Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
PLoS Pathog ; 14(11): e1007418, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30496310

RESUMEN

Malaria is a global health concern caused by infection with Plasmodium parasites. With rising insecticide and drug resistance, there is a critical need to develop novel control strategies, including strategies to block parasite sporogony in key mosquito vector species. MAPK signaling pathways regulated by extracellular signal-regulated kinases (ERKs) and the stress-activated protein kinases (SAPKs) c-Jun N-terminal kinases (JNKs) and p38 MAPKs are highly conserved across eukaryotes, including mosquito vectors of the human malaria parasite Plasmodium falciparum. Some of these pathways in mosquitoes have been investigated in detail, but the mechanisms of integration of parasite development and mosquito fitness by JNK signaling have not been elucidated. To this end, we engineered midgut-specific overexpression of MAPK phosphatase 4 (MKP4), which targets the SAPKs, and used two potent and specific JNK small molecule inhibitors (SMIs) to assess the effects of JNK signaling manipulations on Anopheles stephensi fecundity, lifespan, intermediary metabolism, and P. falciparum development. MKP4 overexpression and SMI treatment reduced the proportion of P. falciparum-infected mosquitoes and decreased oocyst loads relative to controls. SMI-treated mosquitoes exhibited no difference in lifespan compared to controls, whereas genetically manipulated mosquitoes exhibited extended longevity. Metabolomics analyses of SMI-treated mosquitoes revealed insights into putative resistance mechanisms and the physiology behind lifespan extension, suggesting for the first time that P. falciparum-induced JNK signaling reduces mosquito longevity and increases susceptibility to infection, in contrast to previously published reports, likely via a critical interplay between the invertebrate host and parasite for nutrients that play essential roles during sporogonic development.


Asunto(s)
Anopheles/metabolismo , Anopheles/parasitología , Malaria Falciparum/metabolismo , Animales , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Interacciones Huésped-Parásitos/efectos de los fármacos , Proteínas de Insectos/metabolismo , Insectos Vectores/parasitología , Longevidad , Sistema de Señalización de MAP Quinasas/fisiología , Malaria/parasitología , Plasmodium/metabolismo , Plasmodium falciparum/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
2.
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
3.
PLoS Pathog ; 10(6): e1004231, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24968248

RESUMEN

Insulin and insulin-like growth factor signaling (IIS) regulates cell death, repair, autophagy, and renewal in response to stress, damage, and pathogen challenge. Therefore, IIS is fundamental to lifespan and disease resistance. Previously, we showed that insulin-like growth factor 1 (IGF1) within a physiologically relevant range (0.013-0.13 µM) in human blood reduced development of the human parasite Plasmodium falciparum in the Indian malaria mosquito Anopheles stephensi. Low IGF1 (0.013 µM) induced FOXO and p70S6K activation in the midgut and extended mosquito lifespan, whereas high IGF1 (0.13 µM) did not. In this study the physiological effects of low and high IGF1 were examined in detail to infer mechanisms for their dichotomous effects on mosquito resistance and lifespan. Following ingestion, low IGF1 induced phosphorylation of midgut c-Jun-N-terminal kinase (JNK), a critical regulator of epithelial homeostasis, but high IGF1 did not. Low and high IGF1 induced midgut mitochondrial reactive oxygen species (ROS) synthesis and nitric oxide (NO) synthase gene expression, responses which were necessary and sufficient to mediate IGF1 inhibition of P. falciparum development. However, increased ROS and apoptosis-associated caspase-3 activity returned to baseline levels following low IGF1 treatment, but were sustained with high IGF1 treatment and accompanied by aberrant expression of biomarkers for mitophagy, stem cell division and proliferation. Low IGF1-induced ROS are likely moderated by JNK-induced epithelial cytoprotection as well as p70S6K-mediated growth and inhibition of apoptosis over the lifetime of A. stephensi to facilitate midgut homeostasis and enhanced survivorship. Hence, mitochondrial integrity and homeostasis in the midgut, a key signaling center for IIS, can be targeted to coordinately optimize mosquito fitness and anti-pathogen resistance for improved control strategies for malaria and other vector-borne diseases.


