Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
1.
Cell ; 177(2): 315-325.e14, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30929905

RESUMEN

Transmission of malaria parasites occurs when a female Anopheles mosquito feeds on an infected host to acquire nutrients for egg development. How parasites are affected by oogenetic processes, principally orchestrated by the steroid hormone 20-hydroxyecdysone (20E), remains largely unknown. Here we show that Plasmodium falciparum development is intimately but not competitively linked to processes shaping Anopheles gambiae reproduction. We unveil a 20E-mediated positive correlation between egg and oocyst numbers; impairing oogenesis by multiple 20E manipulations decreases parasite intensities. These manipulations, however, accelerate Plasmodium growth rates, allowing sporozoites to become infectious sooner. Parasites exploit mosquito lipids for faster growth, but they do so without further affecting egg development. These results suggest that P. falciparum has adopted a non-competitive evolutionary strategy of resource exploitation to optimize transmission while minimizing fitness costs to its mosquito vector. Our findings have profound implications for currently proposed control strategies aimed at suppressing mosquito populations.


Asunto(s)
Ecdisterona/metabolismo , Interacciones Huésped-Parásitos/fisiología , Malaria Falciparum/parasitología , Animales , Anopheles/parasitología , Culicidae , Ecdisterona/fisiología , Femenino , Células HEK293 , Humanos , Insectos Vectores , Malaria/parasitología , Ratones , Mosquitos Vectores , Células 3T3 NIH , Oogénesis/fisiología , Plasmodium/metabolismo , Plasmodium falciparum , Esporozoítos , Esteroides/metabolismo
2.
Proc Natl Acad Sci U S A ; 121(5): e2316304121, 2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-38261617

RESUMEN

The discovery that Africans were resistant to infection by Plasmodium vivax (P. vivax) led to the conclusion that P. vivax invasion relied on the P. vivax Duffy Binding Protein (PvDBP) interacting with the Duffy Antigen Receptor for Chemokines (DARC) expressed on erythrocytes. However, the recent reporting of P. vivax infections in DARC-negative Africans suggests that the parasite might use an alternate invasion pathway to infect DARC-negative reticulocytes. To identify the parasite ligands and erythrocyte receptors that enable P. vivax invasion of both DARC-positive and -negative erythrocytes, we expressed region II containing the Duffy Binding-Like (DBL) domain of P. vivax erythrocyte binding protein (PvEBP-RII) and verified that the DBL domain binds to both DARC-positive and -negative erythrocytes. Furthermore, an AVidity-based EXtracelluar Interaction Screening (AVEXIS) was used to identify the receptor for PvEBP among over 750 human cell surface receptor proteins, and this approach identified only Complement Receptor 1 (CR1, CD35, or C3b/C4b receptor) as a PvEBP receptor. CR1 is a well-known receptor for P. falciparum Reticulocyte binding protein Homology 4 (PfRh4) and is present on the surfaces of both reticulocytes and normocytes, but its expression decreases as erythrocytes age. Indeed, PvEBP-RII bound to a subpopulation of both reticulocytes and normocytes, and this binding was blocked by the addition of soluble CR1 recombinant protein, indicating that CR1 is the receptor of PvEBP. In addition, we found that the Long Homology Repeat A (LHR-A) subdomain of CR1 is the only subdomain responsible for mediating the interaction with PvEBP-RII.


Asunto(s)
Malaria Falciparum , Plasmodium vivax , Humanos , Receptores de Superficie Celular , Eritrocitos , Reticulocitos , Antígenos CD2 , Moléculas de Adhesión Celular
3.
Proc Natl Acad Sci U S A ; 121(23): e2403796121, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38809710

RESUMEN

Olfactory receptors (Olfr) are G protein-coupled receptors that are normally expressed on olfactory sensory neurons to detect volatile chemicals or odorants. Interestingly, many Olfrs are also expressed in diverse tissues and function in cell-cell recognition, migration, and proliferation as well as immune responses and disease processes. Here, we showed that many Olfr genes were expressed in the mouse spleen, linked to Plasmodium yoelii genetic loci significantly, and/or had genome-wide patterns of LOD scores (GPLSs) similar to those of host Toll-like receptor genes. Expression of specific Olfr genes such as Olfr1386 in HEK293T cells significantly increased luciferase signals driven by IFN-ß and NF-κB promoters, with elevated levels of phosphorylated TBK1, IRF3, P38, and JNK. Mice without Olfr1386 were generated using the CRISPR/Cas9 method, and the Olfr1386-/- mice showed significantly lower IFN-α/ß levels and longer survival than wild-type (WT) littermates after infection with P. yoelii YM parasites. Inhibition of G protein signaling and P38 activity could affect cyclic AMP-responsive element promoter-driven luciferase signals and IFN-ß mRNA levels in HEK293T cells expressing the Olfr1386 gene, respectively. Screening of malaria parasite metabolites identified nicotinamide adenine dinucleotide (NAD) as a potential ligand for Olfr1386, and NAD could stimulate IFN-ß responses and phosphorylation of TBK1 and STAT1/2 in RAW264.7 cells. Additionally, parasite RNA (pRNA) could significantly increase Olfr1386 mRNA levels. This study links multiple Olfrs to host immune response pathways, identifies a candidate ligand for Olfr1386, and demonstrates the important roles of Olfr1386 in regulating type I interferon (IFN-I) responses during malaria parasite infections.


