Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
2.
PLoS One ; 14(9): e0219523, 2019.
Article in English | MEDLINE | ID: mdl-31479460

ABSTRACT

Whole mitogenome sequences (mtDNA) have been exploited for insect ecology studies, using them as molecular markers to reconstruct phylogenies, or to infer phylogeographic relationships and gene flow. Recent Anopheles phylogenomic studies have provided information regarding the time of deep lineage divergences within the genus. Here we report the complete 15,393 bp mtDNA sequences of Anopheles aquasalis, a Neotropical human malaria vector. When comparing its structure and base composition with other relevant and available anopheline mitogenomes, high similarity and conserved genomic features were observed. Furthermore, 22 mtDNA sequences comprising anopheline and Dipteran sibling species were analyzed to reconstruct phylogenies and estimate dates of divergence between taxa. Phylogenetic analysis using complete mtDNA sequences suggests that A. aquasalis diverged from the Anopheles albitarsis complex ~28 million years ago (MYA), and ~38 MYA from Anopheles darlingi. Bayesian analysis suggests that the most recent ancestor of Nyssorhynchus and Anopheles + Cellia was extant ~83 MYA, corroborating current estimates of ~79-100 MYA. Additional sampling and publication of African, Asian, and North American anopheline mitogenomes would improve the resolution of the Anopheles phylogeny and clarify early continental dispersal routes.


Subject(s)
Anopheles/classification , Anopheles/genetics , Genome, Mitochondrial , Genomics , Phylogeny , Phylogeography , Animals , Base Composition , Computational Biology/methods , Evolution, Molecular , Genomics/methods , Humans , Molecular Sequence Annotation , Mosquito Vectors/classification , Mosquito Vectors/genetics , Sequence Analysis, DNA , Whole Genome Sequencing
3.
PLoS Negl Trop Dis ; 12(9): e0006785, 2018 09.
Article in English | MEDLINE | ID: mdl-30248099

ABSTRACT

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.


Subject(s)
Anopheles/immunology , Bacteria/immunology , GATA Transcription Factors/metabolism , Immunity, Innate , Plasmodium/immunology , Animals , Anopheles/genetics , Cell Differentiation , Cell Proliferation , Female , GATA Transcription Factors/genetics , Gene Expression Profiling , Gene Silencing , Hemocytes/immunology , Hemocytes/physiology
4.
PLoS Negl Trop Dis ; 12(8): e0006709, 2018 08.
Article in English | MEDLINE | ID: mdl-30138419

ABSTRACT

BACKGROUND/METHODOLOGY: Triatomine bugs are the vectors of Trypanosoma cruzi, the agent of Chagas disease. Vector control has for decades relied upon insecticide spraying, but insecticide resistance has recently emerged in several triatomine populations. One alternative strategy to reduce T. cruzi transmission is paratransgenesis, whereby symbiotic bacteria are genetically engineered to produce T. cruzi-killing proteins in the vector's gut. This approach requires in-depth knowledge of the vectors' natural gut microbiota. Here, we use metagenomics (16S rRNA 454 pyrosequencing) to describe the gut microbiota of field-caught Triatoma sordida-likely the most common peridomestic triatomine in Brazil. For large nymphs (4th and 5th stage) and adults, we also studied separately the three main digestive-tract segments-anterior midgut, posterior midgut, and hindgut. PRINCIPAL FINDINGS: Bacteria of four phyla (12 genera) were present in both nymphs (all five stages) and adults, thus defining T. sordida's 'bacterial core': Actinobacteria (Brevibacterium, Corynebacterium, Dietzia, Gordonia, Nitriliruptor, Nocardia, Nocardiopsis, Rhodococcus, and Williamsia), Proteobacteria (Pseudomonas and Sphingobium), and Firmicutes (Staphylococcus). We found some clear differences in bacterial composition and relative abundance among development stages; overall, Firmicutes and Proteobacteria increased, but Actinobacteria decreased, through development. Finally, the bacterial microbiotas of the bugs' anterior midgut, posterior midgut, and hindgut were sharply distinct. CONCLUSIONS/SIGNIFICANCE: Our results identify the 'bacterial core set' of T. sordida and reveal important gut microbiota differences among development stages-particularly between 1st-3rd stage nymphs and adults. Further, we show that, within any given development stage, the vectors' gut cannot be regarded as a single homogeneous environment. Cultivable, non-pathogenic 'core' bacterial species may now be tested as candidates for paratransgenic control of T. cruzi transmission by T. sordida.


Subject(s)
Bacteria/classification , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Triatoma/microbiology , Animals , Brazil , Female , Male , Nymph , RNA, Bacterial/genetics , RNA, Ribosomal, 16S/genetics , Triatoma/growth & development
5.
PLoS One ; 10(8): e0135985, 2015.
Article in English | MEDLINE | ID: mdl-26275150

ABSTRACT

Blood-feeding mosquitoes are exposed to high levels of heme, the product of hemoglobin degradation. Heme is a pro-oxidant that influences a variety of cellular processes. We performed a global analysis of heme-regulated Aedes aegypti (yellow fever mosquito) transcriptional changes to better understand influence on mosquito physiology at the molecular level. We observed an iron- and reactive oxygen species (ROS)-independent signaling induced by heme that comprised genes related to redox metabolism. By modulating the abundance of these transcripts, heme possibly acts as a danger signaling molecule. Furthermore, heme triggered critical changes in the expression of energy metabolism and immune response genes, altering the susceptibility towards bacteria and dengue virus. These findings seem to have implications on the adaptation of mosquitoes to hematophagy and consequently on their ability to transmit diseases. Altogether, these results may also contribute to the understanding of heme cell biology in eukaryotic cells.


Subject(s)
Dengue Virus/pathogenicity , Aedes/virology , Animals , Heme/metabolism , Immunity/physiology , Reactive Oxygen Species/metabolism , Signal Transduction/physiology
6.
Mem Inst Oswaldo Cruz ; 110(1): 23-47, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25742262

ABSTRACT

In the Americas, areas with a high risk of malaria transmission are mainly located in the Amazon Forest, which extends across nine countries. One keystone step to understanding the Plasmodium life cycle in Anopheles species from the Amazon Region is to obtain experimentally infected mosquito vectors. Several attempts to colonise Anopheles species have been conducted, but with only short-lived success or no success at all. In this review, we review the literature on malaria transmission from the perspective of its Amazon vectors. Currently, it is possible to develop experimental Plasmodium vivax infection of the colonised and field-captured vectors in laboratories located close to Amazonian endemic areas. We are also reviewing studies related to the immune response to P. vivax infection of Anopheles aquasalis, a coastal mosquito species. Finally, we discuss the importance of the modulation of Plasmodium infection by the vector microbiota and also consider the anopheline genomes. The establishment of experimental mosquito infections with Plasmodium falciparum, Plasmodium yoelii and Plasmodium berghei parasites that could provide interesting models for studying malaria in the Amazonian scenario is important. Understanding the molecular mechanisms involved in the development of the parasites in New World vectors is crucial in order to better determine the interaction process and vectorial competence.


Subject(s)
Anopheles/parasitology , Insect Vectors/parasitology , Malaria/transmission , Plasmodium/classification , Animals , Anopheles/classification , Anopheles/genetics , Anopheles/immunology , Anopheles/ultrastructure , Disease Models, Animal , Insect Vectors/classification , Insect Vectors/genetics , Insect Vectors/immunology , Insect Vectors/ultrastructure , Malaria/immunology , Mosquito Control , Parasite Load , Rainforest
7.
PLoS Pathog ; 11(2): e1004631, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25658622

ABSTRACT

Actin is a highly versatile, abundant, and conserved protein, with functions in a variety of intracellular processes. Here, we describe a novel role for insect cytoplasmic actin as an extracellular pathogen recognition factor that mediates antibacterial defense. Insect actins are secreted from cells upon immune challenge through an exosome-independent pathway. Anopheles gambiae actin interacts with the extracellular MD2-like immune factor AgMDL1, and binds to the surfaces of bacteria, mediating their phagocytosis and direct killing. Globular and filamentous actins display distinct functions as extracellular immune factors, and mosquito actin is a Plasmodium infection antagonist.


