceptibility to rodent malaria infection, but does not play a role in erythrocyte morphology.


Asunto(s)
Malaria , Plasmodium yoelii , Animales , Malaria/parasitología , Ratones , Ratones Endogámicos BALB C , Parasitemia/parasitología , Plasmodium yoelii/fisiología , Roedores , Ubiquitina-Proteína Ligasas/genética
3.
Malar J ; 20(1): 297, 2021 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-34215257

RESUMEN

BACKGROUND: Recent genome wide analysis studies have identified a strong association between single nucleotide variations within the human ATP2B4 gene and susceptibility to severe malaria. The ATP2B4 gene encodes the plasma membrane calcium ATPase 4 (PMCA4), which is responsible for controlling the physiological level of intracellular calcium in many cell types, including red blood cells (RBCs). It is, therefore, postulated that genetic differences in the activity or expression level of PMCA4 alters intracellular Ca2+ levels and affects RBC hydration, modulating the invasion and growth of the Plasmodium parasite within its target host cell. METHODS: In this study the course of three different Plasmodium spp. infections were examined in mice with systemic knockout of Pmca4 expression. RESULTS: Ablation of PMCA4 reduced the size of RBCs and their haemoglobin content but did not affect RBC maturation and reticulocyte count. Surprisingly, knockout of PMCA4 did not significantly alter peripheral parasite burdens or the dynamics of blood stage Plasmodium chabaudi infection or reticulocyte-restricted Plasmodium yoelii infection. Interestingly, although ablation of PMCA4 did not affect peripheral parasite levels during Plasmodium berghei infection, it did promote slight protection against experimental cerebral malaria, associated with a minor reduction in antigen-experienced T cell accumulation in the brain. CONCLUSIONS: The finding suggests that PMCA4 may play a minor role in the development of severe malarial complications, but that this appears independent of direct effects on parasite invasion, growth or survival within RBCs.


Asunto(s)
Resistencia a la Enfermedad/genética , Malaria/genética , ATPasas Transportadoras de Calcio de la Membrana Plasmática/genética , Plasmodium/fisiología , Animales , Membrana Celular , Malaria/sangre , Malaria/parasitología , Malaria Cerebral/genética , Malaria Cerebral/parasitología , Ratones , Ratones Noqueados , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , Plasmodium berghei/fisiología , Plasmodium chabaudi/fisiología , Plasmodium yoelii/fisiología
4.
Malar J ; 20(1): 89, 2021 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-33588839

RESUMEN

BACKGROUND: Malaria has high morbidity and mortality rates in some parts of tropical and subtropical countries. Besides respiratory and metabolic function, lung plays a role in immune system. γδT cells have multiple functions in producing cytokines and chemokines, regulating the immune response by interacting with other cells. It remains unclear about the role of γδT cells in the lung of mice infected by malaria parasites. METHODS: Flow cytometry (FCM) was used to evaluate the frequency of γδT cells and the effects of γδT cells on the phenotype and function of B and T cells in Plasmodium yoelii-infected wild-type (WT) or γδTCR knockout (γδT KO) mice. Haematoxylin-eosin (HE) staining was used to observe the pathological changes in the lungs. RESULTS: The percentage and absolute number of γδT cells in the lung increased after Plasmodium infection (p < 0.01). More γδT cells were expressing CD80, CD11b, or PD-1 post-infection (p < 0.05), while less γδT cells were expressing CD34, CD62L, and CD127 post-infection (p < 0.05). The percentages of IL-4+, IL-5+, IL-6+, IL-21+, IL-1α+, and IL-17+ γδT cells were increased (p < 0.05), but the percentage of IFN-γ-expressing γδT cells decreased (p < 0.05) post-infection. The pathological changes in the lungs of the infected γδT KO mice were not obvious compared with the infected WT mice. The proportion of CD3+ cells and absolute numbers of CD3+ cells, CD3+ CD4+ cells, CD3+ CD8+ cells decreased in γδT KO infected mice (p < 0.05). γδT KO infected mice exhibited no significant difference in the surface molecular expression of T cells compared with the WT infected mice (p > 0.05). While, the percentage of IFN-γ-expressing CD3+ and CD3+ CD8+ cells increased in γδT KO infected mice (p < 0.05). There was no significant difference in the absolute numbers of the total, CD69+, ICOS+, and CD80+ B cells between the WT infected and γδT KO infected mice (p > 0.05). CONCLUSIONS: The content, phenotype, and function of γδT cells in the lung of C57BL/6 mice were changed after Plasmodium infection. γδT cells contribute to T cell immune response in the progress of Plasmodium infection.