Asunto(s)
Anopheles/efectos de los fármacos , Interacciones Huésped-Parásitos/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/farmacología , Mucosa Intestinal/efectos de los fármacos , Intestinos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Plasmodium falciparum/patogenicidad , Animales , Anopheles/crecimiento & desarrollo , Anopheles/metabolismo , Anopheles/parasitología , Control de Enfermedades Transmisibles , Femenino , Homeostasis/efectos de los fármacos , Hormesis , Humanos , Proteínas de Insectos/metabolismo , Insectos Vectores/efectos de los fármacos , Insectos Vectores/crecimiento & desarrollo , Insectos Vectores/metabolismo , Insectos Vectores/parasitología , Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Factor I del Crecimiento Similar a la Insulina/genética , Mucosa Intestinal/metabolismo , Longevidad/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Malaria Falciparum/prevención & control , Malaria Falciparum/transmisión , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Fosforilación/efectos de los fármacos , Plasmodium falciparum/aislamiento & purificación , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Recombinantes/farmacología
4.
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
5.
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
6.
Infect Immun ; 81(10): 3515-26, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23690397

RESUMEN

Coinfection with malaria and nontyphoidal Salmonella serotypes (NTS) can cause life-threatening bacteremia in humans. Coinfection with malaria is a recognized risk factor for invasive NTS, suggesting that malaria impairs intestinal barrier function. Here, we investigated mechanisms and strategies for prevention of coinfection pathology in a mouse model. Our findings reveal that malarial-parasite-infected mice, like humans, develop L-arginine deficiency, which is associated with intestinal mastocytosis, elevated levels of histamine, and enhanced intestinal permeability. Prevention or reversal of L-arginine deficiency blunts mastocytosis in ileal villi as well as bacterial translocation, measured as numbers of mesenteric lymph node CFU of noninvasive Escherichia coli Nissle and Salmonella enterica serotype Typhimurium, the latter of which is naturally invasive in mice. Dietary supplementation of malarial-parasite-infected mice with L-arginine or L-citrulline reduced levels of ileal transcripts encoding interleukin-4 (IL-4), a key mediator of intestinal mastocytosis and macromolecular permeability. Supplementation with L-citrulline also enhanced epithelial adherens and tight junctions in the ilea of coinfected mice. These data suggest that increasing L-arginine bioavailability via oral supplementation can ameliorate malaria-induced intestinal pathology, providing a basis for testing nutritional interventions to reduce malaria-associated mortality in humans.


Asunto(s)
Arginina/deficiencia , Bacteriemia/inmunología , Intestinos/citología , Malaria/complicaciones , Mastocitos/fisiología , Salmonelosis Animal/microbiología , Animales , Bacteriemia/microbiología , Citrulina , Femenino , Intestinos/inmunología , Intestinos/patología , Ratones , Permeabilidad , Plasmodium yoelii , Salmonelosis Animal/patología
7.
bioRxiv ; 2023 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-37397989

RESUMEN

Enhanced host immunity and competition for metabolic resources are two main competing hypotheses for the mechanism of Wolbachia-mediated pathogen inhibition in arthropods. Using an Anopheles mosquito - somatic Wolbachia infection - O'nyong nyong virus (ONNV) model, we demonstrate that the mechanism underpinning Wolbachia-mediated virus inhibition is up-regulation of the Toll innate immune pathway. However, the viral inhibitory properties of Wolbachia were abolished by cholesterol supplementation. This result was due to Wolbachia-dependent cholesterol-mediated suppression of Toll signaling rather than competition for cholesterol between Wolbachia and virus. The inhibitory effect of cholesterol was specific to Wolbachia-infected Anopheles mosquitoes and cells. These data indicate that both Wolbachia and cholesterol influence Toll immune signaling in Anopheles mosquitoes in a complex manner and provide a functional link between the host immunity and metabolic competition hypotheses for explaining Wolbachia-mediated pathogen interference in mosquitoes. In addition, these results provide a mechanistic understanding of the mode of action of Wolbachia-induced pathogen blocking in Anophelines, which is critical to evaluate the long-term efficacy of control strategies for malaria and Anopheles-transmitted arboviruses.

8.
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
9.
PLoS Pathog ; 6(7): e1001003, 2010 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-20664791

RESUMEN

Malaria (Plasmodium spp.) kills nearly one million people annually and this number will likely increase as drug and insecticide resistance reduces the effectiveness of current control strategies. The most important human malaria parasite, Plasmodium falciparum, undergoes a complex developmental cycle in the mosquito that takes approximately two weeks and begins with the invasion of the mosquito midgut. Here, we demonstrate that increased Akt signaling in the mosquito midgut disrupts parasite development and concurrently reduces the duration that mosquitoes are infective to humans. Specifically, we found that increased Akt signaling in the midgut of heterozygous Anopheles stephensi reduced the number of infected mosquitoes by 60-99%. Of those mosquitoes that were infected, we observed a 75-99% reduction in parasite load. In homozygous mosquitoes with increased Akt signaling parasite infection was completely blocked. The increase in midgut-specific Akt signaling also led to an 18-20% reduction in the average mosquito lifespan. Thus, activation of Akt signaling reduced the number of infected mosquitoes, the number of malaria parasites per infected mosquito, and the duration of mosquito infectivity.


Asunto(s)
Anopheles/parasitología , Interacciones Huésped-Parásitos , Estadios del Ciclo de Vida , Malaria/parasitología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Sistema Digestivo/parasitología , Humanos , Prevalencia , Transducción de Señal
10.
PLoS Pathog ; 6(8)2010 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-20714345

RESUMEN

Malaria (Plasmodium spp.) kills nearly one million people annually and this number will likely increase as drug and insecticide resistance reduces the effectiveness of current control strategies. The most important human malaria parasite, Plasmodium falciparum, undergoes a complex developmental cycle in the mosquito that takes approximately two weeks and begins with the invasion of the mosquito midgut. Here, we demonstrate that increased Akt signaling in the mosquito midgut disrupts parasite development and concurrently reduces the duration that mosquitoes are infective to humans. Specifically, we found that increased Akt signaling in the midgut of heterozygous Anopheles stephensi reduced the number of infected mosquitoes by 60-99%. Of those mosquitoes that were infected, we observed a 75-99% reduction in parasite load. In homozygous mosquitoes with increased Akt signaling parasite infection was completely blocked. The increase in midgut-specific Akt signaling also led to an 18-20% reduction in the average mosquito lifespan. Thus, activation of Akt signaling reduced the number of infected mosquitoes, the number of malaria parasites per infected mosquito, and the duration of mosquito infectivity.


Asunto(s)
Anopheles/parasitología , Malaria/parasitología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Sistema Digestivo/parasitología , Interacciones Huésped-Parásitos , Humanos , Estadios del Ciclo de Vida , Prevalencia , Transducción de Señal
11.
Artículo en Inglés | MEDLINE | ID: mdl-34594436

RESUMEN

It is difficult in asynchronous online instruction to keep students engaged and motivated. The rapid and unexpected nature of the move to online instruction has meant that the content presented to students has been primarily static and linear. Thus, there is a need for creative pedagogical approaches that re-create some level of the laboratory experience. One economical and accessible approach to building an interactive lab experience is making web-based interactive slides. In the virtual spaces created by this approach, students can explore different modalities of content in a nonlinear and asynchronous manner. We hope that this approach will make the learning process easier and more enjoyable for students while simultaneously making the complex content normally covered in microbiology labs more approachable. In this article we provide detailed instructions for producing web-based interactive slides as well as an example interactive slide that encompasses content that might normally be presented in an introductory microbiology class.

12.
Infect Immun ; 77(8): 3272-83, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19487470

RESUMEN

The precise role of Leishmania glycoconjugate molecules including phosphoglycans (PGs) and lipophosphoglycan (LPG) on host cellular responses is still poorly defined. Here, we investigated the interaction of Leishmania major LPG2 null mutant (lpg2(-)), which lacks both PGs and LPG, with dendritic cells (DCs) and the subsequent early immune response in infected mice. Surprisingly, the absence of phosphoglycans did not influence expression pattern of major histocompatibility complex class II (MHC II), CD40, CD80, and CD86 on DCs in vitro and in vivo. However, lpg2(-) L. major induced significantly higher production of interleukin-12p40 (IL-12p40) by infected bone marrow-derived DCs (BMDCs) than wild-type (WT) parasites in vitro. Furthermore, the production of IL-12p40 by draining lymph node cells from lpg2(-) mutant-infected mice was higher than those from WT L. major-infected mice. In model antigen presentation experiments, DCs from lpg2(-) mutant-infected mice induced more gamma interferon (IFN-gamma) and IL-2 production by Leishmania-specific T cells than those from WT-infected mice. Lymphocytes isolated from mice infected for 3 days with lpg2(-) parasites produce similar levels of IFN-gamma, but significantly less IL-4 and IL-10 than WT controls. Decreased IL-4 production was also seen in another general PG-deficient mutant lacking the Golgi UDP-galactose transporters (lpg5A(-) lpg5B(-)), but not with the lpg1(-) mutant lacking only LPG, thereby implicating PGs generally in the reduction of IL-4 production. Thus, Leishmania PGs influence host early immune response by modulating DC functions in a way that inhibits antigen presentation and promotes early IL-4 response, and their absence may impact the balance between Th1 and Th2 responses.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/parasitología , Factores Inmunológicos/farmacología , Leishmania major/química , Leishmania major/inmunología , Polisacáridos/farmacología , Animales , Presentación de Antígeno , Antígenos CD/metabolismo , Citocinas/metabolismo , Femenino , Glicoesfingolípidos/deficiencia , Antígenos de Histocompatibilidad Clase II/metabolismo , Factores Inmunológicos/inmunología , Subunidad p40 de la Interleucina-12/metabolismo , Proteínas de la Membrana/deficiencia , Ratones , Ratones Endogámicos BALB C , Polisacáridos/inmunología , Proteínas Protozoarias , Linfocitos T/inmunología
13.
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
15.
Curr Opin Immunol ; 36: 73-9, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26210301

RESUMEN

Malaria parasite-host interactions are complex and have confounded available drugs and the development of vaccines. Further, we now appreciate that interventions for malaria elimination and eradication must include therapeutics with intrinsic transmission blocking activity to treat the patient and prevent disease spread. Studies over the past 15 years have revealed significant conservation in the response to infection in mosquito and human hosts. More recently, we have recognized that conserved cell signaling cascades in mosquitoes and humans dictate infection outcome through the regulation of mitochondrial function and biogenesis, which feed back to host immunity, basic intermediary metabolism, and stress responses. These responses - reflected clearly in the primeval insect host - provide fertile ground for innovative strategies for both treatment and transmission blocking.


Asunto(s)
Interacciones Huésped-Parásitos , Malaria/inmunología , Malaria/metabolismo , Plasmodium/fisiología , Animales , Interacciones Huésped-Parásitos/inmunología , Humanos , Insulina/metabolismo , Malaria/parasitología , Mitocondrias/metabolismo , Proteínas Quinasas/metabolismo , Transducción de Señal , Somatomedinas/metabolismo
16.
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
17.
Int J Parasitol ; 33(13): 1537-44, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14572516

RESUMEN

A forkhead transcription factor gene, fktf-1, which we propose to be orthologous to the Caenorhabditis elegans dauer-regulatory gene daf-16 has been discovered in the parasitic nematode Strongyloides stercoralis. Genomic and cDNA sequences from both species predict alternately spliced a and b message isoforms. In contrast to C. elegans, where two a isoforms, daf-16a1 and daf-16a2, are found, a single fktf-1a isoform is found in S. stercoralis. Five of the 10 introns found in the C. elegans gene are found in the proposed S. stercoralis ortholog. Functional motifs common to DAF-16 and several mammalian forkhead transcription factors are conserved in FKTF-1. These include the forkhead DNA binding domain, four Akt/protein kinase B phosphorylation sites and a C-terminal domain that may associate with factors such as the steroid receptor coactivator and other factors necessary for transcriptional regulation. An N-terminal serine-rich domain found in DAF-16A is greatly expanded in FKTF-1A. This domain is missing in DAF-16B, FKTF-1B and all mammalian orthologs. FKTF-1 shows the closest phylogenetic relationship to DAF-16 among all known mammalian and nematode forkhead transcription factors. Like its proposed Caenorhabditis ortholog, the fktf-1 message is expressed at all stages of the life cycle examined thus far. Discovery of fktf-1 indicates the presence of an insulin-like signalling pathway in S. stercoralis similar to that known to regulate dauer development in C. elegans. This pathway is a likely candidate to control infective larval arrest and reactivation as well as regulation of the switch between parasitic and free-living development in the parasite.


Asunto(s)
Proteínas de Caenorhabditis elegans , Genes de Helminto , Strongyloides stercoralis/genética , Factores de Transcripción/genética , Animales , Secuencia de Bases , Caenorhabditis elegans/genética , Factores de Transcripción Forkhead , Expresión Génica , Humanos , Larva , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa/métodos , ARN Mensajero/análisis , Alineación de Secuencia , Homología de Secuencia , Strongyloides stercoralis/fisiología
18.
Curr Opin Insect Sci ; 3: 1-5, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25401083

RESUMEN

During the process of blood feeding insect vectors are exposed to an array of vertebrate-derived blood factors ranging from byproducts of blood meal digestion to naturally occurring products in the blood including growth hormones, cytokines and factors derived from blood-borne pathogens themselves. In this review, we examine the ability of these ingested vertebrate blood factors to alter the innate pathogen defenses of insect vectors. The ability of these factors to modify the immune responses of insect vectors offers new intriguing targets for blocking or reducing transmission of human disease-causing pathogens.

19.
Genome Biol ; 15(9): 459, 2014 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-25244985

RESUMEN

BACKGROUND: Anopheles stephensi is the key vector of malaria throughout the Indian subcontinent and Middle East and an emerging model for molecular and genetic studies of mosquito-parasite interactions. The type form of the species is responsible for the majority of urban malaria transmission across its range. RESULTS: Here, we report the genome sequence and annotation of the Indian strain of the type form of An. stephensi. The 221 Mb genome assembly represents more than 92% of the entire genome and was produced using a combination of 454, Illumina, and PacBio sequencing. Physical mapping assigned 62% of the genome onto chromosomes, enabling chromosome-based analysis. Comparisons between An. stephensi and An. gambiae reveal that the rate of gene order reshuffling on the X chromosome was three times higher than that on the autosomes. An. stephensi has more heterochromatin in pericentric regions but less repetitive DNA in chromosome arms than An. gambiae. We also identify a number of Y-chromosome contigs and BACs. Interspersed repeats constitute 7.1% of the assembled genome while LTR retrotransposons alone comprise more than 49% of the Y contigs. RNA-seq analyses provide new insights into mosquito innate immunity, development, and sexual dimorphism. CONCLUSIONS: The genome analysis described in this manuscript provides a resource and platform for fundamental and translational research into a major urban malaria vector. Chromosome-based investigations provide unique perspectives on Anopheles chromosome evolution. RNA-seq analysis and studies of immunity genes offer new insights into mosquito biology and mosquito-parasite interactions.


Asunto(s)
Anopheles/genética , Insectos Vectores/genética , Animales , Anopheles/metabolismo , Mapeo Cromosómico , Cromosomas de Insectos/genética , Análisis por Conglomerados , Evolución Molecular , Genoma de los Insectos , Humanos , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Malaria/transmisión , Filogenia , Polimorfismo de Nucleótido Simple , Análisis de Secuencia de ADN , Sintenía , Transcriptoma , Población Urbana
20.
Microbes Infect ; 15(3): 243-54, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23370408

RESUMEN

The blood feeding behavior of disease-transmitting arthropods creates a unique intersection between vertebrate and invertebrate physiology. Here, we review host blood-derived factors that persist through blood digestion to affect the lifespan, reproduction, and immune responses of some of the most common arthropod vectors of human disease.


Asunto(s)
Vectores Artrópodos/fisiología , Conducta Alimentaria , Animales , Vectores Artrópodos/inmunología , Proteínas Sanguíneas/inmunología , Culicidae/inmunología , Culicidae/fisiología , Interacciones Huésped-Parásitos , Humanos , Garrapatas/inmunología , Garrapatas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA