Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Intervalo de año de publicación
1.
Mol Microbiol ; 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38720451

RESUMEN

Aedes aegypti females are natural vectors of important arboviruses such as dengue, zika, and yellow fever. Mosquitoes activate innate immune response signaling pathways upon infection, as a resistance mechanism to fight pathogens and limit their propagation. Despite the beneficial effects of immune activation for insect vectors, phenotypic costs ultimately affect their fitness. However, the underlying mechanisms that mediate these fitness costs remain poorly understood. Given the high energy required to mount a proper immune response, we hypothesized that systemic activation of innate immunity would impair flight muscle mitochondrial function, compromising tissue energy demand and flight activity. Here, we investigated the dynamic effects of activation of innate immunity by intra-thoracic zymosan injection on A. aegypti flight muscle mitochondrial metabolism. Zymosan injection significantly increased defensin A expression in fat bodies in a time-dependent manner that compromised flight activity. Although oxidant levels in flight muscle were hardly altered, ATP-linked respiratory rates driven by mitochondrial pyruvate+proline oxidation were significantly reduced at 24 h upon zymosan injection. Oxidative phosphorylation coupling was preserved regardless of innate immune response activation along 24 h. Importantly, rotenone-sensitive respiration and complex I-III activity were specifically reduced 24 h upon zymosan injection. Also, loss of complex I activity compromised ATP-linked and maximal respiratory rates mediated by mitochondrial proline oxidation. Finally, the magnitude of innate immune response activation negatively correlated with respiratory rates, regardless of the metabolic states. Collectively, we demonstrate that activation of innate immunity is strongly associated with reduced flight muscle complex I activity with direct consequences to mitochondrial proline oxidation and flight activity. Remarkably, our results indicate a trade-off between dispersal and immunity exists in an insect vector, underscoring the potential consequences of disrupted flight muscle mitochondrial energy metabolism to arbovirus transmission.

2.
Nat Commun ; 15(1): 1422, 2024 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-38365823

RESUMEN

A novel cellular response of midgut progenitors (stem cells and enteroblasts) to Plasmodium berghei infection was investigated in Anopheles stephensi. The presence of developing oocysts triggers proliferation of midgut progenitors that is modulated by the Jak/STAT pathway and is proportional to the number of oocysts on individual midguts. The percentage of parasites in direct contact with enteroblasts increases over time, as progenitors proliferate. Silencing components of key signaling pathways through RNA interference (RNAi) that enhance proliferation of progenitor cells significantly decreased oocyst numbers, while limiting proliferation of progenitors increased oocyst survival. Live imaging revealed that enteroblasts interact directly with oocysts and eliminate them. Midgut progenitors sense the presence of Plasmodium oocysts and mount a cellular defense response that involves extensive proliferation and tissue remodeling, followed by oocysts lysis and phagocytosis of parasite remnants by enteroblasts.


Asunto(s)
Anopheles , Malaria , Parásitos , Plasmodium , Animales , Quinasas Janus , Factores de Transcripción STAT , Transducción de Señal , Malaria/parasitología , Anopheles/parasitología , Oocistos , Células Madre , Plasmodium berghei/fisiología
3.
Nature ; 623(7985): 149-156, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37880367

RESUMEN

Host factors that mediate Leishmania genetic exchange are not well defined. Here we demonstrate that natural IgM (IgMn)1-4 antibodies mediate parasite genetic exchange by inducing the transient formation of a spherical parasite clump that promotes parasite fusion and hybrid formation. We establish that IgMn from Leishmania-free animals binds to the surface of Leishmania parasites to induce significant changes in the expression of parasite transcripts and proteins. Leishmania binding to IgMn is partially lost after glycosidase treatment, although parasite surface phosphoglycans, including lipophosphoglycan, are not required for IgMn-induced parasite clumping. Notably, the transient formation of parasite clumps is essential for Leishmania hybridization in vitro. In vivo, we observed a 12-fold increase in hybrid formation in sand flies provided a second blood meal containing IgMn compared with controls. Furthermore, the generation of recombinant progeny from mating hybrids and parental lines were only observed in sand flies provided with IgMn. Both in vitro and in vivo IgM-induced Leishmania crosses resulted in full genome hybrids that show equal patterns of biparental contribution. Leishmania co-option of a host natural antibody to facilitate mating in the insect vector establishes a new paradigm of parasite-host-vector interdependence that contributes to parasite diversity and fitness by promoting genetic exchange.


Asunto(s)
Interacciones Huésped-Parásitos , Inmunoglobulina M , Leishmania , Psychodidae , Reproducción , Animales , Hibridación Genética , Inmunoglobulina M/inmunología , Leishmania/genética , Leishmania/inmunología , Psychodidae/inmunología , Psychodidae/parasitología , Reproducción/genética , Interacciones Huésped-Parásitos/genética , Interacciones Huésped-Parásitos/inmunología , Regulación de la Expresión Génica , Glicósido Hidrolasas/metabolismo
4.
bioRxiv ; 2023 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-37577486

RESUMEN

A novel cellular response of midgut progenitors (stem cells and enteroblasts) to Plasmodium berghei infection was investigated in Anopheles stephensi. The presence of developing oocysts triggers proliferation of midgut progenitors that is modulated by the Jak/STAT pathway, and proportional to the number of oocysts on individual midguts. The percentage of parasites in direct contact with enteroblasts increases over time, as progenitors proliferate. Enhancing proliferation of progenitors significantly decreases oocyst numbers, while limiting proliferation increases oocyst survival. Live imaging revealed that enteroblasts interact directly with oocysts and eliminate them. Midgut progenitors sense the presence of Plasmodium oocysts and mount a cellular defense response that involves extensive proliferation and tissue remodeling, followed by oocysts lysis and phagocytosis of parasite remnants by enteroblasts.

5.
PLoS Pathog ; 19(2): e1011149, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36780872

RESUMEN

Aedes aegypti mosquitoes are the main vectors of arboviruses. The peritrophic matrix (PM) is an extracellular layer that surrounds the blood bolus. It acts as an immune barrier that prevents direct contact of bacteria with midgut epithelial cells during blood digestion. Here, we describe a heme-dependent peroxidase, hereafter referred to as heme peroxidase 1 (HPx1). HPx1 promotes PM assembly and antioxidant ability, modulating vector competence. Mechanistically, the heme presence in a blood meal induces HPx1 transcriptional activation mediated by the E75 transcription factor. HPx1 knockdown increases midgut reactive oxygen species (ROS) production by the DUOX NADPH oxidase. Elevated ROS levels reduce microbiota growth while enhancing epithelial mitosis, a response to tissue damage. However, simultaneous HPx1 and DUOX silencing was not able to rescue bacterial population growth, as explained by increased expression of antimicrobial peptides (AMPs), which occurred only after double knockdown. This result revealed hierarchical activation of ROS and AMPs to control microbiota. HPx1 knockdown produced a 100-fold decrease in Zika and dengue 2 midgut infection, demonstrating the essential role of the mosquito PM in the modulation of arbovirus vector competence. Our data show that the PM connects blood digestion to midgut immunological sensing of the microbiota and viral infections.


Asunto(s)
Aedes , Arbovirus , Infección por el Virus Zika , Virus Zika , Animales , Humanos , Especies Reactivas de Oxígeno/metabolismo , Antioxidantes/metabolismo , Peroxidasa/metabolismo , Mosquitos Vectores , Hemo/metabolismo , Peroxidasas/metabolismo , Virus Zika/metabolismo
6.
PLoS Pathog, v. 19, n. 2, e1011149, fev. 2023
Artículo en Inglés | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: bud-4804

RESUMEN

Aedes aegypti mosquitoes are the main vectors of arboviruses. The peritrophic matrix (PM) is an extracellular layer that surrounds the blood bolus. It acts as an immune barrier that prevents direct contact of bacteria with midgut epithelial cells during blood digestion. Here, we describe a heme-dependent peroxidase, hereafter referred to as heme peroxidase 1 (HPx1). HPx1 promotes PM assembly and antioxidant ability, modulating vector competence. Mechanistically, the heme presence in a blood meal induces HPx1 transcriptional activation mediated by the E75 transcription factor. HPx1 knockdown increases midgut reactive oxygen species (ROS) production by the DUOX NADPH oxidase. Elevated ROS levels reduce microbiota growth while enhancing epithelial mitosis, a response to tissue damage. However, simultaneous HPx1 and DUOX silencing was not able to rescue bacterial population growth, as explained by increased expression of antimicrobial peptides (AMPs), which occurred only after double knockdown. This result revealed hierarchical activation of ROS and AMPs to control microbiota. HPx1 knockdown produced a 100-fold decrease in Zika and dengue 2 midgut infection, demonstrating the essential role of the mosquito PM in the modulation of arbovirus vector competence. Our data show that the PM connects blood digestion to midgut immunological sensing of the microbiota and viral infections.

7.
Elife ; 112022 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-36052991

RESUMEN

Activation of Toll signaling in Anopheles gambiae by silencing Cactus, a suppressor of this pathway, enhances local release of hemocyte-derived microvesicles (HdMv), promoting activation of the mosquito complement-like system, which eliminates Plasmodium ookinetes. We uncovered the mechanism of this immune enhancement. Cactus silencing triggers a Rel1-mediated differentiation of granulocytes to the megacyte lineage, a new subpopulation of giant cells, resulting in a dramatic increase in the proportion of circulating megacytes. Megacytes are very plastic cells that are massively recruited to the basal midgut surface in response to Plasmodium infection. We show that Toll signaling modulates hemocyte differentiation and that megacyte recruitment to the midgut greatly enhances mosquito immunity against Plasmodium.


Malaria causes hundreds of thousands of deaths each year. This devastating disease is caused by Plasmodium parasites, which are transmitted to people through female Anopheles gambiae mosquitos. Mosquitos become infected with Plasmodium when they ingest blood containing these malaria-causing parasites. However, Plasmodium must avoid the mosquito immune system to survive and spread. The mosquito immune system is made up of several types of immune cells, including cells known as granulocytes. Granulocytes can further develop into additional cell subtypes, such as megacytes and antimicrobial granulocytes, but it is not clear how these types of cells work to protect mosquitos against infections. In the mosquitos that transmit malaria, a cell signaling pathway called Toll helps control immune responses to disease-causing microbes, such as Plasmodium. When Toll signaling is strongly triggered in mosquitos, Plasmodium infection is eliminated because immune cell responses are enhanced ­ which results in lower levels of transmission to humans. But what is the underlying mechanism through which high levels of Toll signaling eradicate Plasmodium infection? To find out, Barletta et al. collected cell samples from A. gambiae mosquitos and analyzed what happened when Toll signaling was strongly activated. They observed a large increase in the proportion of megacytes in these mosquitos (from 2% to 80% of all granulocytes). Toll signaling also caused megacytes to become bigger, cluster together, and have higher plasticity ­ meaning they could adopt different shapes. Barletta et al. used microscopy to show that these megacytes were releasing large mitochondria-like structures and membrane vesicles , which may be the trigger activating the mosquito's immune system. In live mosquitos, megacytes move towards the area of the Plasmodium infection and release microvesicles. These microvesicles are known to activate a part of the the mosquito's immune system called the complement-like system, destroying the parasites and preventing mosquito infection and disease transmission. These findings show how strong Toll signaling triggers the mosquito immune system to eliminate Plasmodium infections. Understanding how the mosquito immune system tackles Plasmodium infection may help reveal ways to reduce or block transmission.


Asunto(s)
Anopheles , Malaria , Plasmodium , Animales , Hemocitos , Humanos , Plásticos/metabolismo
9.
Proc Natl Acad Sci U S A ; 118(44)2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34711682

RESUMEN

Immune priming in Anopheles gambiae is mediated by the systemic release of a hemocyte differentiation factor (HDF), a complex of lipoxin A4 bound to Evokin, a lipid carrier. HDF increases the proportion of circulating granulocytes and enhances mosquito cellular immunity. Here, we show that Evokin is present in hemocytes and fat-body cells, and messenger RNA (mRNA) expression increases significantly after immune priming. The double peroxidase (DBLOX) enzyme, present in insects but not in vertebrates, is essential for HDF synthesis. DBLOX is highly expressed in oenocytes in the fat-body tissue, and these cells increase in number in primed mosquitoes. We provide direct evidence that the histone acetyltransferase AgTip60 (AGAP001539) is also essential for a sustained increase in oenocyte numbers, HDF synthesis, and immune priming. We propose that oenocytes may function as a population of cells that are reprogrammed, and orchestrate and maintain a broad, systemic, and long-lasting state of enhanced immune surveillance in primed mosquitoes.


Asunto(s)
Culicidae/inmunología , Histona Acetiltransferasas/metabolismo , Memoria Inmunológica/inmunología , Animales , Anopheles/inmunología , Anopheles/metabolismo , Culicidae/metabolismo , Femenino , Granulocitos/metabolismo , Hemocitos/inmunología , Inmunidad Innata/inmunología , Proteínas de Insectos/genética , Insectos/metabolismo , Lipoxinas/metabolismo , Malaria/inmunología , Masculino , Peroxidasa/metabolismo , Plasmodium/metabolismo , Plasmodium berghei/metabolismo
10.
J Med Entomol ; 58(2): 634-645, 2021 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-33710316

RESUMEN

We investigated by scanning electron microscopy the morphology, distribution, and abundance of antennal sensilla of females Phlebotomus duboscqi sand fly, an important vector of zoonotic cutaneous leishmaniasis at Afrotropical region. Thirteen well-differentiated sensilla were identified, among six types of cuticular sensilla. The probable function of these sensillary types is discussed in relation to their external structure and distribution. Five sensillary types were classified as olfactory sensilla, as they have specific morphological characters of sensilla with this function. Number and distribution of sensilla significantly differed between antennal segments. The results of the present work, besides corroborating in the expansion of the morphological and ultrastructural knowledge of P. duboscqi, can foment future electrophysiological studies for the development of volatile semiochemicals, to be used as attractants in traps for monitoring and selective vector control of this sand fly.


Asunto(s)
Phlebotomus/ultraestructura , Sensilos/ultraestructura , Animales , Femenino , Phlebotomus/fisiología , Sensilos/fisiología
11.
PLoS Negl Trop Dis ; 14(10): e0008706, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33095767

RESUMEN

Prostaglandins (PGs) are immuno-active lipids that mediate the immune response in invertebrates and vertebrates. In insects, PGs play a role on different physiological processes such as reproduction, ion transport and regulation of cellular immunity. However, it is unclear whether PGs play a role in invertebrate's humoral immunity, and, if so, which immune signaling pathways would be modulated by PGs. Here, we show that Aedes aegypti gut microbiota and Gram-negative bacteria challenge induces prostaglandin production sensitive to an irreversible inhibitor of the vertebrate cyclooxygenase, acetylsalicylic acid (ASA). ASA treatment reduced PG synthesis and is associated with decreased expression of components of the Toll and IMD immune pathways, thereby rendering mosquitoes more susceptible to both bacterial and viral infections. We also shown that a cytosolic phospholipase (PLAc), one of the upstream regulators of PG synthesis, is induced by the microbiota in the midgut after blood feeding. The knockdown of the PLAc decreased prostaglandin production and enhanced the replication of Dengue in the midgut. We conclude that in Ae. aegypti, PGs control the amplitude of the immune response to guarantee an efficient pathogen clearance.


Asunto(s)
Aedes/virología , Virus del Dengue/fisiología , Inmunidad Humoral , Prostaglandinas/metabolismo , Aedes/inmunología , Animales , Línea Celular , Virus del Dengue/inmunología , Femenino , Regulación Enzimológica de la Expresión Génica , Interacciones Huésped-Patógeno , Fosfolipasas A2/genética , Fosfolipasas A2/metabolismo , Prostaglandinas/genética
12.
Science ; 369(6507): 1128-1132, 2020 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-32855340

RESUMEN

Hemocytes limit the capacity of mosquitoes to transmit human pathogens. Here we profile the transcriptomes of 8506 hemocytes of Anopheles gambiae and Aedes aegypti mosquito vectors. Our data reveal the functional diversity of hemocytes, with different subtypes of granulocytes expressing distinct and evolutionarily conserved subsets of effector genes. A previously unidentified cell type in An. gambiae, which we term "megacyte," is defined by a specific transmembrane protein marker (TM7318) and high expression of lipopolysaccharide-induced tumor necrosis factor-α transcription factor 3 (LL3). Knockdown experiments indicate that LL3 mediates hemocyte differentiation during immune priming. We identify and validate two main hemocyte lineages and find evidence of proliferating granulocyte populations. This atlas of medically relevant invertebrate immune cells at single-cell resolution identifies cellular events that underpin mosquito immunity to malaria infection.


Asunto(s)
Aedes/inmunología , Anopheles/inmunología , Hemocitos/inmunología , Inmunidad Celular , Malaria/transmisión , Mosquitos Vectores/inmunología , Aedes/genética , Animales , Anopheles/genética , Femenino , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Granulocitos/inmunología , Hemocitos/metabolismo , Malaria/inmunología , Malaria/parasitología , Ratones , Mosquitos Vectores/genética , RNA-Seq , Análisis de la Célula Individual
13.
J Med Entomol ; 57(6): 1722-1734, 2020 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-32761144

RESUMEN

The antennal sensilla and the antenna of females Nyssomyia intermedia, one of the main vectors of American cutaneous leishmaniasis, were studied by scanning electron microscopy. The main goal was to characterize the quantity, typology, and topography of the sensilla with particular attention to the olfactory types. The insects were captured in the city of Corte de Pedra, State of Bahia, Brazil, by CDC-type light traps and raised in a laboratory as a new colony. Fourteen well-differentiated sensilla were identified, among six cuticular types: trichoidea, campaniformia, squamiformia, basiconica, chaetica, and coeloconica. Of these, six sensilla were classified as olfactory sensilla due to their specific morphological features. Smaller noninnervated pilosities of microtrichiae type were also evidenced by covering all antennal segments. The antennal segments differ in shapes and sizes, and the amount and distribution of types and subtypes of sensilla. This study may foment future taxonomic and phylogenetic analysis for a better evolutionary understanding of the sand flies. Besides, it may assist the targeting of future electrophysiological studies by Single Sensillum Recording, and aim to develop alternative measures of monitoring and control of this vector.


Asunto(s)
Antenas de Artrópodos/ultraestructura , Insectos Vectores/ultraestructura , Psychodidae/ultraestructura , Animales , Brasil , Femenino , Leishmaniasis Cutánea , Microscopía Electrónica de Rastreo , Sensilos/ultraestructura
14.
Front Microbiol ; 11: 1496, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32719666

RESUMEN

An effective vaccine to reduce malaria transmission is central to control and ultimately achieve disease eradication. Recently, we demonstrated that antibodies targeting the Plasmodium falciparum surface protein P47 (Pfs47) reduce parasite transmission to Anopheles gambiae mosquitoes. Here, Plasmodium berghei (Pb) was used as a model to assess the in vivo efficacy of a P47-targeted transmission blocking vaccine (Pbs47). Mice were immunized following a prime/boost regimen and infected with P. berghei. The effect of immunization on infectivity to mosquitoes was evaluated by direct feeding on P. berghei-infected mice. The key region in Pbs47 where antibody binding confers protection was mapped, and the immunogenicity of this protective antigen was enhanced by conjugation to a virus-like particle. Passive immunization with 100 and 50 µg/mL of anti-Pbs47 IgG reduced oocyst density by 77 and 67%, respectively. Furthermore, affinity purified Pbs47-specific IgG significantly reduced oocyst density by 88 and 77%, respectively at doses as low as 10 and 1 µg/mL. These studies suggest that P47 is a promising transmission blocking target and show that antibodies to the same specific region in Pfs47 and Pbs47 confer protection.

15.
Nat Commun ; 11(1): 2759, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32488028

RESUMEN

Human noroviruses are a major cause of diarrheal illness, but pathogenesis is poorly understood. Here, we investigate the cellular tropism of norovirus in specimens from four immunocompromised patients. Abundant norovirus antigen and RNA are detected throughout the small intestinal tract in jejunal and ileal tissue from one pediatric intestinal transplant recipient with severe gastroenteritis. Negative-sense viral RNA, a marker of active viral replication, is found predominantly in intestinal epithelial cells, with chromogranin A-positive enteroendocrine cells (EECs) identified as a permissive cell type in this patient. These findings are consistent with the detection of norovirus-positive EECs in the other three immunocompromised patients. Investigation of the signaling pathways induced in EECs that mediate communication between the gut and brain may clarify mechanisms of pathogenesis and lead to the development of in vitro model systems in which to evaluate norovirus vaccines and treatment.


Asunto(s)
Células Enteroendocrinas/inmunología , Células Epiteliales/inmunología , Norovirus/fisiología , Enfermedad Aguda , District of Columbia , Células Enteroendocrinas/metabolismo , Gastroenteritis/virología , Genotipo , Humanos , Intestino Delgado/patología , Intestino Delgado/virología , Norovirus/genética , ARN Viral , Replicación Viral
16.
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
17.
iScience ; 19: 54-62, 2019 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-31351392

RESUMEN

Anopheles gambiae mosquitoes that have been infected with Plasmodium mount a more effective immune response to a subsequent infection. Priming is established when Plasmodium invasion of the mosquito midgut allows contact of the gut microbiota with epithelial cells. This event is followed by a systemic release of a hemocyte differentiation factor (HDF) consisting of Lipoxin A4 bound to Evokin, a lipocalin carrier, which increases the proportion of circulating hemocytes. We show that mosquito midgut cells produce and release prostaglandin E2 (PGE2), which attracts hemocytes to the midgut surface and enhances their patrolling activity. Systemic injection of prostaglandins (PGs) recapitulates the priming response and enhances antiplasmodial immunity by triggering HDF production. Although insects lack cyclooxygenases, two heme peroxidases, HPX7 and HPX8, catalyze essential steps in PG biosynthesis in mosquitoes. Mosquito midgut PGE2 release attracts hemocytes and establishes a long-lasting enhanced systemic cellular immune response to Plasmodium infection.

18.
PLoS Negl Trop Dis ; 12(9): e0006785, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30248099

RESUMEN

Innate immunity is an ancient and conserved defense system that provides an early effective response against invaders. Many immune genes of Anopheles mosquitoes have been implicated in defense against a variety of pathogens, including plasmodia. Nevertheless, only recent work identified some immune genes of Anopheles aquasalis mosquitoes upon P. vivax infection. Among these was a GATA transcription factor gene, which is described here. This is an ortholog of GATA factor Serpent genes described in Drosophila melanogaster and Anopheles gambiae. Gene expression analyses showed an increase of GATA-Serpent mRNA in P. vivax-infected A. aquasalis and functional RNAi experiments identified this transcription factor as an important immune gene of A. aquasalis against both bacteria and P. vivax. Besides, we were able to identify an effect of GATA-Serpent knockdown on A. aquasalis hemocyte proliferation and differentiation. These findings expand our understanding of the poorly studied A. aquasalis-P. vivax interactions and uncover GATA-Serpent as a key player of the mosquito innate immune response.


Asunto(s)
Anopheles/inmunología , Bacterias/inmunología , Factores de Transcripción GATA/metabolismo , Inmunidad Innata , Plasmodium/inmunología , Animales , Anopheles/genética , Diferenciación Celular , Proliferación Celular , Femenino , Factores de Transcripción GATA/genética , Perfilación de la Expresión Génica , Silenciador del Gen , Hemocitos/inmunología , Hemocitos/fisiología
19.
Parasit Vectors ; 10(1): 103, 2017 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-28231846

RESUMEN

BACKGROUND: Aedes aegypti is the main vector of important arboviruses such as dengue, Zika and chikungunya. During infections mosquitoes can activate the immune pathways Toll, IMD and JAK/STAT to limit pathogen replication. RESULTS: Here, we evaluate the immune response profile of Ae. aegypti against Sindbis virus (SINV). We analyzed gene expression of components of Toll, IMD and JAK/STAT pathways and showed that a blood meal and virus infection upregulated aaREL2 in a microbiota-dependent fashion, since this induction was prevented by antibiotic. The presence of the microbiota activates IMD and impaired the replication of SINV in the midgut. Constitutive activation of the IMD pathway, by Caspar depletion, leads to a decrease in microbiota levels and an increase in SINV loads. CONCLUSION: Together, these results suggest that a blood meal is able to activate innate immune pathways, through a nutrient induced growth of microbiota, leading to upregulation of aaREL2 and IMD activation. Microbiota levels seemed to have a reciprocal interaction, where the proliferation of the microbiota activates IMD pathway that in turn controls bacterial levels, allowing SINV replication in Ae. aegypti mosquitoes. The activation of the IMD pathway seems to have an indirect effect in SINV levels that is induced by the microbiota.


Asunto(s)
Aedes/virología , Regulación de la Expresión Génica/inmunología , Microbiota/fisiología , Virus Sindbis/fisiología , Aedes/inmunología , Animales , Antibacterianos/farmacología , Interacciones Huésped-Patógeno , Microbiota/efectos de los fármacos , Penicilinas/farmacología , Estreptomicina/farmacología , Transcriptoma
20.
Curr Biol ; 26(16): 2188-93, 2016 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-27476595

RESUMEN

Blood-feeding arthropods are vectors of infectious diseases such as dengue, Zika, Chagas disease, and malaria [1], and vector control is essential to limiting disease spread. Because these arthropods ingest very large amounts of blood, a protein-rich meal, huge amounts of amino acids are produced during digestion. Previous work on Rhodnius prolixus, a vector of Chagas disease, showed that, among all amino acids, only tyrosine degradation enzymes were overexpressed in the midgut compared to other tissues [2]. Here we demonstrate that tyrosine detoxification is an essential trait in the life history of blood-sucking arthropods. We found that silencing Rhodnius tyrosine aminotransferase (TAT) and 4-hydroxyphenylpyruvate dioxygenase (HPPD), the first two enzymes of the phenylalanine/tyrosine degradation pathway, caused the death of insects after a blood meal. This was confirmed by using the HPPD inhibitor mesotrione, which selectively killed hematophagous arthropods but did not affect non-hematophagous insects. In addition, mosquitoes and kissing bugs died after feeding on mice that had previously received a therapeutic effective oral dose (1 mg/kg) of nitisinone, another HPPD inhibitor used in humans for the treatment of tyrosinemia type I [3]. These findings indicate that HPPD (and TAT) can be a target for the selective control of blood-sucking disease vector populations. Because HPPD inhibitors are extensively used as herbicides and in medicine, these compounds may provide an alternative less toxic to humans and more environmentally friendly than the conventional neurotoxic insecticides that are currently used, with the ability to affect only hematophagous arthropods.


Asunto(s)
4-Hidroxifenilpiruvato Dioxigenasa/genética , Silenciador del Gen , Proteínas de Insectos/genética , Rhodnius/genética , Tirosina Transaminasa/genética , Tirosina/metabolismo , 4-Hidroxifenilpiruvato Dioxigenasa/metabolismo , Animales , Femenino , Inactivación Metabólica , Proteínas de Insectos/metabolismo , Masculino , Ninfa/genética , Ninfa/crecimiento & desarrollo , Ninfa/metabolismo , Rhodnius/crecimiento & desarrollo , Rhodnius/metabolismo , Tirosina Transaminasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA