Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
bioRxiv ; 2024 May 31.
Article in English | MEDLINE | ID: mdl-38853990

ABSTRACT

The malaria-causing parasites have to complete a complex infection cycle in the mosquito vector that also involves attack by the insect's innate immune system, especially at the early stages of midgut infection. However, Anopheles immunity to the late Plasmodium sporogonic stages, such as oocysts, has received little attention as they are considered to be concealed from immune factors due to their location under the midgut basal lamina and for harboring an elaborate cell wall comprising an external layer derived from the basal lamina that confers self-properties to an otherwise foreign structure. Here, we investigated whether Plasmodium berghei oocysts and sporozoites are susceptible to melanization-based immunity in Anopheles gambiae. Silencing of the negative regulator of melanization response, CLIPA14, increased melanization prevalence without significantly increasing the numbers of melanized oocysts, while co-silencing CLIPA14 with CLIPA2, a second negative regulator of melanization, resulted in a significant increase in melanized oocysts and melanization prevalence. Only late-stage oocysts were found to be melanized, suggesting that oocyst rupture was a prerequisite for melanization-based immune attack, presumably due to the loss of the immune-evasive features of their wall. We also found melanized sporozoites inside oocysts and in the hemocoel, suggesting that sporozoites at different maturation stages are susceptible to melanization. Silencing the melanization promoting factors TEP1 and CLIPA28 rescued oocyst melanization in CLIPA2/CLIPA14 co-silenced mosquitoes. Interestingly, silencing of CTL4, that protects early stage ookinetes from melanization, had no effect on oocysts and sporozoites, indicating differential regulation of immunity to early and late sporogonic stages. Similar to previous studies addressing ookinete stage melanization, the melanization of Plasmodium falciparum oocysts was significantly lower than that observed for P. berghei. In summary, our results provide conclusive evidence that late sporogonic malaria parasite stages are susceptible to melanization, and we reveal distinct regulatory mechanisms for ookinete and oocyst melanization.

2.
Sci Rep ; 10(1): 3352, 2020 02 25.
Article in English | MEDLINE | ID: mdl-32099004

ABSTRACT

The midgut microbiota of disease vectors plays a critical role in the successful transmission of human pathogens. The environment influences the microbiota composition; however, the relative mosquito-species contribution has not been rigorously disentangled from the environmental contribution to the microbiota structure. Also, the extent to which the microbiota of the adult sugar food source and larval water can predict that of the adult midgut and vice versa is not fully understood. To address these relationships, larvae and adults of Anopheles gambiae and Aedes albopictus were either reared separately or in a co-rearing system, whereby aquatic and adult stages of both species shared the larval water and sugar food source, respectively. Despite being reared under identical conditions, clear intra- and interspecies differences in midgut microbiota-composition were observed across seven cohorts, collected at different time points over a period of eight months. Fitting a linear model separately for each OTU in the mosquito midgut showed that two OTUs significantly differed between the midguts of the two mosquito species. We also show an effect for the sugar food source and larval water on the adult midgut microbiota. Our findings suggest that the mosquito midgut microbiota is highly dynamic and controlled by multiple factors.


Subject(s)
Aedes/genetics , Anopheles/genetics , Bacteria/genetics , Gastrointestinal Microbiome/genetics , Aedes/microbiology , Animals , Anopheles/microbiology , Bacteria/classification , Coculture Techniques , Humans , Larva/genetics , Larva/microbiology , Mosquito Vectors/genetics , Mosquito Vectors/microbiology , RNA, Ribosomal, 16S/genetics
3.
PLoS Pathog ; 15(11): e1008194, 2019 11.
Article in English | MEDLINE | ID: mdl-31765430

ABSTRACT

Serine protease cascades regulate important insect immune responses namely melanization and Toll pathway activation. An important component of these cascades are clip-domain serine protease homologs (cSPHs), which are non-catalytic, but essential for activating the enzyme prophenoloxidase (PPO) in the melanization response during septic infections. The activation of cSPHs requires their proteolytic cleavage, yet factors that control their activation and the complexity of their interactions within these cascades remain unclear. Here, we report the identification of CLIPA28 as a novel immune-related cSPH in the malaria vector Anopheles gambiae. Functional genetic analysis using RNA interference (RNAi) revealed that CLIPA28 is essential for the melanization of Plasmodium berghei parasites in refractory mosquitoes, and for mosquito resistance to fungal infections. We further show, using combined biochemical and genetic approaches, that CLIPA28 is member of a network of at least four cSPHs, whereby members are activated in a hierarchical manner following septic infections. Depletion of the complement-like protein TEP1 abolished the activation of this network after septic infections, whereas, depletion of the serine protease inhibitor 2 (SRPN2) triggered enhanced network activation, even in naïve mosquitoes, culminating in a dramatic reduction in cSPHs hemolymph levels, which paralleled that of PPO. Our data suggest that cSPHs are engaged in complex and multilayered interactions within serine protease cascades that regulate melanization, and identify TEP1 and SRPN2 as two master regulators of the cSPH network.


Subject(s)
Anopheles/immunology , Immunity, Innate/immunology , Insect Proteins/immunology , Malaria/immunology , Melanins/immunology , Plasmodium berghei/immunology , Serine Proteases/immunology , Animals , Anopheles/metabolism , Female , Insect Proteins/metabolism , Malaria/metabolism , Malaria/parasitology , Melanins/metabolism , Serine Proteases/metabolism
4.
J Biol Chem ; 292(44): 18217-18226, 2017 11 03.
Article in English | MEDLINE | ID: mdl-28928218

ABSTRACT

Clip domain serine protease homologs (SPHs) are positive and negative regulators of Anopheles gambiae immune responses mediated by the complement-like protein TEP1 against Plasmodium malaria parasites and other microbial infections. We have previously reported that the SPH CLIPA2 is a negative regulator of the TEP1-mediated response by showing that CLIPA2 knockdown (kd) enhances mosquito resistance to infections with fungi, bacteria, and Plasmodium parasites. Here, we identify another SPH, CLIPA14, as a novel regulator of mosquito immunity. We found that CLIPA14 is a hemolymph protein that is rapidly cleaved following a systemic infection. CLIPA14 kd mosquitoes elicited a potent melanization response against Plasmodium berghei ookinetes and exhibited significantly increased resistance to Plasmodium infections as well as to systemic and oral bacterial infections. The activity of the enzyme phenoloxidase, which initiates melanin biosynthesis, dramatically increased in the hemolymph of CLIPA14 kd mosquitoes in response to systemic bacterial infections. Ookinete melanization and hemolymph phenoloxidase activity were further increased after cosilencing CLIPA14 and CLIPA2, suggesting that these two SPHs act in concert to control the melanization response. Interestingly, CLIPA14 RNAi phenotypes and its infection-induced cleavage were abolished in a TEP1 loss-of-function background. Our results suggest that a complex network of SPHs functions downstream of TEP1 to regulate the melanization reaction.


Subject(s)
Anopheles/metabolism , Hemolymph/metabolism , Immunity, Innate , Insect Proteins/metabolism , Serine Endopeptidases/metabolism , Animals , Animals, Genetically Modified , Anopheles/immunology , Anopheles/microbiology , Anopheles/parasitology , Enzyme Activation , Escherichia coli/growth & development , Escherichia coli/immunology , Escherichia coli/isolation & purification , Female , Gene Knockdown Techniques/veterinary , Hemolymph/immunology , Hemolymph/microbiology , Insect Proteins/antagonists & inhibitors , Insect Proteins/genetics , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Melanins/genetics , Melanins/metabolism , Monophenol Monooxygenase/genetics , Monophenol Monooxygenase/metabolism , Plasmodium berghei/growth & development , Plasmodium berghei/immunology , Plasmodium berghei/isolation & purification , Proteolysis , RNA Interference , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Serratia marcescens/growth & development , Serratia marcescens/immunology , Serratia marcescens/isolation & purification , Staphylococcus aureus/growth & development , Staphylococcus aureus/immunology , Staphylococcus aureus/isolation & purification , Survival Analysis , Up-Regulation
5.
J Innate Immun ; 8(3): 314-26, 2016.
Article in English | MEDLINE | ID: mdl-26950600

ABSTRACT

The complement-like protein thioester-containing protein 1 (TEP1) is the hallmark effector molecule against Plasmodium ookinetes in the malaria vector Anopheles gambiae. We have previously shown that the knockdown of the noncatalytic clip domain serine protease CLIPA2 increased TEP1-mediated killing rendering mosquitoes more resistant to Plasmodium, bacterial and fungal infections. Here, CLIPA2 coimmunoprecipitation from the hemolymph of Beauveria bassiana-infected mosquitoes followed by mass spectrometry and functional genetic analysis led to the identification of the Apolipophorin-II/I gene, encoding the two lipid carrier proteins Apo-I and II, as a novel negative regulator of TEP1-mediated immune response during mosquito systemic infections. Apo-II/I exhibits a similar RNAi phenotype as CLIPA2 in mosquito bioassays characterized by increased resistance to B. bassiana and Escherichia coli infections. We provide evidence that this enhanced resistance to systemic infections is TEP1 dependent. Interestingly, silencing Apo-II/I but not CLIPA2 upregulated the expression of TEP1 following systemic infections with E. coli and B. bassiana in a c-Jun N-terminal kinase pathway-dependent manner. Our results suggest that mosquito Apo-II/I plays an important immune regulatory role during systemic infections and provide novel insight into the functional interplay between lipid metabolism and immune gene regulation.


Subject(s)
Anopheles/immunology , Apolipoproteins/metabolism , Beauveria/immunology , Immunity, Innate , Insect Proteins/metabolism , Malaria/transmission , Mycoses/immunology , Animals , Apolipoproteins/genetics , Cells, Cultured , Cloning, Molecular , Complement System Proteins/metabolism , Disease Vectors , Insect Proteins/genetics , Plasmodium , RNA, Small Interfering/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...