Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 26
1.
bioRxiv ; 2023 Jun 01.
Article En | MEDLINE | ID: mdl-37397989

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.

2.
Article En | MEDLINE | ID: mdl-34594436

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.

4.
PLoS Pathog ; 14(11): e1007418, 2018 11.
Article En | MEDLINE | ID: mdl-30496310

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.


Anopheles/metabolism , Anopheles/parasitology , Malaria, Falciparum/metabolism , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , Host-Parasite Interactions/drug effects , Insect Proteins/metabolism , Insect Vectors/parasitology , Longevity , MAP Kinase Signaling System/physiology , Malaria/parasitology , Plasmodium/metabolism , Plasmodium falciparum/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Biochem J ; 473(20): 3487-3503, 2016 Oct 15.
Article En | MEDLINE | ID: mdl-27496548

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.


Insulin/chemistry , Peptides/pharmacology , Animals , Anopheles/parasitology , Blotting, Western , Feeding Behavior/physiology , Female , Host-Pathogen Interactions , Insect Proteins/genetics , Insect Proteins/metabolism , Malaria, Falciparum/drug therapy , Malaria, Falciparum/metabolism , Peptides/chemistry , Peptides/therapeutic use , Plasmodium falciparum/drug effects , Plasmodium falciparum/pathogenicity
6.
Malar J ; 15: 231, 2016 Apr 21.
Article En | MEDLINE | ID: mdl-27102766

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.


Anopheles/parasitology , Diabetes Mellitus, Type 2 , Insect Vectors/parasitology , Malaria/transmission , Plasmodium berghei/physiology , Plasmodium yoelii/physiology , Animals , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Experimental/parasitology , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/parasitology , Female , Malaria/parasitology , Mice , Mice, Inbred C57BL
7.
Immunobiology ; 221(3): 468-74, 2016 Mar.
Article En | MEDLINE | ID: mdl-26626201

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.


Histamine/metabolism , Malaria/metabolism , Malaria/parasitology , Mast Cells/metabolism , Mucous Membrane/metabolism , Mucous Membrane/parasitology , Animals , Coinfection , Disease Models, Animal , Female , Histamine/blood , Histamine Antagonists/pharmacology , Macaca mulatta , Malaria/drug therapy , Malaria/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/metabolism , Mast Cells/immunology , Mast Cells/pathology , Mastocytosis/immunology , Mastocytosis/metabolism , Mice , Mice, Knockout , Mucous Membrane/drug effects , Mucous Membrane/microbiology , Permeability , Salmonella Infections/immunology , Salmonella Infections/metabolism
8.
Parasit Vectors ; 8: 424, 2015 Aug 19.
Article En | MEDLINE | ID: mdl-26283222

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.


Anopheles/immunology , Energy Metabolism , Immunity, Innate , Plasmodium falciparum/immunology , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Anopheles/metabolism , Gene Expression Profiling , Oxidative Stress , Proteome/analysis , Reactive Oxygen Species/analysis , Real-Time Polymerase Chain Reaction , Survival Analysis
9.
Curr Opin Immunol ; 36: 73-9, 2015 Oct.
Article En | MEDLINE | ID: mdl-26210301

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.


Host-Parasite Interactions , Malaria/immunology , Malaria/metabolism , Plasmodium/physiology , Animals , Host-Parasite Interactions/immunology , Humans , Insulin/metabolism , Malaria/parasitology , Mitochondria/metabolism , Protein Kinases/metabolism , Signal Transduction , Somatomedins/metabolism
10.
Curr Opin Insect Sci ; 3: 1-5, 2014 Sep 01.
Article En | MEDLINE | ID: mdl-25401083

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.

11.
Genome Biol ; 15(9): 459, 2014 Sep 23.
Article En | MEDLINE | ID: mdl-25244985

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.


Anopheles/genetics , Insect Vectors/genetics , Animals , Anopheles/metabolism , Chromosome Mapping , Chromosomes, Insect/genetics , Cluster Analysis , Evolution, Molecular , Genome, Insect , Humans , Insect Proteins/genetics , Insect Proteins/metabolism , Malaria/transmission , Phylogeny , Polymorphism, Single Nucleotide , Sequence Analysis, DNA , Synteny , Transcriptome , Urban Population
12.
PLoS Pathog ; 10(6): e1004231, 2014 Jun.
Article En | MEDLINE | ID: mdl-24968248

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.


Anopheles/drug effects , Host-Parasite Interactions/drug effects , Insulin-Like Growth Factor I/pharmacology , Intestinal Mucosa/drug effects , Intestines/drug effects , Oxidative Stress/drug effects , Plasmodium falciparum/pathogenicity , Animals , Anopheles/growth & development , Anopheles/metabolism , Anopheles/parasitology , Communicable Disease Control , Female , Homeostasis/drug effects , Hormesis , Humans , Insect Proteins/metabolism , Insect Vectors/drug effects , Insect Vectors/growth & development , Insect Vectors/metabolism , Insect Vectors/parasitology , Insulin-Like Growth Factor I/administration & dosage , Insulin-Like Growth Factor I/genetics , Intestinal Mucosa/metabolism , Longevity/drug effects , MAP Kinase Signaling System/drug effects , Malaria, Falciparum/prevention & control , Malaria, Falciparum/transmission , Mitochondria/drug effects , Mitochondria/metabolism , Phosphorylation/drug effects , Plasmodium falciparum/isolation & purification , Protein Processing, Post-Translational/drug effects , Recombinant Proteins/pharmacology
13.
PLoS One ; 8(10): e76535, 2013.
Article En | MEDLINE | ID: mdl-24146884

Anopheline mosquitoes are the primary vectors of parasites in the genus Plasmodium, the causative agents of malaria. Malaria parasites undergo a series of complex transformations upon ingestion by the mosquito host. During this process, the physical barrier of the midgut epithelium, along with innate immune defenses, functionally restrict parasite development. Although these defenses have been studied for some time, the regulatory factors that control them are poorly understood. The protein kinase C (PKC) gene family consists of serine/threonine kinases that serve as central signaling molecules and regulators of a broad spectrum of cellular processes including epithelial barrier function and immunity. Indeed, PKCs are highly conserved, ranging from 7 isoforms in Drosophila to 16 isoforms in mammals, yet none have been identified in mosquitoes. Despite conservation of the PKC gene family and their potential as targets for transmission-blocking strategies for malaria, no direct connections between PKCs, the mosquito immune response or epithelial barrier integrity are known. Here, we identify and characterize six PKC gene family members--PKCδ, PKCε, PKCζ, PKD, PKN, and an indeterminate conventional PKC--in Anopheles gambiae and Anopheles stephensi. Sequence and phylogenetic analyses of the anopheline PKCs support most subfamily assignments. All six PKCs are expressed in the midgut epithelia of A. gambiae and A. stephensi post-blood feeding, indicating availability for signaling in a tissue that is critical for malaria parasite development. Although inhibition of PKC enzymatic activity decreased NF-κB-regulated anti-microbial peptide expression in mosquito cells in vitro, PKC inhibition had no effect on expression of a panel of immune genes in the midgut epithelium in vivo. PKC inhibition did, however, significantly increase midgut barrier integrity and decrease development of P. falciparum oocysts in A. stephensi, suggesting that PKC-dependent signaling is a negative regulator of epithelial barrier function and a potential new target for transmission-blocking strategies.


Anopheles/enzymology , Anopheles/parasitology , Digestive System/parasitology , Epithelium/parasitology , Malaria/parasitology , Protein Kinase C/metabolism , Signal Transduction , Animals , Anopheles/drug effects , Bayes Theorem , Digestive System/drug effects , Enzyme Activation/drug effects , Epithelium/drug effects , Lipopolysaccharides/pharmacology , Multigene Family , NF-kappa B/genetics , Parasites/drug effects , Parasites/physiology , Phylogeny , Plasmodium falciparum/drug effects , Plasmodium falciparum/physiology , Promoter Regions, Genetic/genetics , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/chemistry , Protein Kinase C/genetics , Receptors, Pattern Recognition/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
14.
Microbes Infect ; 15(12): 775-87, 2013 Nov.
Article En | MEDLINE | ID: mdl-23774695

The insulin/insulin-like growth factor signaling (IIS) cascade is highly conserved and regulates diverse physiological processes such as metabolism, lifespan, reproduction and immunity. Transgenic overexpression of Akt, a critical regulator of IIS, was previously shown to shorten mosquito lifespan and increase resistance to the human malaria parasite Plasmodium falciparum. To further understand how IIS controls mosquito physiology and resistance to malaria parasite infection, we overexpressed an inhibitor of IIS, phosphatase and tensin homolog (PTEN), in the Anopheles stephensi midgut. PTEN overexpression inhibited phosphorylation of the IIS protein FOXO, an expected target for PTEN, in the midgut of A. stephensi. Further, PTEN overexpression extended mosquito lifespan and increased resistance to P. falciparum development. The reduction in parasite development did not appear to be due to alterations in an innate immune response, but rather was associated with increased expression of genes regulating autophagy and stem cell maintenance in the midgut and with enhanced midgut barrier integrity. In light of previous success in genetically targeting the IIS pathway to alter mosquito lifespan and malaria parasite transmission, these data confirm that multiple strategies to genetically manipulate IIS can be leveraged to generate fit, resistant mosquitoes for malaria control.


Anopheles/immunology , Gene Expression , Host-Pathogen Interactions , PTEN Phosphohydrolase/biosynthesis , Plasmodium falciparum/growth & development , Animals , Anopheles/enzymology , Anopheles/genetics , Anopheles/parasitology , Arthropod Proteins/genetics , Arthropod Proteins/metabolism , Autophagy , Female , Longevity , PTEN Phosphohydrolase/genetics , Plasmodium falciparum/immunology , Stem Cells/physiology
15.
Infect Immun ; 81(10): 3515-26, 2013 Oct.
Article En | MEDLINE | ID: mdl-23690397

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.


Arginine/deficiency , Bacteremia/immunology , Intestines/cytology , Malaria/complications , Mast Cells/physiology , Salmonella Infections, Animal/microbiology , Animals , Bacteremia/microbiology , Citrulline , Female , Intestines/immunology , Intestines/pathology , Mice , Permeability , Plasmodium yoelii , Salmonella Infections, Animal/pathology
16.
PLoS Pathog ; 9(2): e1003180, 2013 Feb.
Article En | MEDLINE | ID: mdl-23468624

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.


Anopheles/parasitology , Host-Parasite Interactions , Malaria, Falciparum/prevention & control , Mitochondrial Diseases/metabolism , Plasmodium falciparum/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Disease Resistance , Female , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/parasitology , Humans , Insect Proteins/biosynthesis , Male , Mitochondria/metabolism , Mitochondria/parasitology , Mitochondria/ultrastructure , Mitochondrial Diseases/parasitology , Signal Transduction
17.
Microbes Infect ; 15(3): 243-54, 2013 Mar.
Article En | MEDLINE | ID: mdl-23370408

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.


Arthropod Vectors/physiology , Feeding Behavior , Animals , Arthropod Vectors/immunology , Blood Proteins/immunology , Culicidae/immunology , Culicidae/physiology , Host-Parasite Interactions , Humans , Ticks/immunology , Ticks/physiology
18.
Infect Immun ; 80(6): 2141-9, 2012 Jun.
Article En | MEDLINE | ID: mdl-22473605

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.


Anopheles/drug effects , Anopheles/immunology , Insulin/pharmacology , NF-kappa B/metabolism , Plasmodium falciparum/immunology , Animals , Anopheles/parasitology , Cells, Cultured , Female , Gene Expression Regulation/drug effects , Humans , Lipopolysaccharides , NF-kappa B/genetics , Phosphatidylinositol 3-Kinases/metabolism , Promoter Regions, Genetic , Signal Transduction
19.
Antioxid Redox Signal ; 14(6): 943-55, 2011 Mar 15.
Article En | MEDLINE | ID: mdl-21126166

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.


Culicidae/parasitology , Malaria/immunology , Plasmodium falciparum/growth & development , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , Animals , Blotting, Western , Cells, Cultured , Culicidae/immunology , Female , Humans
20.
PLoS Pathog ; 6(8)2010 Aug 10.
Article En | MEDLINE | ID: mdl-20714345

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.


Anopheles/parasitology , Malaria/parasitology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Digestive System/parasitology , Host-Parasite Interactions , Humans , Life Cycle Stages , Prevalence , Signal Transduction
...