Asunto(s)
Interferón Tipo I , Malaria , Plasmodium yoelii , Receptores Odorantes , Animales , Ratones , Malaria/inmunología , Malaria/parasitología , Malaria/metabolismo , Humanos , Células HEK293 , Receptores Odorantes/genética , Receptores Odorantes/metabolismo , Interferón Tipo I/metabolismo , Interferón Tipo I/inmunología , Ratones Noqueados , Transducción de Señal , Ratones Endogámicos C57BL
4.
Proc Natl Acad Sci U S A ; 120(1): e2215003120, 2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36577076

RESUMEN

We used a transgenic parasite in which Plasmodium falciparum parasites were genetically modified to express Plasmodium vivax apical membrane antigen 1 (PvAMA1) protein in place of PfAMA1 to study PvAMA1-mediated invasion. In P. falciparum, AMA1 interaction with rhoptry neck protein 2 (RON2) is known to be crucial for invasion, and PfRON2 peptides (PfRON2p) blocked the invasion of PfAMA1 wild-type parasites. However, PfRON2p has no effect on the invasion of transgenic parasites expressing PvAMA1 indicating that PfRON2 had no role in the invasion of PvAMA1 transgenic parasites. Interestingly, PvRON2p blocked the invasion of PvAMA1 transgenic parasites in a dose-dependent manner. We found that recombinant PvAMA1 domains 1 and 2 (rPvAMA1) bound to reticulocytes and normocytes indicating that PvAMA1 directly interacts with erythrocytes during the invasion, and invasion blocking of PvRON2p may result from it interfering with PvAMA1 binding to erythrocytes. It was previously shown that the peptide containing Loop1a of PvAMA1 (PvAMA1 Loop1a) is also bound to reticulocytes. We found that the Loop1a peptide blocked the binding of PvAMA1 to erythrocytes. PvAMA1 Loop1a has no polymorphisms in contrast to other PvAMA1 loops and may be an attractive vaccine target. We thus present the evidence that PvAMA1 binds to erythrocytes in addition to interacting with PvRON2 suggesting that the P. vivax merozoites may exploit complex pathways during the invasion process.


Asunto(s)
Malaria Falciparum , Plasmodium vivax , Humanos , Proteínas Protozoarias/química , Antígenos de Protozoos , Eritrocitos/metabolismo , Plasmodium falciparum/metabolismo , Reticulocitos/metabolismo
5.
PLoS Pathog ; 19(3): e1010538, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36862755

RESUMEN

Malaria is caused by the unicellular parasite Plasmodium which is transmitted to humans through the bite of infected female Anopheles mosquitoes. To initiate sexual reproduction and to infect the midgut of the mosquito, Plasmodium gametocytes are able to recognize the intestinal environment after being ingested during blood feeding. A shift in temperature, pH change and the presence of the insect-specific compound xanthurenic acid have been shown to be important stimuli perceived by gametocytes to become activated and proceed to sexual reproduction. Here we report that the salivary protein Saglin, previously proposed to be a receptor for the recognition of salivary glands by sporozoites, facilitates Plasmodium colonization of the mosquito midgut, but does not contribute to salivary gland invasion. In mosquito mutants lacking Saglin, Plasmodium infection of Anopheles females is reduced, resulting in impaired transmission of sporozoites at low infection densities. Interestingly, Saglin can be detected in high amounts in the midgut of mosquitoes after blood ingestion, possibly indicating a previously unknown host-pathogen interaction between Saglin and midgut stages of Plasmodium. Furthermore, we were able to show that saglin deletion has no fitness cost in laboratory conditions, suggesting this gene would be an interesting target for gene drive approaches.


Asunto(s)
Anopheles , Malaria , Parásitos , Plasmodium , Animales , Humanos , Femenino , Anopheles/parasitología , Mosquitos Vectores , Malaria/parasitología , Esporozoítos , Proteínas y Péptidos Salivales
6.
Proc Natl Acad Sci U S A ; 118(22)2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34035177

RESUMEN

Cytoadhesion of Plasmodium falciparum-infected erythrocytes (IEs) to the endothelial lining of blood vessels protects parasites from splenic destruction, but also leads to detrimental inflammation and vessel occlusion. Surface display of the P. falciparum erythrocyte membrane protein 1 (PfEMP1) adhesion ligands exposes them to host antibodies and serum proteins. PfEMP1 are important targets of acquired immunity to malaria, and through evolution, the protein family has expanded and diversified to bind a select set of host receptors through antigenically diversified receptor-binding domains. Here, we show that complement component 1s (C1s) in serum cleaves PfEMP1 at semiconserved arginine motifs located at interdomain regions between the receptor-binding domains, rendering the IE incapable of binding the two main PfEMP1 receptors, CD36 and endothelial protein C receptor (EPCR). Bioinformatic analyses of PfEMP1 protein sequences from 15 P. falciparum genomes found the C1s motif was present in most PfEMP1 variants. Prediction of C1s cleavage and loss of binding to endothelial receptors was further corroborated by testing of several different parasite lines. These observations suggest that the parasites have maintained susceptibility for cleavage by the serine protease, C1s, and provides evidence for a complex relationship between the complement system and the P. falciparum cytoadhesion virulence determinant.


Asunto(s)
Adhesión Bacteriana , Complemento C1/metabolismo , Plasmodium falciparum/fisiología , Proteínas Protozoarias/metabolismo , Secuencia de Aminoácidos , Línea Celular , Secuencia Conservada , Humanos
7.
J Biol Chem ; 298(6): 101971, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35460690

RESUMEN

To successfully feed on blood, hematophagous arthropods must combat the host's natural hemostatic and inflammatory responses. Salivary proteins of blood-feeding insects such as mosquitoes contain compounds that inhibit these common host defenses against blood loss, including vasoconstriction, platelet aggregation, blood clotting, pain, and itching. The D7 proteins are some of the most abundantly expressed proteins in female mosquito salivary glands and have been implicated in inhibiting host hemostatic and inflammatory responses. Anopheles gambiae, the primary vector of malaria, expresses three D7 long-form and five D7 short-form proteins. Previous studies have characterized the AngaD7 short-forms, but the D7 long-form proteins have not yet been characterized in detail. Here, we characterized the A. gambiae D7 long-forms by first determining their binding kinetics to hemostatic agonists such as leukotrienes and serotonin, which are potent activators of vasoconstriction, edema formation, and postcapillary venule leakage, followed by ex vivo functional assays. We found that AngaD7L1 binds leukotriene C4 and thromboxane A2 analog U-46619; AngaD7L2 weakly binds leukotrienes B4 and D4; and AngaD7L3 binds serotonin. Subsequent functional assays confirmed AngaD7L1 inhibits U-46619-induced platelet aggregation and vasoconstriction, and AngaD7L3 inhibits serotonin-induced platelet aggregation and vasoconstriction. It is therefore possible that AngaD7L proteins counteract host hemostasis by scavenging these mediators. Finally, we demonstrate that AngaD7L2 had a dose-dependent anticoagulant effect via the intrinsic coagulation pathway by interacting with factors XII, XIIa, and XI. The uncovering of these interactions in the present study will be essential for comprehensive understanding of the vector-host biochemical interface.


Asunto(s)
Anopheles , Hemostáticos , Proteínas de Insectos/metabolismo , Proteínas y Péptidos Salivales/metabolismo , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico , Animales , Anopheles/química , Femenino , Hemostáticos/metabolismo , Leucotrienos/metabolismo , Malaria , Mosquitos Vectores , Serotonina/metabolismo , Serotonina/farmacología
8.
BMC Genomics ; 24(1): 135, 2023 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-36941562

RESUMEN

INTRODUCTION: During evolution, blood-feeding arthropods developed a complex salivary mixture that can interfere with host haemostatic and immune response, favoring blood acquisition and pathogen transmission. Therefore, a survey of the salivary gland contents can lead to the identification of molecules with potent pharmacological activity in addition to increase our understanding of the molecular mechanisms underlying the hematophagic behaviour of arthropods. The southern house mosquito, Culex quinquefasciatus, is a vector of several pathogenic agents, including viruses and filarial parasites that can affect humans and wild animals. RESULTS: Previously, a Sanger-based transcriptome of the salivary glands (sialome) of adult C. quinquefasciatus females was published based on the sequencing of 503 clones organized into 281 clusters. Here, we revisited the southern mosquito sialome using an Illumina-based RNA-sequencing approach of both male and female salivary glands. Our analysis resulted in the identification of 7,539 coding DNA sequences (CDS) that were functionally annotated into 25 classes, in addition to 159 long non-coding RNA (LncRNA). Additionally, comparison of male and female libraries allowed the identification of female-enriched transcripts that are potentially related to blood acquisition and/or pathogen transmission. CONCLUSION: Together, these findings represent an extended reference for the identification and characterization of the proteins containing relevant pharmacological activity in the salivary glands of C. quinquefasciatus mosquitoes.


Asunto(s)
Culex , Culicidae , Humanos , Animales , Masculino , Femenino , Culex/genética , Culex/metabolismo , Culicidae/genética , Mosquitos Vectores/genética , Proteínas/metabolismo , Transcriptoma
9.
Proc Natl Acad Sci U S A ; 117(5): 2597-2605, 2020 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-31969456

RESUMEN

The surface protein Pfs47 allows Plasmodium falciparum parasites to survive and be transmitted by making them "undetectable" to the mosquito immune system. P. falciparum parasites express Pfs47 haplotypes compatible with their sympatric vectors, while those with incompatible haplotypes are eliminated by the mosquito. We proposed that Pfs47 serves as a "key" that mediates immune evasion by interacting with a mosquito receptor "the lock," which differs in evolutionarily divergent anopheline mosquitoes. Recombinant Pfs47 (rPfs47) was used to identify the mosquito Pfs47 receptor protein (P47Rec) using far-Western analysis. rPfs47 bound to a single 31-kDa band and the identity of this protein was determined by mass spectrometry. The mosquito P47Rec has two natterin-like domains and binds to Pfs47 with high affinity (17 to 32 nM). P47Rec is a highly conserved protein with submicrovillar localization in midgut cells. It has structural homology to a cytoskeleton-interacting protein and accumulates at the site of ookinete invasion. Silencing P47Rec expression reduced P. falciparum infection, indicating that the interaction of Pfs47 with the receptor is critical for parasite survival. The binding specificity of P47Rec from distant anophelines (Anopheles gambiae, Anopheles dirus, and Anopheles albimanus) with Pfs47-Africa (GB4) and Pfs47-South America (7G8) haplotypes was evaluated, and it is in agreement with the previously documented compatibility between P. falciparum parasites expressing different Pfs47 haplotypes and these three anopheline species. Our findings give further support to the role of Pfs47 in the adaptation of P. falciparum to different vectors.


Asunto(s)
Anopheles/inmunología , Anopheles/parasitología , Proteínas de Insectos/inmunología , Glicoproteínas de Membrana/inmunología , Mosquitos Vectores/inmunología , Mosquitos Vectores/parasitología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Animales , Anopheles/genética , Interacciones Huésped-Parásitos , Evasión Inmune , Proteínas de Insectos/genética , Cinética , Glicoproteínas de Membrana/genética , Mosquitos Vectores/genética , Plasmodium falciparum/genética , Proteínas Protozoarias/genética
10.
PLoS Pathog ; 16(1): e1008288, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31961911

RESUMEN

Insects rely on the innate immune system for defense against pathogens, some aspects of which are under hormonal control. Here we provide direct experimental evidence showing that the juvenile hormone-binding protein (mJHBP) of Aedes aegypti is required for the regulation of innate immune responses and the development of mosquito blood cells (hemocytes). Using an mJHBP-deficient mosquito line generated by means of CRISPR-Cas9 gene editing technology we uncovered a mutant phenotype characterized by immunosuppression at the humoral and cellular levels, which profoundly affected susceptibility to bacterial infection. Bacteria-challenged mosquitoes exhibited significantly higher levels of septicemia and mortality relative to the wild type (WT) strain, delayed expression of antimicrobial peptides (AMPs), severe developmental dysregulation of embryonic and larval hemocytes (reduction in the total number of hemocytes) and increased differentiation of the granulocyte lineage. Interestingly, injection of recombinant wild type mJHBP protein into adult females three-days before infection was sufficient to restore normal immune function. Similarly, injection of mJHBP into fourth-instar larvae fully restored normal larval/pupal hemocyte populations in emerging adults. More importantly, the recovery of normal immuno-activation and hemocyte development requires the capability of mJHBP to bind JH III. These results strongly suggest that JH III functions in mosquito immunity and hemocyte development in a manner that is perhaps independent of canonical JH signaling, given the lack of developmental and reproductive abnormalities. Because of the prominent role of hemocytes as regulators of mosquito immunity, this novel discovery may have broader implications for the understanding of vector endocrinology, hemocyte development, vector competence and disease transmission.


Asunto(s)
Aedes/crecimiento & desarrollo , Aedes/inmunología , Proteínas Portadoras/inmunología , Proteínas de Insectos/inmunología , Aedes/genética , Aedes/microbiología , Animales , Proteínas Portadoras/genética , Femenino , Hemocitos/inmunología , Hemocitos/microbiología , Inmunidad Innata , Proteínas de Insectos/genética , Hormonas Juveniles/inmunología , Larva/genética , Larva/crecimiento & desarrollo , Larva/inmunología , Larva/microbiología , Masculino , Serratia marcescens/fisiología
11.
Malar J ; 20(1): 11, 2021 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-33407511

RESUMEN

BACKGROUND: The invasion of the mosquito salivary glands by Plasmodium sporozoites is a critical step that defines the success of malaria transmission and a detailed understanding of the molecules responsible for salivary gland invasion could be leveraged towards control of vector-borne pathogens. Antibodies directed against the mosquito salivary gland protein SGS1 have been shown to reduce Plasmodium gallinaceum sporozoite invasion of Aedes aegypti salivary glands, but the specific role of this protein in sporozoite invasion and in other stages of the Plasmodium life cycle remains unknown. METHODS: RNA interference and CRISPR/Cas9 were used to evaluate the role of A. aegypti SGS1 in the P. gallinaceum life cycle. RESULTS: Knockdown and knockout of SGS1 disrupted sporozoite invasion of the salivary gland. Interestingly, mosquitoes lacking SGS1 also displayed fewer oocysts. Proteomic analyses confirmed the abolishment of SGS1 in the salivary gland of SGS1 knockout mosquitoes and revealed that the C-terminus of the protein is absent in the salivary gland of control mosquitoes. In silico analyses indicated that SGS1 contains two potential internal cleavage sites and thus might generate three proteins. CONCLUSION: SGS1 facilitates, but is not essential for, invasion of A. aegypti salivary glands by P. gallinaceum and has a dual role as a facilitator of parasite development in the mosquito midgut. SGS1 could, therefore, be part of a strategy to decrease malaria transmission by the mosquito vector, for example in a transgenic mosquito that blocks its interaction with the parasite.


Asunto(s)
Aedes/genética , Proteínas de Insectos/genética , Plasmodium gallinaceum/fisiología , Proteínas y Péptidos Salivales/genética , Aedes/parasitología , Secuencia de Aminoácidos , Animales , Femenino , Tracto Gastrointestinal/parasitología , Proteínas de Insectos/química , Proteínas de Insectos/metabolismo , Mosquitos Vectores/genética , Mosquitos Vectores/parasitología , Glándulas Salivales/parasitología , Proteínas y Péptidos Salivales/química , Proteínas y Péptidos Salivales/metabolismo , Alineación de Secuencia , Esporozoítos/fisiología
12.
Int J Mol Sci ; 22(17)2021 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-34502092

RESUMEN

Arthropod-borne viruses, referred to collectively as arboviruses, infect millions of people worldwide each year and have the potential to cause severe disease. They are predominately transmitted to humans through blood-feeding behavior of three main groups of biting arthropods: ticks, mosquitoes, and sandflies. The pathogens harbored by these blood-feeding arthropods (BFA) are transferred to animal hosts through deposition of virus-rich saliva into the skin. Sometimes these infections become systemic and can lead to neuro-invasion and life-threatening viral encephalitis. Factors intrinsic to the arboviral vectors can greatly influence the pathogenicity and virulence of infections, with mounting evidence that BFA saliva and salivary proteins can shift the trajectory of viral infection in the host. This review provides an overview of arbovirus infection and ways in which vectors influence viral pathogenesis. In particular, we focus on how saliva and salivary gland extracts from the three dominant arbovirus vectors impact the trajectory of the cellular immune response to arbovirus infection in the skin.


Asunto(s)
Infecciones por Arbovirus/transmisión , Arbovirus/patogenicidad , Vectores Artrópodos/virología , Saliva/virología , Animales , Vectores Artrópodos/fisiología , Interacciones Huésped-Patógeno , Humanos , Saliva/metabolismo
13.
Int J Mol Sci ; 22(23)2021 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-34884537

RESUMEN

The PIWI-interacting RNA (piRNA) pathway provides an RNA interference (RNAi) mechanism known from Drosophila studies to maintain the integrity of the germline genome by silencing transposable elements (TE). Aedes aegypti mosquitoes, which are the key vectors of several arthropod-borne viruses, exhibit an expanded repertoire of Piwi proteins involved in the piRNA pathway, suggesting functional divergence. Here, we investigate RNA-binding dynamics and subcellular localization of A. aegypti Piwi4 (AePiwi4), a Piwi protein involved in antiviral immunity and embryonic development, to better understand its function. We found that AePiwi4 PAZ (Piwi/Argonaute/Zwille), the domain that binds the 3' ends of piRNAs, bound to mature (3' 2' O-methylated) and unmethylated RNAs with similar micromolar affinities (KD = 1.7 ± 0.8 µM and KD of 5.0 ± 2.2 µM, respectively; p = 0.05) in a sequence independent manner. Through site-directed mutagenesis studies, we identified highly conserved residues involved in RNA binding and found that subtle changes in the amino acids flanking the binding pocket across PAZ proteins have significant impacts on binding behaviors, likely by impacting the protein secondary structure. We also analyzed AePiwi4 subcellular localization in mosquito tissues. We found that the protein is both cytoplasmic and nuclear, and we identified an AePiwi4 nuclear localization signal (NLS) in the N-terminal region of the protein. Taken together, these studies provide insights on the dynamic role of AePiwi4 in RNAi and pave the way for future studies aimed at understanding Piwi interactions with diverse RNA populations.


Asunto(s)
Proteínas Argonautas/química , Proteínas Argonautas/metabolismo , Núcleo Celular/metabolismo , Elementos Transponibles de ADN , Proteínas de Insectos/química , Proteínas de Insectos/metabolismo , ARN Interferente Pequeño/metabolismo , Aedes , Secuencia de Aminoácidos , Animales , Proteínas Argonautas/genética , Núcleo Celular/genética , Proteínas de Insectos/genética , Mosquitos Vectores , Conformación Proteica , ARN Interferente Pequeño/genética , Homología de Secuencia
14.
PLoS Pathog ; 14(5): e1007006, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29723281

RESUMEN

Salivary components from disease vectors help arthropods to acquire blood and have been shown to enhance pathogen transmission in different model systems. Here we show that two salivary enzymes from Lutzomyia longipalpis have a synergist effect that facilitates a more efficient blood meal intake and diffusion of other sialome components. We have previously shown that Lundep, a highly active endonuclease, enhances parasite infection and prevent blood clotting by inhibiting the intrinsic pathway of coagulation. To investigate the physiological role of a salivary hyaluronidase in blood feeding we cloned and expressed a recombinant hyaluronidase from Lu. longipalpis. Recombinant hyaluronidase (LuloHya) was expressed in mammalian cells and biochemically characterized in vitro. Our study showed that expression of neutrophil CXC chemokines and colony stimulating factors were upregulated in HMVEC cells after incubation with LuloHya and Lundep. These results were confirmed by the acute hemorrhage, edema and inflammation in a dermal necrosis (dermonecrotic) assay involving a massive infiltration of leukocytes, especially neutrophils, in mice co-injected with hemorrhagic factor and these two salivary proteins. Moreover, flow cytometry results showed that LuloHya and Lundep promote neutrophil recruitment to the bite site that may serve as a vehicle for establishment of Leishmania infection. A vaccination experiment demonstrated that LuloHya and Lundep confer protective immunity against cutaneous leishmaniasis using the Lu. longipalpis-Leishmania major combination as a model. Animals (C57BL/6) immunized with LuloHya or Lundep showed minimal skin damage while lesions in control animals remained ulcerated. This protective immunity was abrogated when B-cell-deficient mice were used indicating that antibodies against both proteins play a significant role for disease protection. Rabbit-raised anti-LuloHya antibodies completely abrogated hyaluronidase activity in vitro. Moreover, in vivo experiments demonstrated that blocking LuloHya with specific antibodies interferes with sand fly blood feeding. This work highlights the relevance of vector salivary components in blood feeding and parasite transmission and further suggests the inclusion of these salivary proteins as components for an anti-Leishmania vaccine.


Asunto(s)
Hialuronoglucosaminidasa/inmunología , Leishmania major/inmunología , Leishmania major/patogenicidad , Leishmaniasis Cutánea/inmunología , Leishmaniasis Cutánea/prevención & control , Psychodidae/inmunología , Animales , Simulación por Computador , Endonucleasas/inmunología , Femenino , Interacciones Huésped-Patógeno/inmunología , Humanos , Hialuronoglucosaminidasa/química , Proteínas de Insectos/química , Proteínas de Insectos/inmunología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Modelos Moleculares , Neutrófilos/inmunología , Polisacárido Liasas/inmunología , Conejos , Saliva/enzimología , Saliva/inmunología
15.
Cell Mol Life Sci ; 76(10): 2003-2013, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30747251

RESUMEN

To successfully feed, ticks inject pharmacoactive molecules into the vertebrate host including cystatin cysteine protease inhibitors. However, the molecular and cellular events modulated by tick saliva remain largely unknown. Here, we describe and characterize a novel immunomodulatory cystatin, Iristatin, which is upregulated in the salivary glands of feeding Ixodes ricinus ticks. We present the crystal structure of Iristatin at 1.76 Å resolution. Purified recombinant Iristatin inhibited the proteolytic activity of cathepsins L and C and diminished IL-2, IL-4, IL-9, and IFN-γ production by different T-cell populations, IL-6 and IL-9 production by mast cells, and nitric oxide production by macrophages. Furthermore, Iristatin inhibited OVA antigen-induced CD4+ T-cell proliferation and leukocyte recruitment in vivo and in vitro. Our results indicate that Iristatin affects wide range of anti-tick immune responses in the vertebrate host and may be exploitable as an immunotherapeutic.


Asunto(s)
Proteínas de Artrópodos/farmacología , Cistatinas/farmacología , Inmunosupresores/farmacología , Cistatinas Salivales/farmacología , Secuencia de Aminoácidos , Animales , Proteínas de Artrópodos/química , Proteínas de Artrópodos/genética , Cristalografía por Rayos X , Cistatinas/clasificación , Cistatinas/genética , Citocinas/metabolismo , Compuestos Epoxi/metabolismo , Femenino , Inmunosupresores/química , Inmunosupresores/metabolismo , Ixodes/química , Ixodes/genética , Ixodes/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Óxido Nítrico/metabolismo , Filogenia , Proteolisis/efectos de los fármacos , Cistatinas Salivales/química , Cistatinas Salivales/genética , Homología de Secuencia de Aminoácido , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo
16.
BMC Genomics ; 20(1): 166, 2019 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-30832587

RESUMEN

BACKGROUND: Saliva of mosquitoes contains anti-platelet, anti-clotting, vasodilatory, anti-complement and anti-inflammatory substances that help the blood feeding process. The salivary polypeptides are at a fast pace of evolution possibly due to their relative lack of structural constraint and possibly also by positive selection on their genes leading to evasion of host immune pressure. RESULTS: In this study, we used deep mRNA sequence to uncover for the first time the sialomes of four Amazonian anophelines species (Anopheles braziliensis, A. marajorara, A. nuneztovari and A. triannulatus) and extend the knowledge of the A. darlingi sialome. Two libraries were generated from A. darlingi mosquitoes, sampled from two localities separated ~ 1100 km apart. A total of 60,016 sequences were submitted to GenBank, which will help discovery of novel pharmacologically active polypeptides and the design of specific immunological markers of mosquito exposure. Additionally, in these analyses we identified and characterized novel phasmaviruses and anpheviruses associated to the sialomes of A. triannulatus, A. marajorara and A. darlingi species. CONCLUSIONS: Besides their pharmacological properties, which may be exploited for the development of new drugs (e.g. anti-thrombotics), salivary proteins of blood feeding arthropods may be turned into tools to prevent and/or better control vector borne diseases; for example, through the development of vaccines or biomarkers to evaluate human exposure to vector bites. The sialotranscriptome study reported here provided novel data on four New World anopheline species and allowed to extend our knowledge on the salivary repertoire of A. darlingi. Additionally, we discovered novel viruses following analysis of the transcriptomes, a procedure that should become standard within future RNAseq studies.


Asunto(s)
Anopheles/genética , Péptidos/genética , Saliva/química , Proteínas y Péptidos Salivales/genética , Secuencia de Aminoácidos/genética , Animales , Anopheles/química , Brasil , Humanos , Insectos Vectores/química , Insectos Vectores/genética , Mosquitos Vectores/genética , Ácido N-Acetilneuramínico/química , Péptidos/química , ARN Mensajero/genética , Proteínas y Péptidos Salivales/química , Selección Genética/genética
17.
Proc Natl Acad Sci U S A ; 111(19): 6946-51, 2014 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-24778255

RESUMEN

Mosquito salivary glands have important roles in blood feeding and pathogen transmission. However, the biological relevance of many salivary components has yet to be determined. Aegyptin, a secreted salivary protein from Aedes aegypti, binds collagen and inhibits platelet aggregation and adhesion. We used a transgenic approach to study the relevance of Aegyptin in mosquito blood feeding. Aedes aegypti manipulated genetically to express gene-specific inverted-repeat RNA sequences exhibited significant reductions in Aegyptin mRNA accumulation (85-87%) and protein levels (>80-fold) in female mosquito salivary glands. Transgenic mosquitoes had longer probing times (78-300 s, P < 0.0001) when feeding on mice compared with controls (15-56 s), feeding success was reduced, and those feeding took smaller blood meals. However, no differences in feeding success or blood meal size were found in membrane feeding experiments using defibrinated human blood. Salivary gland extracts from transgenic mosquitoes failed to inhibit collagen-induced platelet aggregation in vitro. Reductions of Aegyptin did not affect salivary ADP-induced platelet aggregation inhibition or disturb anticlotting activities. Our results demonstrate the relevance of Aegyptin for A. aegypti blood feeding, providing further support for the hypothesis that platelet aggregation inhibition is a vital salivary function in blood feeding arthropods. It has been suggested that the multiple mosquito salivary components mediating platelet aggregation (i.e., Aegyptin, apyrase, D7) represent functional redundancy. Our findings do not support this hypothesis; instead, they indicate that multiple salivary components work synergistically and are necessary to achieve maximum blood feeding efficiency.


Asunto(s)
Aedes/genética , Aedes/virología , Virus del Dengue/genética , Conducta Alimentaria , Proteínas de Insectos/genética , Proteínas y Péptidos Salivales/genética , Aedes/fisiología , Animales , Animales Modificados Genéticamente , Especificidad de Anticuerpos , Secuencia de Bases , Coagulación Sanguínea/fisiología , Proteínas Sanguíneas/fisiología , Colágeno/metabolismo , Femenino , Silenciador del Gen , Humanos , Proteínas de Insectos/metabolismo , Masculino , Ratones , Datos de Secuencia Molecular , Agregación Plaquetaria/fisiología , Conejos , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Saliva/metabolismo , Proteínas y Péptidos Salivales/metabolismo
18.
Infect Immun ; 84(6): 1796-1805, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27045038

RESUMEN

Tick saliva contains a number of effector molecules that inhibit host immunity and facilitate pathogen transmission. How tick proteins regulate immune signaling, however, is incompletely understood. Here, we describe that loop 2 of sialostatin L2, an anti-inflammatory tick protein, binds to annexin A2 and impairs the formation of the NLRC4 inflammasome during infection with the rickettsial agent Anaplasma phagocytophilum Macrophages deficient in annexin A2 secreted significantly smaller amounts of interleukin-1ß (IL-1ß) and IL-18 and had a defect in NLRC4 inflammasome oligomerization and caspase-1 activation. Accordingly, Annexin a2-deficient mice were more susceptible to A. phagocytophilum infection and showed splenomegaly, thrombocytopenia, and monocytopenia. Providing translational support to our findings, better binding of annexin A2 to sialostatin L2 in sera from 21 out of 23 infected patients than in sera from control individuals was also demonstrated. Overall, we establish a unique mode of inflammasome evasion by a pathogen, centered on a blood-feeding arthropod.


Asunto(s)
Anaplasma phagocytophilum/inmunología , Anexina A2/inmunología , Proteínas Reguladoras de la Apoptosis/inmunología , Proteínas de Unión al Calcio/inmunología , Cistatinas/inmunología , Ehrlichiosis/microbiología , Evasión Inmune , Secuencia de Aminoácidos , Anaplasma phagocytophilum/genética , Animales , Anexina A2/química , Anexina A2/genética , Proteínas Reguladoras de la Apoptosis/química , Proteínas Reguladoras de la Apoptosis/genética , Vectores Arácnidos/química , Vectores Arácnidos/genética , Vectores Arácnidos/inmunología , Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/genética , Caspasa 1/genética , Caspasa 1/inmunología , Caspasas/genética , Caspasas/inmunología , Caspasas Iniciadoras , Cistatinas/química , Cistatinas/genética , Ehrlichiosis/inmunología , Ehrlichiosis/patología , Escherichia coli/genética , Escherichia coli/metabolismo , Regulación de la Expresión Génica , Humanos , Inflamasomas/genética , Inflamasomas/inmunología , Interleucina-18/genética , Interleucina-18/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Ixodes/química , Ixodes/genética , Ixodes/inmunología , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Modelos Moleculares , Unión Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Transducción de Señal
19.
PLoS Pathog ; 10(2): e1003923, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24516388

RESUMEN

Neutrophils are the host's first line of defense against infections, and their extracellular traps (NET) were recently shown to kill Leishmania parasites. Here we report a NET-destroying molecule (Lundep) from the salivary glands of Lutzomyia longipalpis. Previous analysis of the sialotranscriptome of Lu. longipalpis showed the potential presence of an endonuclease. Indeed, not only was the cloned cDNA (Lundep) shown to encode a highly active ss- and dsDNAse, but also the same activity was demonstrated to be secreted by salivary glands of female Lu. longipalpis. Lundep hydrolyzes both ss- and dsDNA with little sequence specificity with a calculated DNase activity of 300000 Kunitz units per mg of protein. Disruption of PMA (phorbol 12 myristate 13 acetate)- or parasite-induced NETs by treatment with recombinant Lundep or salivary gland homogenates increases parasite survival in neutrophils. Furthermore, co-injection of recombinant Lundep with metacyclic promastigotes significantly exacerbates Leishmania infection in mice when compared with PBS alone or inactive (mutagenized) Lundep. We hypothesize that Lundep helps the parasite to establish an infection by allowing it to escape from the leishmanicidal activity of NETs early after inoculation. Lundep may also assist blood meal intake by lowering the local viscosity caused by the release of host DNA and as an anticoagulant by inhibiting the intrinsic pathway of coagulation.


Asunto(s)
Endonucleasas/metabolismo , Interacciones Huésped-Parásitos/fisiología , Leishmaniasis/enzimología , Psychodidae/enzimología , Psychodidae/parasitología , Secuencia de Aminoácidos , Animales , Coagulación Sanguínea/fisiología , Western Blotting , Vectores de Enfermedades , Endonucleasas/inmunología , Factor XIIa/metabolismo , Humanos , Leishmania , Leishmaniasis/inmunología , Ratones , Datos de Secuencia Molecular , Neutrófilos/inmunología , Neutrófilos/parasitología , Reacción en Cadena de la Polimerasa , Psychodidae/inmunología , Glándulas Salivales/enzimología , Glándulas Salivales/inmunología
20.
Blood ; 119(12): 2799-809, 2012 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-22302738

RESUMEN

CD300a is an immunoreceptor tyrosine-based inhibitory motif (ITIM) containing molecule that belongs to the CD300 family of paired activating/inhibitory receptors. It has been shown that its ligation inhibits activation signals on cells of both myeloid and lymphoid lineages. The ligands for CD300a have not been identified. Here, we show that a CD300a-Ig fusion protein specifically binds to apoptotic cells that are evolutionary apart, such as human and insect cells, suggesting that the ligand has to be conserved. Using surface plasmon resonance, ultracentrifugation, ELISA, and reporter cell assays, we identified phosphatidylethanolamine (PE) and phosphatidylserine (PS), 2 phospholipids that translocate to the outer leaflet of the plasma membrane of dead cells, as the ligands for CD300a. Mutational and structural modeling studies identified residues that are involved in the binding of CD300a to PE and PS and that form a cavity where the hydrophilic heads of PE and PS, can penetrate. CD300a down-regulates the uptake of apoptotic cells by macrophages and its ectopic expression in CD300a-negative cell lines also decreased the engulfment of dead cells. Collectively, our results indicate that PE and PS are ligands for CD300a, and that this interaction plays an important role in regulating the removal of dead cells.


Asunto(s)
Antígenos CD/química , Antígenos CD/metabolismo , Fagocitosis/fisiología , Fosfatidiletanolaminas/metabolismo , Fosfatidilserinas/metabolismo , Receptores Inmunológicos/química , Receptores Inmunológicos/metabolismo , Secuencia de Aminoácidos , Muerte Celular , Citometría de Flujo , Células HEK293 , Humanos , Ligandos , Macrófagos/inmunología , Macrófagos/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Resonancia por Plasmón de Superficie , Ultracentrifugación
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda