14 research outputs found

    Testing non-autonomous antimalarial gene drive effectors using self-eliminating drivers in the African mosquito vector Anopheles gambiae

    Get PDF
    Gene drives for mosquito population modification are novel tools for malaria control. Strategies to safely test antimalarial effectors in the field are required. Here, we modified the Anopheles gambiae zpg locus to host a CRISPR/Cas9 integral gene drive allele (zpgD) and characterized its behaviour and resistance profile. We found that zpgD dominantly sterilizes females but can induce efficient drive at other loci when it itself encounters resistance. We combined zpgD with multiple previously characterized non-autonomous payload drives and found that, as zpgD self-eliminates, it leads to conversion of mosquito cage populations at these loci. Our results demonstrate how self-eliminating drivers could allow safe testing of non-autonomous effector-traits by local population modification. They also suggest that after engendering resistance, gene drives intended for population suppression could nevertheless serve to propagate subsequently released non-autonomous payload genes, allowing modification of vector populations initially targeted for suppression

    PIMMS43 is required for malaria parasite immune evasion and sporogonic development in the mosquito vector.

    Get PDF
    After being ingested by a female Anopheles mosquito during a bloodmeal on an infected host, and before they can reach the mosquito salivary glands to be transmitted to a new host, Plasmodium parasites must establish an infection of the mosquito midgut in the form of oocysts. To achieve this, they must first survive a series of robust innate immune responses that take place prior to, during, and immediately after ookinete traversal of the midgut epithelium. Understanding how parasites may evade these responses could highlight new ways to block malaria transmission. We show that an ookinete and sporozoite surface protein designated as PIMMS43 (Plasmodium Infection of the Mosquito Midgut Screen 43) is required for parasite evasion of the Anopheles coluzzii complement-like response. Disruption of PIMMS43 in the rodent malaria parasite Plasmodium berghei triggers robust complement activation and ookinete elimination upon mosquito midgut traversal. Silencing components of the complement-like system through RNAi largely restores ookinete-to-oocyst transition but oocysts remain small in size and produce a very small number of sporozoites that additionally are not infectious, indicating that PIMMS43 is also essential for sporogonic development in the oocyst. Antibodies that bind PIMMS43 interfere with parasite immune evasion when ingested with the infectious blood meal and significantly reduce the prevalence and intensity of infection. PIMMS43 genetic structure across African Plasmodium falciparum populations indicates allelic adaptation to sympatric vector populations. These data add to our understanding of mosquito-parasite interactions and identify PIMMS43 as a target of malaria transmission blocking

    Microbiota-induced peritrophic matrix regulates midgut homeostasis and prevents systemic infection of malaria vector mosquitoes

    Get PDF
    <div><p>Manipulation of the mosquito gut microbiota can lay the foundations for novel methods for disease transmission control. Mosquito blood feeding triggers a significant, transient increase of the gut microbiota, but little is known about the mechanisms by which the mosquito controls this bacterial growth whilst limiting inflammation of the gut epithelium. Here, we investigate the gut epithelial response to the changing microbiota load upon blood feeding in the malaria vector <i>Anopheles coluzzii</i>. We show that the synthesis and integrity of the peritrophic matrix, which physically separates the gut epithelium from its luminal contents, is microbiota dependent. We reveal that the peritrophic matrix limits the growth and persistence of <i>Enterobacteriaceae</i> within the gut, whilst preventing seeding of a systemic infection. Our results demonstrate that the peritrophic matrix is a key regulator of mosquito gut homeostasis and establish functional analogies between this and the mucus layers of the mammalian gastrointestinal tract.</p></div

    The peritrophic matrix prevents microbiota dissemination and systemic immune induction.

    No full text
    <p><b>(A-C)</b><i>CEC1</i> expression (A), <i>16S rRNA</i> quantification (B) and <i>Enterobacteriaceae 16S rRNA</i> quantification (C) in the carcass 72h after feeding with a blood meal supplemented with 100μM polyoxin D or water as a control, plus or minus antibiotic treatment, as determined by qRT-PCR. Each dot represents a pool of 8–10 carcasses, derived from 4 independent experiments. Data show mean and standard error. In A, ratios are normalized within biological replicates to the mean of the control pools (no polyoxin D, no antibiotics). In B-C, ratios are normalized within each biological replicate to the highest value across all conditions (‘100%’). In A-C, statistical significance was assessed by an ANOVA on a linear mixed effect regression model. <b>(D)</b> Scatter plots of relative <i>Enterobacteriaceae</i> load against <i>CEC1</i> expression in the carcass at 72h post blood feeding. Each dot represents a pool of 8–10 carcasses, derived from 4 independent experiments; data are normalized as in B and C. Spearman’s rank correlation coefficient and associated p-values are indicated. ‘*’ p<0.05.</p

    The peritrophic matrix regulates immune resistance to the microbiota.

    No full text
    <p><b>(A)</b> RNA-seq transcriptional profiles of <i>CEC1</i> and <i>GAM1</i> in the midgut of control (black lines) and antibiotic fed (grey lines) mosquitoes over a two blood meal (BM1 and BM2) time course. Dots indicate normalized counts in each of four biological replicates, with the line connecting the means. Statistical significance of a pairwise comparison of counts at each time point was assessed by a Wald test with a Benjamini-Hochberg correction. ‘*’ p<0.05; ‘**’ p<0.01; ‘***’ p<0.001 (<b>B)</b> <i>GAM1</i> expression, relative to AgS7, in the midgut 24h after feeding with blood supplemented with 100μM polyoxin D (PxD) or an equal volume of water (control), plus or minus antibiotic treatment, as determined by qRT-PCR. Mean plus/minus standard error is indicated. Statistical significance was assessed by an ANOVA on a linear mixed effect regression model. Each dot represents a pool of 8–10 guts, derived from 4–5 independent experiments. Ratios are normalized within biological replicates to the mean of the control pools (no polyoxin D, no antibiotics). <b>(C)</b> <i>Enterobacteriaceae</i> load, relative to AgS7, as determined by qRT-PCR using <i>Enterobacteriaceae</i> specific 16S primers. Normalization and statistical analysis were performed as described for (B). <b>(D)</b> Scatter plots of the load of specific bacteria families commonly found in the mosquito gut against <i>GAM1</i> expression in the midguts of control (top row) or polyoxin D-treated (bottom row) mosquitoes. Spearman’s rank correlation coefficient (r) and associated p-values (p) are indicated.</p

    The peritrophic matrix promotes restoration of gut homeostasis after blood feeding.

    No full text
    <p><b>(A)</b> Midgut bacterial load at 24h and 48h after a blood meal, as determined by qRT-PCR. At 48h, midguts were pooled according to whether (+) or not (-) the blood bolus was present. Each dot represents a pool of 5 guts, derived from two independent experiments. Ratios are normalized within biological replicates to the mean of the 24h pools. Mean plus/minus standard error is indicated. Statistical significance was assessed by an ANOVA on a linear mixed effect regression model. ‘*’ p<0.05; ‘**’ p<0.01; ‘***’ p<0.001. <b>(B)</b> <i>Enterobacteriaceae</i> and <i>Acetobacteraceae</i> load in midguts 72h after feeding on blood supplemented with 100μM polyoxin D (PxD) or an equal volume of water (control), as determined by qRT-PCR. Each dot represents a pool of 8–10 guts, derived from 4 independent experiments. Ratios are normalized within biological replicates to the mean of the control pools. Mean plus/minus standard error is indicated. Statistical significance was assessed and is presented as described above. <b>(C)</b> Thin abdominal section 24h post blood meal stained with anti-LPS antibody. White arrowheads indicate LPS staining. <b>(D)</b> Gram-stained thin abdominal sections 24h post blood meal, with or without polyoxin D supplementation. Bacteria are stained light purple. “Lu” lumen; “Ep” epithelium.</p

    Effect of oral antibiotic treatment on the gene expression of the mosquito midgut.

    No full text
    <p><b>(A)</b> Bacterial load in the guts of 4 control and 4 antibiotic treated mosquito cohorts throughout a two blood meal (BM1 and BM2) time course assessed by qRT-PCR using universal 16S primers. Data were normalized within each biological replicate to the bacterial load in the control 96h sample. The mean plus/minus the standard error is shown. <b>(B)</b> Principal components analysis (PCA) plot of the 40 sequenced midgut samples after variance stabilizing transformation of count data. <b>(C)</b> Number of genes that were significantly upregulated (white bars) or downregulated (black bars) in each of the time points following antibiotic treatment. Genes showing an adjusted p-value <0.1 (Wald test with a Benjamini-Hochberg correction) were considered to be significantly regulated.</p
    corecore