Asunto(s)
Linfocitos Intraepiteliales/inmunología , Pulmón/inmunología , Malaria/inmunología , Plasmodium yoelii/fisiología , Animales , Linfocitos B/inmunología , Femenino , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Linfocitos T/inmunología
5.
J Cell Mol Med ; 24(19): 11535-11545, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32885594

RESUMEN

Early type I interferon is essential for antagonizing against malaria infection, which remains a significant global infectious disease. After Plasmodium yoelii YM infection, the activation of MAVS-, STING- and inflammasome-IRF3-mediated pathway could trigger the Socs1 expression to inhibit the TLR7-MyD88-IRF7-induced type I interferon production. However, the dynamic regulatory mechanisms of type I interferon response to YM infection and delicate cross-regulation of these signalling are far from clear. In current study, we established a mathematical model to systematically demonstrate that the MAVS-, STING- and inflammasome-mediated signalling pathways play distinct roles in regulating type I interferon response after YM infection; and the YM dose could significantly affect the difference of resistance to YM infection among MAVS, STING and inflammasome deficiency. Collectively, our study systematically elucidated the precise regulatory mechanisms of type I interferon signalling after YM infection and advanced the research on therapy of plasmodium infection by incorporating multiple signalling pathways at diverse time.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Inflamasomas/metabolismo , Interferón Tipo I/metabolismo , Proteínas de la Membrana/metabolismo , Modelos Biológicos , Factor 88 de Diferenciación Mieloide/metabolismo , Animales , Resistencia a la Enfermedad , Femenino , Malaria/metabolismo , Malaria/parasitología , Malaria/patología , Ratones Endogámicos C57BL , Plasmodium yoelii/fisiología , Transducción de Señal , Proteína 1 Supresora de la Señalización de Citocinas/metabolismo , Factores de Tiempo
6.
J Immunol ; 205(6): 1608-1619, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32817333

RESUMEN

CD4+ T cell functional inhibition (exhaustion) is a hallmark of malaria and correlates with impaired parasite control and infection chronicity. However, the mechanisms of CD4+ T cell exhaustion are still poorly understood. In this study, we show that Ag-experienced (Ag-exp) CD4+ T cell exhaustion during Plasmodium yoelii nonlethal infection occurs alongside the reduction in mammalian target of rapamycin (mTOR) activity and restriction in CD4+ T cell glycolytic capacity. We demonstrate that the loss of glycolytic metabolism and mTOR activity within the exhausted Ag-expCD4+ T cell population during infection coincides with reduction in T-bet expression. T-bet was found to directly bind to and control the transcription of various mTOR and metabolism-related genes within effector CD4+ T cells. Consistent with this, Ag-expTh1 cells exhibited significantly higher and sustained mTOR activity than effector T-bet- (non-Th1) Ag-expT cells throughout the course of malaria. We identified mTOR to be redundant for sustaining T-bet expression in activated Th1 cells, whereas mTOR was necessary but not sufficient for maintaining IFN-γ production by Th1 cells. Immunotherapy targeting PD-1, CTLA-4, and IL-27 blocked CD4+ T cell exhaustion during malaria infection and was associated with elevated T-bet expression and a concomitant increased CD4+ T cell glycolytic metabolism. Collectively, our data suggest that mTOR activity is linked to T-bet in Ag-expCD4+ T cells but that reduction in mTOR activity may not directly underpin Ag-expTh1 cell loss and exhaustion during malaria infection. These data have implications for therapeutic reactivation of exhausted CD4+ T cells during malaria infection and other chronic conditions.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Malaria/inmunología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Plasmodium yoelii/fisiología , Proteínas de Dominio T Box/metabolismo , Células TH1/inmunología , Animales , Senescencia Celular , Regulación de la Expresión Génica , Glucólisis , Humanos , Tolerancia Inmunológica , Memoria Inmunológica , Interferón gamma/metabolismo , Interleucina-27/metabolismo , Activación de Linfocitos , Malaria/terapia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Dominio T Box/genética
7.
Proc Natl Acad Sci U S A ; 117(28): 16567-16578, 2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32606244

RESUMEN

Malaria infection induces complex and diverse immune responses. To elucidate the mechanisms underlying host-parasite interaction, we performed a genetic screen during early (24 h) Plasmodium yoelii infection in mice and identified a large number of interacting host and parasite genes/loci after transspecies expression quantitative trait locus (Ts-eQTL) analysis. We next investigated a host E3 ubiquitin ligase gene (March1) that was clustered with interferon (IFN)-stimulated genes (ISGs) based on the similarity of the genome-wide pattern of logarithm of the odds (LOD) scores (GPLS). March1 inhibits MAVS/STING/TRIF-induced type I IFN (IFN-I) signaling in vitro and in vivo. However, in malaria-infected hosts, deficiency of March1 reduces IFN-I production by activating inhibitors such as SOCS1, USP18, and TRIM24 and by altering immune cell populations. March1 deficiency increases CD86+DC (dendritic cell) populations and levels of IFN-γ and interleukin 10 (IL-10) at day 4 post infection, leading to improved host survival. T cell depletion reduces IFN-γ level and reverse the protective effects of March1 deficiency, which can also be achieved by antibody neutralization of IFN-γ. This study reveals functions of MARCH1 (membrane-associated ring-CH-type finger 1) in innate immune responses and provides potential avenues for activating antimalaria immunity and enhancing vaccine efficacy.


Asunto(s)
Malaria/inmunología , Plasmodium yoelii/fisiología , Linfocitos T/inmunología , Ubiquitina-Proteína Ligasas/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Interacciones Huésped-Parásitos , Humanos , Inmunidad Innata , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-10/genética , Interleucina-10/inmunología , Malaria/enzimología , Malaria/genética , Malaria/parasitología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plasmodium yoelii/inmunología , Ubiquitina-Proteína Ligasas/genética
8.
BMC Biol ; 18(1): 83, 2020 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-32620114

RESUMEN

BACKGROUND: Experimental reproducibility in mouse models is impacted by both genetics and environment. The generation of reproducible data is critical for the biomedical enterprise and has become a major concern for the scientific community and funding agencies alike. Among the factors that impact reproducibility in experimental mouse models is the variable composition of the microbiota in mice supplied by different commercial vendors. Less attention has been paid to how the microbiota of mice supplied by a particular vendor might change over time. RESULTS: In the course of conducting a series of experiments in a mouse model of malaria, we observed a profound and lasting change in the severity of malaria in mice infected with Plasmodium yoelii; while for several years mice obtained from a specific production suite of a specific commercial vendor were able to clear the parasites effectively in a relatively short time, mice subsequently shipped from the same unit suffered much more severe disease. Gut microbiota analysis of frozen cecal samples identified a distinct and lasting shift in bacteria populations that coincided with the altered response of the later shipments of mice to infection with malaria parasites. Germ-free mice colonized with cecal microbiota from mice within the same production suite before and after this change followed by Plasmodium infection provided a direct demonstration that the change in gut microbiota profoundly impacted the severity of malaria. Moreover, spatial changes in gut microbiota composition were also shown to alter the acute bacterial burden following Salmonella infection, and tumor burden in a lung tumorigenesis model. CONCLUSION: These changes in gut bacteria may have impacted the experimental reproducibility of diverse research groups and highlight the need for both laboratory animal providers and researchers to collaborate in determining the methods and criteria needed to stabilize the gut microbiota of animal breeding colonies and research cohorts, and to develop a microbiota solution to increase experimental rigor and reproducibility.


Asunto(s)
Modelos Animales de Enfermedad , Microbioma Gastrointestinal , Malaria/fisiopatología , Plasmodium yoelii/fisiología , Animales , Femenino , Ratones , Ratones Endogámicos C57BL , Análisis Espacio-Temporal
9.
Proc Natl Acad Sci U S A ; 117(32): 19465-19474, 2020 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-32709745

RESUMEN

Infection by malaria parasites triggers dynamic immune responses leading to diverse symptoms and pathologies; however, the molecular mechanisms responsible for these reactions are largely unknown. We performed Trans-species Expression Quantitative Trait Locus analysis to identify a large number of host genes that respond to malaria parasite infections. Here we functionally characterize one of the host genes called receptor transporter protein 4 (RTP4) in responses to malaria parasite and virus infections. RTP4 is induced by type I IFN (IFN-I) and binds to the TANK-binding kinase (TBK1) complex where it negatively regulates TBK1 signaling by interfering with expression and phosphorylation of both TBK1 and IFN regulatory factor 3. Rtp4-/- mice were generated and infected with malaria parasite Plasmodiun berghei ANKA. Significantly higher levels of IFN-I response in microglia, lower parasitemia, fewer neurologic symptoms, and better survival rates were observed in Rtp4-/- than in wild-type mice. Similarly, RTP4 deficiency significantly reduced West Nile virus titers in the brain, but not in the heart and the spleen, of infected mice, suggesting a specific role for RTP4 in brain infection and pathology. This study reveals functions of RTP4 in IFN-I response and a potential target for therapy in diseases with neuropathology.


Asunto(s)
Encéfalo/patología , Interferón Tipo I/metabolismo , Malaria Cerebral/patología , Chaperonas Moleculares/metabolismo , Animales , Encéfalo/parasitología , Encéfalo/virología , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Factor 3 Regulador del Interferón , Malaria Cerebral/metabolismo , Malaria Cerebral/parasitología , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/metabolismo , Chaperonas Moleculares/genética , Fosforilación , Plasmodium berghei/fisiología , Plasmodium yoelii/fisiología , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Fiebre del Nilo Occidental/metabolismo , Fiebre del Nilo Occidental/patología , Fiebre del Nilo Occidental/virología , Virus del Nilo Occidental/fisiología
10.
PLoS One ; 15(5): e0232234, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32407410

RESUMEN

Only a small fraction of the antigens expressed by malaria parasites have been evaluated as vaccine candidates. A successful malaria subunit vaccine will likely require multiple antigenic targets to achieve broad protection with high protective efficacy. Here we describe protective efficacy of a novel antigen, Plasmodium yoelii (Py) E140 (PyE140), evaluated against P. yoelii challenge of mice. Vaccines targeting PyE140 reproducibly induced up to 100% sterile protection in both inbred and outbred murine challenge models. Although PyE140 immunization induced high frequency and multifunctional CD8+ T cell responses, as well as CD4+ T cell responses, protection was mediated by PyE140 antibodies acting against blood stage parasites. Protection in mice was long-lasting with up to 100% sterile protection at twelve weeks post-immunization and durable high titer anti-PyE140 antibodies. The E140 antigen is expressed in all Plasmodium species, is highly conserved in both P. falciparum lab-adapted strains and endemic circulating parasites, and is thus a promising lead vaccine candidate for future evaluation against human malaria parasite species.


Asunto(s)
Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/inmunología , Inmunización , Malaria/prevención & control , Plasmodium yoelii/fisiología , Animales , Antígenos de Protozoos/genética , Reacciones Cruzadas , Femenino , Regulación de la Expresión Génica , Ratones , Plasmodium yoelii/genética , Plasmodium yoelii/inmunología
11.
PLoS Negl Trop Dis ; 14(2): e0008059, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32032359

RESUMEN

During a blood meal, female Anopheles mosquitoes are potentially exposed to diverse microbes in addition to the malaria parasite, Plasmodium. Human and animal African trypanosomiases are frequently co-endemic with malaria in Africa. It is not known whether exposure of Anopheles to trypanosomes influences their fitness or ability to transmit Plasmodium. Using cell and molecular biology approaches, we found that Trypanosoma brucei brucei parasites survive for at least 48h after infectious blood meal in the midgut of the major malaria vector, Anopheles coluzzii before being cleared. This transient survival of trypanosomes in the midgut is correlated with a dysbiosis, an alteration in the abundance of the enteric bacterial flora in Anopheles coluzzii. Using a developmental biology approach, we found that the presence of live trypanosomes in mosquito midguts also reduces their reproductive fitness, as it impairs the viability of laid eggs by affecting their hatching. Furthermore, we found that Anopheles exposure to trypanosomes enhances their vector competence for Plasmodium, as it increases their infection prevalence. A transcriptomic analysis revealed that expression of only two Anopheles immune genes are modulated during trypanosome exposure and that the increased susceptibility to Plasmodium was microbiome-dependent, while the reproductive fitness cost was dependent only on the presence of live trypanosomes but was microbiome independent. Taken together, these results demonstrate multiple effects upon Anopheles vector competence for Plasmodium caused by eukaryotic microbes interacting with the host and its microbiome, which may in turn have implications for malaria control strategies in co-endemic areas.


Asunto(s)
Anopheles/parasitología , Malaria/parasitología , Plasmodium yoelii/fisiología , Trypanosoma/fisiología , Animales , Coinfección , Interacciones Huésped-Parásitos , Ratones , Reacción en Cadena de la Polimerasa , Reproducción
12.
mBio ; 11(1)2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31911494

RESUMEN

Erythrocyte-binding-like (EBL) proteins are known to play an important role in malaria parasite invasion of red blood cells (RBCs); however, any roles of EBL proteins in regulating host immune responses remain unknown. Here, we show that Plasmodium yoelii EBL (PyEBL) can shape disease severity by modulating the surface structure of infected RBCs (iRBCs) and host immune responses. We identified an amino acid substitution (a change of C to Y at position 741 [C741Y]) in the protein trafficking domain of PyEBL between isogenic P. yoelliinigeriensis strain N67 and N67C parasites that produce different disease phenotypes in C57BL/6 mice. Exchanges of the C741Y alleles altered parasite growth and host survival accordingly. The C741Y substitution also changed protein processing and trafficking in merozoites and in the cytoplasm of iRBCs, reduced PyEBL binding to band 3, increased phosphatidylserine (PS) surface exposure, and elevated the osmotic fragility of iRBCs, but it did not affect invasion of RBCs in vitro The modified iRBC surface triggered PS-CD36-mediated phagocytosis of iRBCs, host type I interferon (IFN-I) signaling, and T cell differentiation, leading to improved host survival. This study reveals a previously unknown role of PyEBL in regulating host-pathogen interaction and innate immune responses, which may be explored for developing disease control strategies.IMPORTANCE Malaria is a deadly parasitic disease that continues to afflict hundreds of millions of people every year. Infections with malaria parasites can be asymptomatic, with mild symptoms, or fatal, depending on a delicate balance of host immune responses. Malaria parasites enter host red blood cells (RBCs) through interactions between parasite ligands and host receptors, such as erythrocyte-binding-like (EBL) proteins and host Duffy antigen receptor for chemokines (DARC). Plasmodium yoelii EBL (PyEBL) is known to play a role in parasite invasion of RBCs. Here, we show that PyEBL also affects disease severity through modulation of host immune responses, particularly type I interferon (IFN-I) signaling. This discovery assigns a new function to PyEBL and provides a mechanism for developing disease control strategies.


Asunto(s)
Antígenos de Protozoos/inmunología , Eritrocitos/inmunología , Eritrocitos/parasitología , Malaria/inmunología , Malaria/parasitología , Proteínas de la Membrana/metabolismo , Plasmodium yoelii/fisiología , Proteínas Protozoarias/metabolismo , Alelos , Antígenos de Protozoos/metabolismo , Biomarcadores , Citocinas/metabolismo , Técnica del Anticuerpo Fluorescente , Interacciones Huésped-Parásitos , Inmunohistoquímica , Malaria/diagnóstico , Malaria/metabolismo , Proteínas de la Membrana/inmunología , Fragilidad Osmótica , Fagocitosis/inmunología , Proteínas Protozoarias/genética , Proteínas Protozoarias/inmunología , Índice de Severidad de la Enfermedad , Bazo/inmunología , Bazo/metabolismo , Bazo/patología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo
13.
Nat Commun ; 10(1): 3950, 2019 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-31477704

RESUMEN

Immunization with attenuated whole Plasmodium sporozoites constitutes a promising vaccination strategy. Compared to replication-deficient parasites, immunization with replication-competent parasites confers better protection and also induces a type I IFN (IFN-1) response, but whether this IFN-1 response has beneficial or adverse effects on vaccine-induced adaptive immunity is not known. Here, we show that IFN-1 signaling-deficient mice immunized with replication-competent sporozoites exhibit superior protection against infection. This correlates with superior CD8 T cell memory including reduced expression of the exhaustion markers PD-1 and LAG-3 on these cells and increased numbers of memory CD8 T cells in the liver. Moreover, the adoptive transfer of memory CD8 T cells from the livers of previously immunized IFN-1 signaling-deficient mice confers greater protection against liver stage parasites. However, the detrimental role of IFN-1 signaling is not CD8 T cell intrinsic. Together, our data demonstrate that liver stage-engendered IFN-1 signaling impairs hepatic CD8 T cell memory via a CD8 T cell-extrinsic mechanism.


Asunto(s)
Inmunidad Adaptativa/inmunología , Eritrocitos/inmunología , Inmunidad Innata/inmunología , Malaria/inmunología , Plasmodium yoelii/inmunología , Esporozoítos/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/parasitología , Eritrocitos/parasitología , Femenino , Inmunización , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Hígado/inmunología , Hígado/metabolismo , Hígado/parasitología , Malaria/parasitología , Malaria/prevención & control , Vacunas contra la Malaria/administración & dosificación , Vacunas contra la Malaria/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Plasmodium yoelii/fisiología , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología
14.
Int J Parasitol ; 49(9): 705-714, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31202685

RESUMEN

CRISPR/Cas9 is a powerful genome editing method that has greatly facilitated functional studies in many eukaryotic organisms including malaria parasites. Due to the lack of genes encoding enzymes necessary for the non-homologous end joining DNA repair pathway, genetic manipulation of malaria parasite genomes is generally accomplished through homologous recombination requiring the presence of DNA templates. Recently, an alternative double-strand break repair pathway, microhomology-mediated end joining, was found in the Plasmodium falciparum parasite. Taking advantage of the MMEJ pathway, we developed a MMEJ-based CRISPR/Cas9 (mCRISPR) strategy to efficiently generate multiple mutant parasites simultaneously in genes with repetitive sequences. As a proof of principle, we successfully produced various size mutants in the central repeat region of the Plasmodium yoelii circumsporozoite surface protein without the use of template DNA. Monitoring mixed parasite populations and individual parasites with different sizes of CSP-CRR showed that the CSP-CRR plays a role in the development of mosquito stages, with severe developmental defects in parasites with large deletions in the repeat region. However, the majority of the csp mutant parasite clones grew similarly to the wild type P. yoelii 17XL parasite in mice. This study develops a useful technique to efficiently generate mutant parasites with deletions or insertions, and shows that the CSP-CRR plays a role in parasite development in mosquito.


Asunto(s)
Proteína 9 Asociada a CRISPR/genética , Eliminación de Gen , Plasmodium yoelii/genética , Animales , Anopheles/parasitología , Proteína 9 Asociada a CRISPR/inmunología , Clonación Molecular , Culicidae/parasitología , Reparación del ADN , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Mosquitos Vectores/parasitología , Plásmidos , Plasmodium yoelii/crecimiento & desarrollo , Plasmodium yoelii/fisiología , Proteínas Protozoarias/genética
15.
Front Immunol ; 10: 1225, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31214184

RESUMEN

The enzyme acid sphingomyelinase (ASM) hydrolyzes sphingomyelin to ceramide and is thereby involved in several cellular processes such as differentiation, proliferation, and apoptosis in different cell types. However, the function of ASM in T cells is still not well characterized. Here, we used T cell-specific ASM overexpressing mice (t-ASM/CD4cre) to clarify the impact of cell-intrinsic ASM activity on T cell function in vitro and in vivo. We showed that t-ASM/CD4cre mice exhibit decreased frequencies of Foxp3+ T regulatory cells (Tregs) within the spleen. Enforced T cell-specific ASM expression resulted in less efficient induction of Tregs and promoted differentiation of CD4+CD25- naïve T cells into IFN-γ producing Th1 cells in vitro. Further analysis revealed that ASM-overexpressing T cells from t-ASM/CD4cre mice show elevated T cell receptor (TCR) signaling activity accompanied with increased proliferation upon stimulation in vitro. Plasmodium yoelii infection of t-ASM/CD4cre mice resulted in enhanced T cell activation and was associated with reduced parasitemia in comparison to infected control mice. Hence, our results provide evidence that ASM activity modulates T cell function in vitro and in vivo.


Asunto(s)
Malaria/inmunología , Plasmodium yoelii/fisiología , Esfingomielina Fosfodiesterasa/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Diferenciación Celular , Células Cultivadas , Factores de Transcripción Forkhead/metabolismo , Humanos , Activación de Linfocitos , Ratones , Parasitemia , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Regulación hacia Arriba
16.
PLoS One ; 14(3): e0214449, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30917184

RESUMEN

Malaria is a devastating disease resulting in significant morbidity and mortality, especially in the developing world. Previously, we showed that the gut microbiome modulates severity of malaria in mice, though the exact mechanism was unknown. One well-studied mechanism by which the intestinal microbiota exerts an effect on host health is by synthesis of short-chain fatty acids (SCFAs). SCFAs have pleiotropic effects on the host, including modulating the immune system and altering susceptibility to pathogens. The objective of the current work was to explore if gut microbiota-mediated resistance and susceptibility to malaria in mice is through differential production of SCFAs. Of the eight detected SCFAs, only propionic acid (C3) was different between two groups of resistant and two groups of susceptible mice, with higher levels in feces of susceptible mice compared to resistant mice. Nevertheless, subsequent analysis revealed no robust correlation between malaria severity and levels of fecal propionic acid. In spite of the broad effect of SCFAs on host physiology, including host immunity, this study shows that gut microbiota-mediated modulation of malaria severity in mice is independent of fecal SCFA levels. Additionally, our data indicates that intestinal SCFAs do not function as biomarkers for prediction of malaria disease severity.


Asunto(s)
Ácidos Grasos Volátiles/metabolismo , Microbioma Gastrointestinal , Mucosa Intestinal/metabolismo , Malaria/metabolismo , Malaria/microbiología , Animales , Ácidos Grasos Volátiles/química , Heces/química , Femenino , Ratones , Ratones Endogámicos C57BL , Plasmodium yoelii/fisiología
17.
Sci Rep ; 9(1): 3472, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30837607

RESUMEN

Experimental models of malaria have shown that infection with specific Plasmodium species in certain mouse strains can transiently modulate gut microbiota and cause intestinal shortening, indicating a disruption of gut homeostasis. Importantly, changes in gut homeostasis have not been characterized in the context of mild versus severe malaria. We show that severe Plasmodium infection in mice disrupts homeostasis along the gut-liver axis in multiple ways compared to mild infection. High parasite burden results in a larger influx of immune cells in the lamina propria and mice with high parasitemia display specific metabolomic profiles in the ceca and plasma during infection compared to mice with mild parasitemia. Liver damage was also more pronounced and longer lasting during severe infection, with concomitant changes in bile acids in the gut. Finally, severe Plasmodium infection changes the functional capacity of the microbiota, enhancing bacterial motility and amino acid metabolism in mice with high parasite burden compared to a mild infection. Taken together, Plasmodium infections have diverse effects on host gut homeostasis relative to the severity of infection that may contribute to enteric bacteremia that is associated with malaria.


Asunto(s)
Susceptibilidad a Enfermedades , Homeostasis , Intestinos , Hígado , Malaria/parasitología , Plasmodium yoelii/fisiología , Animales , Modelos Animales de Enfermedad , Microbioma Gastrointestinal , Hígado/metabolismo , Malaria/inmunología , Malaria/metabolismo , Metaboloma , Metabolómica/métodos , Ratones , Ratones Endogámicos C57BL , Membrana Mucosa/inervación , Membrana Mucosa/metabolismo , Membrana Mucosa/parasitología , Carga de Parásitos , Permeabilidad
18.
Int Immunopharmacol ; 70: 387-395, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30852294

RESUMEN

Vitamin C (ascorbate) is maintained at high levels in most immune cells and can affect many aspects of the immune response. Here, we evaluated the effect of vitamin C supplementation on the immune response to Plasmodium yoelii 17XL (P. yoelii 17XL) infection in BALB/c mice. Two orally administered doses (25 mg/kg/day and 250 mg/kg/day) of vitamin C significantly reduced levels of parasitemia during the early stages of P. yoelii 17XL infection. The numbers of activated Th1 cells and macrophages in the groups receiving vitamin C supplementation were both higher than those in the untreated group. Meanwhile, vitamin C administration reduced the levels of tumor necrosis factor α secreted by splenocytes. Vitamin C also regulated the protective anti-malarial immune response by increasing the number of plasmacytoid dendritic cells, as well as the expression of dendritic cell maturation markers, such as major histocompatibility complex class II and cluster of differentiation 86. In conclusion, the doses of vitamin C (25 mg/kg/day, 250 mg/kg/day) during the early stages of malaria infection may better enhance host protective immunity, but have no dose dependence.


Asunto(s)
Antimaláricos/uso terapéutico , Ácido Ascórbico/uso terapéutico , Células Dendríticas/inmunología , Malaria/terapia , Plasmodium yoelii/fisiología , Células TH1/inmunología , Animales , Diferenciación Celular , Células Cultivadas , Suplementos Dietéticos , Cálculo de Dosificación de Drogas , Femenino , Humanos , Inmunidad Celular , Activación de Linfocitos , Malaria/inmunología , Ratones , Ratones Endogámicos BALB C
19.
Vaccine ; 36(49): 7463-7471, 2018 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-30420038

RESUMEN

Malaria transmission-blocking vaccines aim to inhibit the development of malaria parasites in mosquitoes by inducing antibodies targeting surface proteins of sexual stage parasites. We have recently identified PyMiGS, a protein specifically expressed in the osmiophilic body of male gametocytes of Plasmodium yoelii (Py). PyMiGS is translocated to the surface of microgametes, and potent transmission-blocking activity was observed in mosquitoes fed on mice passively immunized with antibodies against PyMiGS. Here we demonstrate using a direct feeding assay that recombinant PyMiGS successfully induces anti-PyMiGS antibodies in mice and that the antibodies block parasite development in mosquitoes. We also show using the membrane-feeding assay that rabbit anti-PyMiGS antibody inhibits parasite development in mosquitoes in a dose-dependent manner without complement involvement. To investigate the mode of action of anti-PyMiGS antibodies against parasite development, we observed exflagellation after mixing Py gametocytes with activation medium containing anti-PyMiGS or anti-GST control antibodies. Whereas most microgametes were released from activated male gametocytes in the control group, a significantly reduced number of microgametes were released in the anti-PyMiGS group, with most of the microgametes left attached to the activated male gametocytes. Moreover, anti-PyMiGS antibodies shortened the duration of the active movement of microgametes after the onset of exflagellation. Taken together, these findings suggest that anti-PyMiGS antibodies bind to the microgamete surface immediately after exflagellation, thereby reducing microgamete motility and inhibiting microgamete release from the activated male gametocytes. These results strongly suggest that PyMiGS orthologues in Plasmodium falciparum and Plasmodium vivax can be promising TBV candidates.


Asunto(s)
Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/inmunología , Antígenos de Superficie/inmunología , Culicidae/parasitología , Malaria/inmunología , Plasmodium yoelii/inmunología , Animales , Bioensayo , Femenino , Inmunización , Inmunización Pasiva , Vacunas contra la Malaria , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Movimiento , Plasmodium yoelii/fisiología
20.
Nat Commun ; 9(1): 4964, 2018 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-30470758

RESUMEN

The inflammasome plays a critical role in inflammation and immune responses against pathogens. However, whether or how inflammasome activation regulates type I interferon (IFN-I) signaling in the context of malaria infection remain unknown. Here we show mice deficient in inflammasome sensors AIM2, NLRP3 or adaptor Caspase-1 produce high levels of IFN-I cytokines and are resistant to lethal Plasmodium yoelii YM infection. Inactivation of inflammasome signaling reduces interleukin (IL)-1ß production, but increases IFN-I production. Mechanistically, we show inflammsome activation enhances IL-1ß-mediated MyD88-TRAF3-IRF3 signaling and SOCS1 upregulation. However, SOCS1 inhibits MyD88-IRF7-mediated-IFN-I signaling and cytokine production in plasmacytoid dendritic cells. By contrast, ablation of inflammsome components reduces SOCS1 induction, and relieves its inhibition on MyD88-IRF7-dependent-IFN-I signaling, leading to high levels of IFN-α/ß production and host survival. Our study identifies a previously unrecognized role of inflammasome activation in the negative regulation of IFN-I signaling pathways and provides potential targets for developing effective malaria vaccines.


Asunto(s)
Inflamasomas/inmunología , Factor 7 Regulador del Interferón/inmunología , Malaria/inmunología , Factor 88 de Diferenciación Mieloide/inmunología , Plasmodium yoelii/fisiología , Animales , Regulación hacia Abajo , Femenino , Humanos , Inflamasomas/genética , Factor 7 Regulador del Interferón/genética , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Malaria/genética , Malaria/parasitología , Ratones , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/genética , Transducción de Señal , Proteína 1 Supresora de la Señalización de Citocinas/genética , Proteína 1 Supresora de la Señalización de Citocinas/inmunología , Factor 3 Asociado a Receptor de TNF/genética , Factor 3 Asociado a Receptor de TNF/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...