Subject(s)
Actins/immunology , Anopheles/immunology , Insect Proteins/immunology , Malaria/immunology , Plasmodium falciparum/immunology , Actins/metabolism , Animals , Anopheles/metabolism , Blotting, Western , Cell Line , Cytoplasm/immunology , Cytoplasm/metabolism , Host-Parasite Interactions/immunology , Insect Proteins/metabolism , Malaria/metabolism , Phagocytosis/immunology , Polymerase Chain Reaction , Two-Hybrid System Techniques
8.
Mem. Inst. Oswaldo Cruz ; 110(1): 23-47, 03/02/2015. graf
Article in English | LILACS | ID: lil-741609

ABSTRACT

In the Americas, areas with a high risk of malaria transmission are mainly located in the Amazon Forest, which extends across nine countries. One keystone step to understanding the Plasmodium life cycle in Anopheles species from the Amazon Region is to obtain experimentally infected mosquito vectors. Several attempts to colonise Ano- pheles species have been conducted, but with only short-lived success or no success at all. In this review, we review the literature on malaria transmission from the perspective of its Amazon vectors. Currently, it is possible to develop experimental Plasmodium vivax infection of the colonised and field-captured vectors in laboratories located close to Amazonian endemic areas. We are also reviewing studies related to the immune response to P. vivax infection of Anopheles aquasalis, a coastal mosquito species. Finally, we discuss the importance of the modulation of Plasmodium infection by the vector microbiota and also consider the anopheline genomes. The establishment of experimental mosquito infections with Plasmodium falciparum, Plasmodium yoelii and Plasmodium berghei parasites that could provide interesting models for studying malaria in the Amazonian scenario is important. Understanding the molecular mechanisms involved in the development of the parasites in New World vectors is crucial in order to better determine the interaction process and vectorial competence.


Subject(s)
Adult , Female , Humans , Male , Middle Aged , Amoxicillin/administration & dosage , Anti-Bacterial Agents/administration & dosage , Drugs, Chinese Herbal/administration & dosage , Helicobacter Infections/drug therapy , Helicobacter pylori/drug effects , Omeprazole/analogs & derivatives , Peptic Ulcer/drug therapy , Anti-Ulcer Agents/administration & dosage , Clarithromycin/administration & dosage , Double-Blind Method , Drug Therapy, Combination , Follow-Up Studies , Helicobacter Infections/pathology , Lansoprazole , Omeprazole/administration & dosage , Prospective Studies , Peptic Ulcer/microbiology , Peptic Ulcer/pathology , Recurrence , Wound Healing/drug effects
9.
PLoS Pathog ; 10(10): e1004398, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25340821

ABSTRACT

Plasmodium and dengue virus, the causative agents of the two most devastating vector-borne diseases, malaria and dengue, are transmitted by the two most important mosquito vectors, Anopheles gambiae and Aedes aegypti, respectively. Insect-bacteria associations have been shown to influence vector competence for human pathogens through multi-faceted actions that include the elicitation of the insect immune system, pathogen sequestration by microbes, and bacteria-produced anti-pathogenic factors. These influences make the mosquito microbiota highly interesting from a disease control perspective. Here we present a bacterium of the genus Chromobacterium (Csp_P), which was isolated from the midgut of field-caught Aedes aegypti. Csp_P can effectively colonize the mosquito midgut when introduced through an artificial nectar meal, and it also inhibits the growth of other members of the midgut microbiota. Csp_P colonization of the midgut tissue activates mosquito immune responses, and Csp_P exposure dramatically reduces the survival of both the larval and adult stages. Ingestion of Csp_P by the mosquito significantly reduces its susceptibility to Plasmodium falciparum and dengue virus infection, thereby compromising the mosquito's vector competence. This bacterium also exerts in vitro anti-Plasmodium and anti-dengue activities, which appear to be mediated through Csp_P -produced stable bioactive factors with transmission-blocking and therapeutic potential. The anti-pathogen and entomopathogenic properties of Csp_P render it a potential candidate for the development of malaria and dengue control strategies.


Subject(s)
Anopheles/microbiology , Chromobacterium/metabolism , Dengue Virus , Gram-Negative Bacterial Infections/metabolism , Malaria/microbiology , Animals , Culicidae , Genetic Vectors/genetics , Humans , In Vitro Techniques , Plasmodium falciparum/microbiology , Virulence Factors
10.
Environ Microbiol ; 16(9): 2980-94, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24428613

ABSTRACT

Malaria parasite transmission requires the successful development of Plasmodium gametocytes into flagellated microgametes upon mosquito blood ingestion, and the subsequent fertilization of microgametes and macrogametes for the development of motile zygotes, called ookinetes, which invade and transverse the Anopheles vector mosquito midgut at around 18-36 h after blood ingestion. Within the mosquito midgut, the malaria parasite has to withstand the mosquito's innate immune response and the detrimental effect of its commensal bacterial flora. We have assessed the midgut colonization capacity of five gut bacterial isolates from field-derived, and two from laboratory colony, mosquitoes and their effect on Plasmodium development in vivo and in vitro, along with their impact on mosquito survival. Some bacterial isolates activated the mosquito's immune system, affected the mosquito's lifespan, and were capable of blocking Plasmodium development. We have also shown that the ability of these bacteria to inhibit the parasites is likely to involve different mechanisms and factors. A Serratia marcescens isolate was particularly efficient in colonizing the mosquitoes' gut, compromising mosquito survival and inhibiting both Plasmodium sexual- and asexual-stage through secreted factors, thereby rendering it a potential candidate for the development of a malaria transmission intervention strategy.


Subject(s)
Anopheles/microbiology , Digestive System/microbiology , Plasmodium/microbiology , Serratia marcescens/physiology , Animals , Anopheles/immunology , Anopheles/parasitology , Bacteria/isolation & purification , Female , Immunity, Innate , Mice , Serratia marcescens/isolation & purification
11.
PLoS One ; 8(2): e57014, 2013.
Article in English | MEDLINE | ID: mdl-23441231

ABSTRACT

Malaria affects millions of people worldwide and hundreds of thousands of people each year in Brazil. The mosquito Anopheles aquasalis is an important vector of Plasmodium vivax, the main human malaria parasite in the Americas. Reactive oxygen species (ROS) have been shown to have a role in insect innate immune responses as a potent pathogen-killing agent. We investigated the mechanisms of free radicals modulation after A. aquasalis infection with P. vivax. ROS metabolism was evaluated in the vector by studying expression and activity of three key detoxification enzymes, one catalase and two superoxide dismutases (SOD3A and SOD3B). Also, the involvement of free radicals in the mosquito immunity was measured by silencing the catalase gene followed by infection of A. aquasalis with P. vivax. Catalase, SOD3A and SOD3B expression in whole A. aquasalis were at the same levels of controls at 24 h and upregulated 36 h after ingestion of blood containing P. vivax. However, in the insect isolated midgut, the mRNA for these enzymes was not regulated by P. vivax infection, while catalase activity was reduced 24 h after the infectious meal. RNAi-mediated silencing of catalase reduced enzyme activity in the midgut, resulted in increased P. vivax infection and prevalence, and decreased bacterial load in the mosquito midgut. Our findings suggest that the interactions between A. aquasalis and P. vivax do not follow the model of ROS-induced parasite killing. It appears that P. vivax manipulates the mosquito detoxification system in order to allow its own development. This can be an indirect effect of fewer competitive bacteria present in the mosquito midgut caused by the increase of ROS after catalase silencing. These findings provide novel information on unique aspects of the main malaria parasite in the Americas interaction with one of its natural vectors.


Subject(s)
Anopheles/metabolism , Anopheles/parasitology , Plasmodium vivax/physiology , Reactive Oxygen Species/metabolism , Amino Acid Sequence , Animals , Anopheles/genetics , Catalase/genetics , Catalase/metabolism , Disease Susceptibility , Enzyme Activation , Female , Gene Silencing , Humans , Male , Molecular Sequence Data , Phylogeny , Sequence Alignment , Superoxide Dismutase/chemistry , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Transcription, Genetic
12.
PLoS Pathog ; 8(6): e1002737, 2012.
Article in English | MEDLINE | ID: mdl-22685401

ABSTRACT

The Anopheles gambiae immune response against Plasmodium falciparum, an etiological agent of human malaria, has been identified as a source of potential anti-Plasmodium genes and mechanisms to be exploited in efforts to control the malaria transmission cycle. One such mechanism is the Imd pathway, a conserved immune signaling pathway that has potent anti-P. falciparum activity. Silencing the expression of caspar, a negative regulator of the Imd pathway, or over-expressing rel2, an Imd pathway-controlled NFkappaB transcription factor, confers a resistant phenotype on A. gambiae mosquitoes that involves an array of immune effector genes. However, unexplored features of this powerful mechanism that may be essential for the implementation of a malaria control strategy still remain. Using RNA interference to singly or dually silence caspar and other components of the Imd pathway, we have identified genes participating in the anti-Plasmodium signaling module regulated by Caspar, each of which represents a potential target to achieve over-activation of the pathway. We also determined that the Imd pathway is most potent against the parasite's ookinete stage, yet also has reasonable activity against early oocysts and lesser activity against late oocysts. We further demonstrated that caspar silencing alone is sufficient to induce a robust anti-P. falciparum response even in the relative absence of resident gut microbiota. Finally, we established the relevance of the Imd pathway components and regulated effectors TEP1, APL1, and LRIM1 in parasite infection intensity-dependent defense, thereby shedding light on the relevance of laboratory versus natural infection intensity models. Our results highlight the physiological considerations that are integral to a thoughtful implementation of Imd pathway manipulation in A. gambiae as part of an effort to limit the malaria transmission cycle, and they reveal a variety of previously unrecognized nuances in the Imd-directed immune response against P. falciparum.


Subject(s)
Anopheles/immunology , Anopheles/parasitology , Insect Proteins/immunology , Malaria/immunology , Plasmodium falciparum/immunology , Animals , Insect Vectors/immunology , Malaria, Falciparum/prevention & control , RNA Interference , Real-Time Polymerase Chain Reaction , Signal Transduction
13.
PLoS Negl Trop Dis ; 5(11): e1317, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22069502

ABSTRACT

Malaria affects 300 million people worldwide every year and 450,000 in Brazil. In coastal areas of Brazil, the main malaria vector is Anopheles aquasalis, and Plasmodium vivax is responsible for the majority of malaria cases in the Americas. Insects possess a powerful immune system to combat infections. Three pathways control the insect immune response: Toll, IMD, and JAK-STAT. Here we analyze the immune role of the A. aquasalis JAK-STAT pathway after P. vivax infection. Three genes, the transcription factor Signal Transducers and Activators of Transcription (STAT), the regulatory Protein Inhibitors of Activated STAT (PIAS) and the Nitric Oxide Synthase enzyme (NOS) were characterized. Expression of STAT and PIAS was higher in males than females and in eggs and first instar larvae when compared to larvae and pupae. RNA levels for STAT and PIAS increased 24 and 36 hours (h) after P. vivax challenge. NOS transcription increased 36 h post infection (hpi) while this protein was already detected in some midgut epithelial cells 24 hpi. Imunocytochemistry experiments using specific antibodies showed that in non-infected insects STAT and PIAS were found mostly in the fat body, while in infected mosquitoes the proteins were found in other body tissues. The knockdown of STAT by RNAi increased the number of oocysts in the midgut of A. aquasalis. This is the first clear evidence for the involvement of a specific immune pathway in the interaction of the Brazilian malaria vector A. aquasalis with P. vivax, delineating a potential target for the future development of disease controlling strategies.


Subject(s)
Anopheles/immunology , Anopheles/parasitology , Nitric Oxide Synthase/biosynthesis , Plasmodium vivax/immunology , Plasmodium vivax/isolation & purification , Protein Inhibitors of Activated STAT/biosynthesis , STAT Transcription Factors/biosynthesis , Animals , Brazil , Female , Gene Expression Profiling , Gene Knockdown Techniques , Immunohistochemistry , Male , Molecular Sequence Data , Nitric Oxide Synthase/immunology , Protein Inhibitors of Activated STAT/immunology , STAT Transcription Factors/immunology , Sequence Analysis, DNA
14.
PLoS One ; 5(3): e9795, 2010 Mar 22.
Article in English | MEDLINE | ID: mdl-20339545

ABSTRACT

Malaria affects 300 million people worldwide every year and is endemic in 22 countries in the Americas where transmission occurs mainly in the Amazon Region. Most malaria cases in the Americas are caused by Plasmodium vivax, a parasite that is almost impossible to cultivate in vitro, and Anopheles aquasalis is an important malaria vector. Understanding the interactions between this vector and its parasite will provide important information for development of disease control strategies. To this end, we performed mRNA subtraction experiments using A. aquasalis 2 and 24 hours after feeding on blood and blood from malaria patients infected with P. vivax to identify changes in the mosquito vector gene induction that could be important during the initial steps of infection. A total of 2,138 clones of differentially expressed genes were sequenced and 496 high quality unique sequences were obtained. Annotation revealed 36% of sequences unrelated to genes in any database, suggesting that they were specific to A. aquasalis. A high number of sequences (59%) with no matches in any databases were found 24 h after infection. Genes related to embryogenesis were down-regulated in insects infected by P. vivax. Only a handful of genes related to immune responses were detected in our subtraction experiment. This apparent weak immune response of A. aquasalis to P. vivax infection could be related to the susceptibility of this vector to this important human malaria parasite. Analysis of some genes by real time PCR corroborated and expanded the subtraction results. Taken together, these data provide important new information about this poorly studied American malaria vector by revealing differences between the responses of A. aquasalis to P. vivax infection, in relation to better studied mosquito-Plasmodium pairs. These differences may be important for the development of malaria transmission-blocking strategies in the Americas.


Subject(s)
Anopheles/parasitology , Gene Expression Profiling , Gene Expression Regulation , Plasmodium vivax/metabolism , Actins/genetics , Amino Acid Sequence , Animals , Expressed Sequence Tags , Female , Gene Library , Male , Models, Genetic , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Species Specificity
15.
J Med Entomol ; 44(6): 903-14, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18047187

ABSTRACT

Lutzomyia (Nyssomyia) intermedia (Lutz & Neiva 1912) and Lutzomyia (Nyssomyia) whitmani (Antunes & Coutinho 1939) (Diptera: Psychodidae) are vectors of American cutaneous leishmaniasis in several endemic regions of Brazil. We analyzed the external morphological aspects of the immature stages of these two vectors by using scanning electron microscopy. In general, the larval stages of the two species are morphologically similar, although some differences were noted. Detailed examination of the eggs of both species revealed similar exchorionic ornamentations of unconnected parallel ridges. The larval head capsules are well defined, heavily sclerotized, and bear prominent chewing mouthparts. The abdominal segments are easily recognized by the presence of prolegs on their ventral surfaces. The morphology of the anal lobe on the terminal abdominal segment differs between the two species. We found the following three types of sensillae inserted on the antennae: (1) clavate basiconic; (2) small, blunt coeloconic; and (3) multipourous clavate coleoconic. In addition; five subtypes of trichoid sensillae were found on the larval body: (1) long, (2) short, (3) curved long, (4) brush-like, and (5) weakly brush-like. The caudal filaments located on the last abdominal segment were recognized as long trichoid sensillae. We observed pores on the surface of the clavate coelonic sensillae and on the caudal filaments that presumably function as chemoreceptors. The larvae of the two species show similarities in the lobular-form antennae of L1 larvae, which changes to digitiform in second instar (L2), L3, and L4. This study demonstrated that the external surface of the eggs and larvae of Lu. intermedia and Lu. whitmani are morphologically similar, but they can be distinguished by details in the microanatomy observed by scanning electron microscopy.


Subject(s)
Leishmaniasis, Cutaneous/transmission , Psychodidae/ultrastructure , Animals , Insect Vectors , Larva/ultrastructure , Microscopy, Electron, Scanning , Ovum/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...