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











Base de datos
Intervalo de año de publicación
1.
Insect Biochem Mol Biol ; 139: 103669, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34666189

RESUMEN

In vertebrates and invertebrates, the insulin/insulin-like growth factor 1 (IGF1) signaling (IIS) cascade is highly conserved and plays a vital role in many different physiological processes. Among the many tissues that respond to IIS in mosquitoes, the fat body has a central role in metabolism, lifespan, reproduction, and innate immunity. We previously demonstrated that fat body specific expression of active Akt, a key IIS signaling molecule, in adult Anopheles stephensi and Aedes aegypti activated the IIS cascade and extended lifespan. Additionally, we found that transgenic females produced more vitellogenin (Vg) protein than non-transgenic mosquitoes, although this did not translate into increased fecundity. These results prompted us to further examine how IIS impacts immunity, metabolism, growth and development of these transgenic mosquitoes. We observed significant changes in glycogen, trehalose, triglycerides, glucose, and protein in young (3-5 d) transgenic mosquitoes relative to non-transgenic sibling controls, while only triglycerides were significantly changed in older (18 d) transgenic mosquitoes. More importantly, we demonstrated that enhanced fat body IIS decreased both the prevalence and intensity of Plasmodium falciparum infection in transgenic An. stephensi. Additionally, challenging transgenic An. stephensi with Gram-positive and Gram-negative bacteria altered the expression of several antimicrobial peptides (AMPs) and two anti-Plasmodium genes, nitric oxide synthase (NOS) and thioester complement-like protein (TEP1), relative to non-transgenic controls. Increased IIS in the fat body of adult female An. stephensi had little to no impact on body size, growth or development of progeny from transgenic mosquitoes relative to non-transgenic controls. This study both confirms and expands our understanding of the critical roles insulin signaling plays in regulating the diverse functions of the mosquito fat body.


Asunto(s)
Anopheles/fisiología , Cuerpo Adiposo/metabolismo , Interacciones Huésped-Patógeno , Insulina/fisiología , Transducción de Señal , Animales , Anopheles/microbiología , Anopheles/parasitología , Femenino , Bacterias Gramnegativas/fisiología , Bacterias Grampositivas/fisiología , Interacciones Huésped-Parásitos , Plasmodium falciparum/fisiología
2.
Ann Transl Med ; 9(15): 1273, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34532410

RESUMEN

BACKGROUND: Diabetic retinopathy is a retinal vasculopathy involving all three retinal capillary plexus layers. Since human CD34+ bone marrow stem cells (BMSCs) have the potential to promote revascularization of ischemic tissue, this study tests the hypothesis that intravitreal injection of human CD34+ BMSCs can have protective effects on all layers of the retinal vasculature in eyes with diabetic retinopathy. METHODS: Streptozotocin (STZ)-induced diabetic mice were injected intravitreally with 50,000 human CD34+ BMSCs or phosphate-buffered saline (PBS) into the right eye. Systemic immunosuppression with rapamycin and tacrolimus was started 5 days before the injection and maintained for study duration to prevent rejection of human cells. All mice were euthanized 4 weeks after intravitreal injection; both eyes were enucleated for retinal flat mount immunohistochemistry. The retinal vasculature was stained with Isolectin-GS-IB4. Confocal microscopy was used to image four circular areas of interest of retina, 1-mm diameter around the optic disc. Images of superficial, intermediate, and deep retinal capillary plexus layers within the areas of interest were obtained and analyzed using ImageJ software with the Vessel Analysis plugin to quantitate the retinal vascular density and vascular length density in the three plexus layers. RESULTS: Three distinct retinal capillary plexus layers were visualized and imaged using confocal microscopy. Eyes that received intravitreal injection of CD34+ BMSCs (N=9) had significantly higher vascular density and vascular length density in the superficial retinal capillary plexus when compared to the untreated contralateral eyes (N=9) or PBS treated control eyes (N=12; P values <0.05 using ANOVA followed by post-hoc tests). For the intermediate and deep plexus layers, the difference was not statistically significant. CONCLUSIONS: The protective effect of intravitreal injection of the human CD34+ BMSCs on the superficial retinal capillary plexus layers is demonstrated using confocal microscopy in this murine model of diabetic retinopathy.

4.
Biochem J ; 473(20): 3487-3503, 2016 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-27496548

RESUMEN

Insulin-like peptides (ILPs) play important roles in growth and metabolic homeostasis, but have also emerged as key regulators of stress responses and immunity in a variety of vertebrates and invertebrates. Furthermore, a growing literature suggests that insulin signaling-dependent metabolic provisioning can influence host responses to infection and affect infection outcomes. In line with these studies, we previously showed that knockdown of either of two closely related, infection-induced ILPs, ILP3 and ILP4, in the mosquito Anopheles stephensi decreased infection with the human malaria parasite Plasmodium falciparum through kinetically distinct effects on parasite death. However, the precise mechanisms by which ILP3 and ILP4 control the response to infection remained unknown. To address this knowledge gap, we used a complementary approach of direct ILP supplementation into the blood meal to further define ILP-specific effects on mosquito biology and parasite infection. Notably, we observed that feeding resulted in differential effects of ILP3 and ILP4 on blood-feeding behavior and P. falciparum development. These effects depended on ILP-specific regulation of intermediary metabolism in the mosquito midgut, suggesting a major contribution of ILP-dependent metabolic shifts to the regulation of infection resistance and parasite transmission. Accordingly, our data implicate endogenous ILP signaling in balancing intermediary metabolism for the host response to infection, affirming this emerging tenet in host-pathogen interactions with novel insights from a system of significant public health importance.


Asunto(s)
Insulina/química , Péptidos/farmacología , Animales , Anopheles/parasitología , Western Blotting , Conducta Alimentaria/fisiología , Femenino , Interacciones Huésped-Patógeno , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/metabolismo , Péptidos/química , Péptidos/uso terapéutico , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/patogenicidad
5.
Malar J ; 15: 231, 2016 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-27102766

RESUMEN

BACKGROUND: More than half of the world's population is at risk of malaria and simultaneously, many malaria-endemic regions are facing dramatic increases in the prevalence of type 2 diabetes. Studies in murine malaria models have examined the impact of malaria infection on type 2 diabetes pathology, it remains unclear how this chronic metabolic disorder impacts the transmission of malaria. In this report, the ability type 2 diabetic rodents infected with malaria to transmit parasites to Anopheles stephensi mosquitoes is quantified. METHODS: The infection prevalence and intensity of An. stephensi mosquitoes that fed upon control or type 2 diabetic C57BL/6 db/db mice infected with either lethal Plasmodium berghei NK65 or non-lethal Plasmodium yoelii 17XNL murine malaria strains were determined. Daily parasitaemias were also recorded. RESULTS: A higher percentage of mosquitoes (87.5 vs 61.5 % for P. yoelii and 76.9 vs 50 % for P. berghei) became infected following blood feeding on Plasmodium-infected type 2 diabetic mice compared to mosquitoes that fed on infected control animals, despite no significant differences in circulating gametocyte levels. CONCLUSIONS: These results suggest that type 2 diabetic mice infected with malaria are more efficient at infecting mosquitoes, raising the question of whether a similar synergy exists in humans.


Asunto(s)
Anopheles/parasitología , Diabetes Mellitus Tipo 2 , Insectos Vectores/parasitología , Malaria/transmisión , Plasmodium berghei/fisiología , Plasmodium yoelii/fisiología , Animales , Diabetes Mellitus Experimental/etiología , Diabetes Mellitus Experimental/parasitología , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/parasitología , Femenino , Malaria/parasitología , Ratones , Ratones Endogámicos C57BL
6.
Immunobiology ; 221(3): 468-74, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26626201

RESUMEN

Co-infections with malaria and non-typhoidal Salmonella serotypes (NTS) can present as life-threatening bacteremia, in contrast to self-resolving NTS diarrhea in healthy individuals. In previous work with our mouse model of malaria/NTS co-infection, we showed increased gut mastocytosis and increased ileal and plasma histamine levels that were temporally associated with increased gut permeability and bacterial translocation. Here, we report that gut mastocytosis and elevated plasma histamine are also associated with malaria in an animal model of falciparum malaria, suggesting a broader host distribution of this biology. In support of mast cell function in this phenotype, malaria/NTS co-infection in mast cell-deficient mice was associated with a reduction in gut permeability and bacteremia. Further, antihistamine treatment reduced bacterial translocation and gut permeability in mice with malaria, suggesting a contribution of mast cell-derived histamine to GI pathology and enhanced risk of bacteremia during malaria/NTS co-infection.


Asunto(s)
Histamina/metabolismo , Malaria/metabolismo , Malaria/parasitología , Mastocitos/metabolismo , Membrana Mucosa/metabolismo , Membrana Mucosa/parasitología , Animales , Coinfección , Modelos Animales de Enfermedad , Femenino , Histamina/sangre , Antagonistas de los Receptores Histamínicos/farmacología , Macaca mulatta , Malaria/tratamiento farmacológico , Malaria/inmunología , Malaria Falciparum/inmunología , Malaria Falciparum/metabolismo , Mastocitos/inmunología , Mastocitos/patología , Mastocitosis/inmunología , Mastocitosis/metabolismo , Ratones , Ratones Noqueados , Membrana Mucosa/efectos de los fármacos , Membrana Mucosa/microbiología , Permeabilidad , Infecciones por Salmonella/inmunología , Infecciones por Salmonella/metabolismo
7.
Parasit Vectors ; 8: 424, 2015 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-26283222

RESUMEN

BACKGROUND: Fruit flies and mammals protect themselves against infection by mounting immune and metabolic responses that must be balanced against the metabolic needs of the pathogens. In this context, p38 mitogen-activated protein kinase (MAPK)-dependent signaling is critical to regulating both innate immunity and metabolism during infection. Accordingly, we asked to what extent the Asian malaria mosquito Anopheles stephensi utilizes p38 MAPK signaling during infection with the human malaria parasite Plasmodium falciparum. METHODS: A. stephensi p38 MAPK (AsP38 MAPK) was identified and patterns of signaling in vitro and in vivo (midgut) were analyzed using phospho-specific antibodies and small molecule inhibitors. Functional effects of AsP38 MAPK inhibition were assessed using P. falciparum infection, quantitative real-time PCR, assays for reactive oxygen species and survivorship under oxidative stress, proteomics, and biochemical analyses. RESULTS: The genome of A. stephensi encodes a single p38 MAPK that is activated in the midgut in response to parasite infection. Inhibition of AsP38 MAPK signaling significantly reduced P. falciparum sporogonic development. This phenotype was associated with AsP38 MAPK regulation of mitochondrial physiology and stress responses in the midgut epithelium, a tissue critical for parasite development. Specifically, inhibition of AsP38 MAPK resulted in reduction in mosquito protein synthesis machinery, a shift in glucose metabolism, reduced mitochondrial metabolism, enhanced production of mitochondrial reactive oxygen species, induction of an array of anti-parasite effector genes, and decreased resistance to oxidative stress-mediated damage. Hence, P. falciparum-induced activation of AsP38 MAPK in the midgut facilitates parasite infection through a combination of reduced anti-parasite immune defenses and enhanced host protein synthesis and bioenergetics to minimize the impact of infection on the host and to maximize parasite survival, and ultimately, transmission. CONCLUSIONS: These observations suggest that, as in mammals, innate immunity and mitochondrial responses are integrated in mosquitoes and that AsP38 MAPK-dependent signaling facilitates mosquito survival during parasite infection, a fact that may attest to the relatively longer evolutionary relationship of these parasites with their invertebrate compared to their vertebrate hosts. On a practical level, improved understanding of the balances and trade-offs between resistance and metabolism could be leveraged to generate fit, resistant mosquitoes for malaria control.


Asunto(s)
Anopheles/inmunología , Metabolismo Energético , Inmunidad Innata , Plasmodium falciparum/inmunología , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Anopheles/metabolismo , Perfilación de la Expresión Génica , Estrés Oxidativo , Proteoma/análisis , Especies Reactivas de Oxígeno/análisis , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Supervivencia
8.
Parasit Vectors ; 7: 287, 2014 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-24957684

RESUMEN

BACKGROUND: Susceptibility to Plasmodium infection in Anopheles gambiae has been proposed to result from naturally occurring polymorphisms that alter the strength of endogenous innate defenses. Despite the fact that some of these mutations are known to introduce non-synonymous substitutions in coding sequences, these mutations have largely been used to rationalize knockdown of associated target proteins to query the effects on parasite development in the mosquito host. Here, we assay the effects of engineered mutations on an immune signaling protein target that is known to control parasite sporogonic development. By this proof-of-principle work, we have established that naturally occurring mutations can be queried for their effects on mosquito protein function and on parasite development and that this important signaling pathway can be genetically manipulated to enhance mosquito resistance. METHODS: We introduced SNPs into the A. gambiae MAPK kinase MEK to alter key residues in the N-terminal docking site (D-site), thus interfering with its ability to interact with the downstream kinase target ERK. ERK phosphorylation levels in vitro and in vivo were evaluated to confirm the effects of MEK D-site mutations. In addition, overexpression of various MEK D-site alleles was used to assess P. berghei infection in A. gambiae. RESULTS: The MEK D-site contains conserved lysine residues predicted to mediate protein-protein interaction with ERK. As anticipated, each of the D-site mutations (K3M, K6M) suppressed ERK phosphorylation and this inhibition was significant when both mutations were present. Tissue-targeted overexpression of alleles encoding MEK D-site polymorphisms resulted in reduced ERK phosphorylation in the midgut of A. gambiae. Furthermore, as expected, inhibition of MEK-ERK signaling due to D-site mutations resulted in reduction in P. berghei development relative to infection in the presence of overexpressed catalytically active MEK. CONCLUSION: MEK-ERK signaling in A. gambiae, as in model organisms and humans, depends on the integrity of conserved key residues within the MEK D-site. Disruption of signal transmission via engineered SNPs provides a purposeful proof-of-principle model for the study of naturally occurring mutations that may be associated with mosquito resistance to parasite infection as well as an alternative genetic basis for manipulation of this important immune signaling pathway.


Asunto(s)
Anopheles/genética , Anopheles/parasitología , Proteínas de Insectos/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Plasmodium berghei/crecimiento & desarrollo , Polimorfismo de Nucleótido Simple , Secuencia de Aminoácidos , Animales , Anopheles/metabolismo , Sitios de Unión , Línea Celular , Femenino , Proteínas de Insectos/genética , Leviviridae , Malaria/parasitología , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Datos de Secuencia Molecular , Plasmodium berghei/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos
9.
PLoS Pathog ; 9(2): e1003180, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23468624

RESUMEN

The overexpression of activated, myristoylated Akt in the midgut of female transgenic Anopheles stephensi results in resistance to infection with the human malaria parasite Plasmodium falciparum but also decreased lifespan. In the present study, the understanding of mitochondria-dependent midgut homeostasis has been expanded to explain this apparent paradox in an insect of major medical importance. Given that Akt signaling is essential for cell growth and survival, we hypothesized that sustained Akt activation in the mosquito midgut would alter the balance of critical pathways that control mitochondrial dynamics to enhance parasite killing at some cost to survivorship. Toxic reactive oxygen and nitrogen species (RNOS) rise to high levels in the midgut after blood feeding, due to a combination of high NO production and a decline in FOXO-dependent antioxidants. Despite an apparent increase in mitochondrial biogenesis in young females (3 d), energy deficiencies were apparent as decreased oxidative phosphorylation and increased [AMP]/[ATP] ratios. In addition, mitochondrial mass was lower and accompanied by the presence of stalled autophagosomes in the posterior midgut, a critical site for blood digestion and stem cell-mediated epithelial maintenance and repair, and by functional degradation of the epithelial barrier. By 18 d, the age at which An. stephensi would transmit P. falciparum to human hosts, mitochondrial dysfunction coupled to Akt-mediated repression of autophagy/mitophagy was more evident and midgut epithelial structure was markedly compromised. Inhibition of RNOS by co-feeding of the nitric-oxide synthase inhibitor L-NAME at infection abrogated Akt-dependent killing of P. falciparum that begins within 18 h of infection in 3-5 d old mosquitoes. Hence, Akt-induced changes in mitochondrial dynamics perturb midgut homeostasis to enhance parasite resistance and decrease mosquito infective lifespan. Further, quality control of mitochondrial function in the midgut is necessary for the maintenance of midgut health as reflected in energy homeostasis and tissue repair and renewal.


Asunto(s)
Anopheles/parasitología , Interacciones Huésped-Parásitos , Malaria Falciparum/prevención & control , Enfermedades Mitocondriales/metabolismo , Plasmodium falciparum/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Resistencia a la Enfermedad , Femenino , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/parasitología , Humanos , Proteínas de Insectos/biosíntesis , Masculino , Mitocondrias/metabolismo , Mitocondrias/parasitología , Mitocondrias/ultraestructura , Enfermedades Mitocondriales/parasitología , Transducción de Señal
10.
J Exp Biol ; 216(Pt 2): 208-17, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23255191

RESUMEN

The highly conserved insulin/insulin-like growth factor (IGF) signaling (IIS) pathway regulates metabolism, development, lifespan and immunity across a wide range of organisms. Previous studies have shown that human insulin ingested in the blood meal can activate mosquito IIS, resulting in attenuated lifespan and increased malaria parasite infection. Because human IGF1 is present at higher concentrations in blood than insulin and is functionally linked with lifespan and immune processes, we predicted that human IGF1 ingested in a blood meal would affect lifespan and malaria parasite infection in the mosquito Anopheles stephensi. Here we demonstrate that physiological levels of ingested IGF1, like insulin, can persist intact in the blood-filled midgut for up to 30 h and disseminate into the mosquito body, and that both peptides activate IIS in mosquito cells and midgut. At these same levels, ingested IGF1 alone extended average mosquito lifespan by 23% compared with controls and, more significantly, when ingested in infected blood meals, reduced the prevalence of Plasmodium falciparum-infected mosquitoes by >20% and parasite load by 35-50% compared with controls. Thus, the effects of ingested IGF1 on mosquito lifespan and immunity are opposite to those of ingested insulin. These results offer the first evidence that insect cells can functionally discriminate between mammalian insulin and IGF1. Further, in light of previous success in genetically targeting IIS to alter mosquito lifespan and malaria parasite transmission, this study indicates that a more complete understanding of the IIS-activating ligands in blood can be used to optimize transgenic strategies for malaria control.


Asunto(s)
Anopheles/parasitología , Interacciones Huésped-Parásitos , Insectos Vectores/parasitología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Malaria/transmisión , Plasmodium falciparum/patogenicidad , Animales , Anopheles/fisiología , Línea Celular , Sistema Digestivo/metabolismo , Sistema Digestivo/parasitología , Eritrocitos/parasitología , Femenino , Factores de Transcripción Forkhead/metabolismo , Humanos , Proteínas de Insectos/metabolismo , Insectos Vectores/fisiología , Insulina/metabolismo , Malaria/parasitología , Plasmodium falciparum/crecimiento & desarrollo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
11.
Infect Immun ; 80(6): 2141-9, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22473605

RESUMEN

We showed previously that ingested human insulin activates the insulin/IGF-1 signaling pathway in Anopheles stephensi and increases the susceptibility of these mosquitoes to Plasmodium falciparum. In other organisms, insulin can alter immune responsiveness through regulation of NF-κB transcription factors, critical elements for innate immunity that are also central to mosquito immunity. We show here that insulin signaling decreased expression of NF-κB-regulated immune genes in mosquito cells stimulated with either bacterial or malarial soluble products. Further, human insulin suppressed mosquito immunity through sustained phosphatidylinositol 3-kinase activation, since inhibition of this pathway led to decreased parasite development in the mosquito. Together, these data demonstrate that activation of the insulin/IGF-1 signaling pathway by ingested human insulin can alter NF-κB-dependent immunity, and ultimately the susceptibility, of mosquitoes to P. falciparum.


Asunto(s)
Anopheles/efectos de los fármacos , Anopheles/inmunología , Insulina/farmacología , FN-kappa B/metabolismo , Plasmodium falciparum/inmunología , Animales , Anopheles/parasitología , Células Cultivadas , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Lipopolisacáridos , FN-kappa B/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Regiones Promotoras Genéticas , Transducción de Señal
12.
Antioxid Redox Signal ; 14(6): 943-55, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21126166

RESUMEN

Reactive oxygen species (ROS) have been implicated in direct killing of pathogens, increased tissue damage, and regulation of immune signaling pathways in mammalian cells. Available research suggests that analogous phenomena affect the establishment of Plasmodium infection in Anopheles mosquitoes. We have previously shown that provision of human insulin in a blood meal leads to increased ROS levels in Anopheles stephensi. Here, we demonstrate that provision of human insulin significantly increased parasite development in the same mosquito host in a manner that was not consistent with ROS-induced parasite killing or parasite escape through damaged tissue. Rather, our studies demonstrate that ROS are important mediators of both the mitogen-activated protein kinase and phosphatidylinositol 3-kinase/Akt signaling branches of the mosquito insulin signaling cascade. Further, ROS alone can directly activate these signaling pathways and this activation is growth factor specific. Our data, therefore, highlight a novel role for ROS as signaling mediators in the mosquito innate immune response to Plasmodium parasites.


Asunto(s)
Culicidae/parasitología , Malaria/inmunología , Plasmodium falciparum/crecimiento & desarrollo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología , Animales , Western Blotting , Células Cultivadas , Culicidae/inmunología , Femenino , Humanos
13.
Infect Immun ; 78(4): 1520-7, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20100860

RESUMEN

Severe pediatric malaria is an important risk factor for developing disseminated infections with nontyphoidal Salmonella serotypes (NTS). While recent animal studies on this subject are lacking, early work suggests that an increased risk for developing systemic NTS infection during malaria is caused by hemolytic anemia, which leads to reduced macrophage microbicidal activity. Here we established a model for oral Salmonella enterica serotype Typhimurium challenge in mice infected with Plasmodium yoelii nigeriensis. Initial characterization of this model showed that 5 days after coinoculation, P. yoelii nigeriensis infection increased the recovery of S. Typhimurium from liver and spleen by approximately 1,000-fold. The increased bacterial burden could be only partially recapitulated by antibody-mediated hemolysis, which increased the recovery of S. Typhimurium from liver and spleen by 10-fold. These data suggested that both hemolysis and P. yoelii nigeriensis-specific factors contributed to the increased susceptibility to S. Typhimurium. The mechanism by which hemolysis impaired resistance to S. Typhimurium was further investigated. In vitro, S. Typhimurium was recovered 24 h after infection of hemophagocytic macrophages in 2-fold-higher numbers than after infection of mock-treated macrophages, making it unlikely that reduced macrophage microbicidal activity was solely responsible for hemolysis-induced immunosuppression during malaria. Infection with P. yoelii nigeriensis, but not antibody-mediated hemolysis, reduced serum levels of interleukin-12p70 (IL-12p70) in response to S. Typhimurium challenge. Collectively, studies establishing a mouse model for this coinfection suggest that multiple distinct malaria-induced immune defects contribute to increased susceptibility to S. Typhimurium.


Asunto(s)
Anemia Hemolítica/complicaciones , Malaria/complicaciones , Salmonelosis Animal/microbiología , Salmonella typhimurium/patogenicidad , Animales , Recuento de Colonia Microbiana , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Femenino , Humanos , Interleucina-12/sangre , Hígado/microbiología , Ratones , Plasmodium yoelii/patogenicidad , Salmonelosis Animal/inmunología , Salmonella typhimurium/inmunología , Bazo/microbiología
14.
J Vis Exp ; (46)2010 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-21206476

RESUMEN

Plasmodium parasites, the causative agent of malaria, are transmitted through the bites of infected Anopheles mosquitoes resulting in over 250 million new infections each year. Despite decades of research, there is still no vaccine against malaria, highlighting the need for novel control strategies. One innovative approach is the use of genetically modified mosquitoes to effectively control malaria parasite transmission. Deliberate alterations of cell signaling pathways in the mosquito, via targeted mutagenesis, have been found to regulate parasite development (1). From these studies, we can begin to identify potential gene targets for transformation. Targeted mutagenesis has traditionally relied upon the homologous recombination between a target gene and a large DNA molecule. However, the construction and use of such complex DNA molecules for generation of stably transformed cell lines is costly, time consuming and often inefficient. Therefore, a strategy using locked nucleic acid-modified oligonucleotides (LNA-ONs) provides a useful alternative for introducing artificial single nucleotide substitutions into episomal and chromosomal DNA gene targets (reviewed in (2)). LNA-ON-mediated targeted mutagenesis has been used to introduce point mutations into genes of interest in cultured cells of both yeast and mice (3,4). We show here that LNA-ONs can be used to introduce a single nucleotide change in a transfected episomal target that results in a switch from blue fluorescent protein (BFP) expression to green fluorescent protein (GFP) expression in both Anopheles gambiae and Anopheles stephensi cells. This conversion demonstrates for the first time that effective mutagenesis of target genes in mosquito cells can be mediated by LNA-ONs and suggests that this technique may be applicable to mutagenesis of chromosomal targets in vitro and in vivo.


Asunto(s)
Anopheles/genética , Insectos Vectores/genética , Mutagénesis Sitio-Dirigida/métodos , Transfección/métodos , Animales , Anopheles/metabolismo , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Insectos Vectores/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo
15.
PLoS Pathog ; 5(4): e1000366, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19343212

RESUMEN

Malaria is caused by infection with intraerythrocytic protozoa of the genus Plasmodium that are transmitted by Anopheles mosquitoes. Although a variety of anti-parasite effector genes have been identified in anopheline mosquitoes, little is known about the signaling pathways that regulate these responses during parasite development. Here we demonstrate that the MEK-ERK signaling pathway in Anopheles is controlled by ingested human TGF-beta1 and finely tunes mosquito innate immunity to parasite infection. Specifically, MEK-ERK signaling was dose-dependently induced in response to TGF-beta1 in immortalized cells in vitro and in the A. stephensi midgut epithelium in vivo. At the highest treatment dose of TGF-beta1, inhibition of ERK phosphorylation increased TGF-beta1-induced expression of the anti-parasite effector gene nitric oxide synthase (NOS), suggesting that increasing levels of ERK activation negatively feed back on induced NOS expression. At infection levels similar to those found in nature, inhibition of ERK activation reduced P. falciparum oocyst loads and infection prevalence in A. stephensi and enhanced TGF-beta1-mediated control of P. falciparum development. Taken together, our data demonstrate that malaria parasite development in the mosquito is regulated by a conserved MAPK signaling pathway that mediates the effects of an ingested cytokine.


Asunto(s)
Anopheles/inmunología , Anopheles/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas de Insectos/metabolismo , Sistema de Señalización de MAP Quinasas , Plasmodium falciparum/inmunología , Factor de Crecimiento Transformador beta1/metabolismo , Análisis de Varianza , Animales , Células Cultivadas , Distribución de Chi-Cuadrado , Regulación de la Expresión Génica , Inmunidad Innata , Óxido Nítrico Sintasa de Tipo II/metabolismo , Plasmodium falciparum/crecimiento & desarrollo , Transducción de Señal , Estadísticas no Paramétricas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA