1
|
Differential expression of Phlebotomus tobbi Adler, Theodor & Lourie, 1930 (Diptera: Psychodidae) genes under different environmental conditions. Acta Trop 2023; 239:106808. [PMID: 36577475 DOI: 10.1016/j.actatropica.2022.106808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/23/2022] [Accepted: 12/24/2022] [Indexed: 12/26/2022]
Abstract
Phlebotomus tobbi is a widely distributed sand fly species in Turkey and is the proven vector of Leishmania infantum and several Phleboviruses. Information regarding the genetic basis of phenotypic plasticity is crucial for managing vector-borne diseases, as the changing environmental conditions have consequences for the survival of arthropods and the disease agents they transmit. However, limited data is available on the impacts of environmental conditions on the traits associated with sand fly survival, reproduction, and vectorial competence. The present study aimed to reveal the changes in the expression levels of three selected P. tobbi genes using laboratory-reared and wild-caught populations. A nervous system protein, Cacophony (PtCac), related to the life history traits of sand flies, and two sand fly salivary protein genes, PtSP32 and PtSP38, influence the infection of the vertebrate hosts, were assessed. Sand flies were maintained at 23 °C and 27 °C in the laboratory to evaluate the relationship between temperature and the expressed phenotypes. Field collections were carried out in three climatically distinct regions of Turkey to establish the regional differences in the gene expression levels of natural P. tobbi populations. In the laboratory, PtCac expression increased with the temperature. However, PtCac expression was negatively correlated with local temperature and humidity conditions. No differences were detected in the PtSP32 gene expression levels of both laboratory-reared and wild-caught females, but a negative correlation was observed with relative humidity in natural populations. Although the expression levels of PtSP38 did not differ among the females collected from distinct regions, a positive correlation was detected in the laboratory-reared colony. The findings indicated that changes in environmental conditions could drive the expression levels of P. tobbi genes, which influence population dynamics and the transmission risk of the disease.
Collapse
|
2
|
Schneider CA, Calvo E, Peterson KE. Arboviruses: How Saliva Impacts the Journey from Vector to Host. Int J Mol Sci 2021; 22:ijms22179173. [PMID: 34502092 PMCID: PMC8431069 DOI: 10.3390/ijms22179173] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/19/2021] [Accepted: 08/22/2021] [Indexed: 12/21/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Christine A. Schneider
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA;
| | - Eric Calvo
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA;
| | - Karin E. Peterson
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA;
- Correspondence:
| |
Collapse
|
3
|
Karmakar S, Nath S, Sarkar B, Chakraborty S, Paul S, Karan M, Pal C. Insect vectors' saliva and gut microbiota as a blessing in disguise: probability versus possibility. Future Microbiol 2021; 16:657-670. [PMID: 34100305 DOI: 10.2217/fmb-2020-0239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Drawing of host blood is a natural phenomenon during the bite of blood-probing insect vectors. Along with the blood meal, the vectors introduce salivary components and a trail of microbiota. In the case of infected vectors, the related pathogen accompanies the aforementioned biological components. In addition to Anopheles gambiae or Anopheles stephensi, the bites of other nonmalarial vectors cannot be ignored in malaria-endemic regions. Similarly, the bite incidence of Phlebotomus papatasi cannot be ignored in visceral leishmaniasis-endemic regions. Even the chances of getting bitten by uninfected vectors are higher than the infected vectors. We have discussed the probability or possibility of uninfected, infected, and/or nonvector's saliva and gut microbiota as a therapeutic option leading to the initial deterrent to pathogen establishment.
Collapse
Affiliation(s)
- Suman Karmakar
- Cellular Immunology & Experimental Therapeutics Laboratory, Department of Zoology, West Bengal State University, Barasat, North 24 Parganas, West Bengal, 700126, India.,Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, Barasat, North 24 Parganas, West Bengal, 700126, India
| | - Supriya Nath
- Cellular Immunology & Experimental Therapeutics Laboratory, Department of Zoology, West Bengal State University, Barasat, North 24 Parganas, West Bengal, 700126, India.,Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, Barasat, North 24 Parganas, West Bengal, 700126, India
| | - Biswajyoti Sarkar
- Cellular Immunology & Experimental Therapeutics Laboratory, Department of Zoology, West Bengal State University, Barasat, North 24 Parganas, West Bengal, 700126, India.,Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, Barasat, North 24 Parganas, West Bengal, 700126, India
| | - Sondipon Chakraborty
- Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, Barasat, North 24 Parganas, West Bengal, 700126, India
| | - Sharmistha Paul
- Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, Barasat, North 24 Parganas, West Bengal, 700126, India
| | - Mintu Karan
- Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, Barasat, North 24 Parganas, West Bengal, 700126, India
| | - Chiranjib Pal
- Cellular Immunology & Experimental Therapeutics Laboratory, Department of Zoology, West Bengal State University, Barasat, North 24 Parganas, West Bengal, 700126, India.,Vector Molecular Biology Laboratory, Department of Zoology, West Bengal State University, Barasat, North 24 Parganas, West Bengal, 700126, India
| |
Collapse
|
4
|
RNA-sequencing of the Nyssomyia neivai sialome: a sand fly-vector from a Brazilian endemic area for tegumentary leishmaniasis and pemphigus foliaceus. Sci Rep 2020; 10:17664. [PMID: 33077743 PMCID: PMC7572365 DOI: 10.1038/s41598-020-74343-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 09/17/2020] [Indexed: 02/07/2023] Open
Abstract
Leishmaniasis encompasses a spectrum of diseases caused by a protozoan belonging to the genus Leishmania. The parasite is transmitted by the bite of sand flies, which inoculate the promastigote forms into the host’s skin while acquiring a blood meal. Nyssomyia neivai is one of the main vectors of tegumentary leishmaniasis (TL) in Brazil. Southeastern Brazil is an endemic region for TL but also overlaps with an endemic focus for pemphigus foliaceus (PF), also known as Fogo Selvagem. Salivary proteins of sand flies, specifically maxadilan and LJM11, have been related to pemphigus etiopathogenesis in the New World, being proposed as an environmental trigger for autoimmunity. We present a comprehensive description of the salivary transcriptome of the N. neivai, using deep sequencing achieved by the Illumina protocol. In addition, we highlight the abundances of several N. neivai salivary proteins and use phylogenetic analysis to compare with Old- and New-World sand fly salivary proteins. The collection of protein sequences associated with the salivary glands of N. neivai can be useful for monitoring vector control strategies as biomarkers of N. neivai, as well as driving vector-vaccine design for leishmaniasis. Additionally, this catalog will serve as reference to screen for possible antigenic peptide candidates triggering anti-Desmoglein-1 autoantibodies.
Collapse
|
5
|
Maryam Ghafari S, Ebrahimi S, Nateghi Rostami M, Bordbar A, Parvizi P. Comparative evaluation of salivary glands proteomes from wild Phlebotomus papatasi-proven vector of zoonotic cutaneous leishmaniasis in Iran. Vet Med Sci 2020; 7:362-369. [PMID: 32969601 PMCID: PMC8025609 DOI: 10.1002/vms3.368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 08/01/2020] [Accepted: 09/09/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Zoonotic Cutaneous Leishmaniasis is increasing in the world and Phlebotomus papatasi as a proven vector was considered in different aspects for disease control. Sandfly saliva contains proteins which provoke host immune system. These proteins are candidates for developing vaccines. OBJECTIVES The main purpose of this research was comparing evaluation of salivary glands proteomes from wild P. papatasi. Extracting these proteins and purifying of original SP15 as inducer agent in vector salivary glands from endemic leishmaniasis foci were other objectives. METHODS Adult sandflies were sampled using aspirators and funnel traps from three endemic foci in 2017-2018. Each pair of salivary glands of unfed females was dissected and proteins were extracted using thermal shocking and sonication methods. Purification was performed through RP-HPLC. All equivalent fractions were added together in order to reach sufficient protein concentration. Protein content and profile determination were examined with SDS-PAGE. RESULTS The protein concentration of whole-salivary glands of specimens was determined approximately 1.6 µg/µl (Isfahan) and 1 µg/µl (Varamin and Kashan). SDS-PAGE revealed 10 distinct bands between 10 and 63 kDa. Analysis of proteomes showed some similarities and differences in the chromatograms of different foci. SDS-PAGE of all collected fractions revealed SP15-like proteins were isolated in 24 min from Varamin, 26 to 30 min from Kashan and 29.4 min from Isfahan and were around 15 kDa. CONCLUSIONS Isolation of salivary components of Iranian wild P. papatasi is very important for finding potential proteins in vaccine development and measuring control strategy of zoonotic cutaneous leishmaniasis in Iran and this could be concluded elsewhere in the world.
Collapse
Affiliation(s)
- Seyedeh Maryam Ghafari
- Molecular Systematics Laboratory, Parasitology Department, Pasteur Institute of Iran, Tehran, Iran
| | - Sahar Ebrahimi
- Molecular Systematics Laboratory, Parasitology Department, Pasteur Institute of Iran, Tehran, Iran
| | | | - Ali Bordbar
- Molecular Systematics Laboratory, Parasitology Department, Pasteur Institute of Iran, Tehran, Iran
| | - Parviz Parvizi
- Molecular Systematics Laboratory, Parasitology Department, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
6
|
Mondragon-Shem K, Wongtrakul-Kish K, Kozak RP, Yan S, Wilson IBH, Paschinger K, Rogers ME, Spencer DIR, Acosta-Serrano A. Insights into the salivary N-glycome of Lutzomyia longipalpis, vector of visceral leishmaniasis. Sci Rep 2020; 10:12903. [PMID: 32737362 PMCID: PMC7395719 DOI: 10.1038/s41598-020-69753-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 07/14/2020] [Indexed: 01/08/2023] Open
Abstract
During Leishmania transmission sand flies inoculate parasites and saliva into the skin of vertebrates. Saliva has anti-haemostatic and anti-inflammatory activities that evolved to facilitate bloodfeeding, but also modulate the host's immune responses. Sand fly salivary proteins have been extensively studied, but the nature and biological roles of protein-linked glycans remain overlooked. Here, we characterised the profile of N-glycans from the salivary glycoproteins of Lutzomyia longipalpis, vector of visceral leishmaniasis in the Americas. In silico predictions suggest half of Lu. longipalpis salivary proteins may be N-glycosylated. SDS-PAGE coupled to LC-MS analysis of sand fly saliva, before and after enzymatic deglycosylation, revealed several candidate glycoproteins. To determine the diversity of N-glycan structures in sand fly saliva, enzymatically released sugars were fluorescently tagged and analysed by HPLC, combined with highly sensitive LC-MS/MS, MALDI-TOF-MS, and exoglycosidase treatments. We found that the N-glycan composition of Lu. longipalpis saliva mostly consists of oligomannose sugars, with Man5GlcNAc2 being the most abundant, and a few hybrid-type species. Interestingly, some glycans appear modified with a group of 144 Da, whose identity has yet to be confirmed. Our work presents the first detailed structural analysis of sand fly salivary glycans.
Collapse
Affiliation(s)
- Karina Mondragon-Shem
- Department of Vector Biology, Liverpool School of Tropical Medicine, Liverpool, L3 5QA, UK
| | - Katherine Wongtrakul-Kish
- Ludger Ltd., Culham Science Centre, Oxfordshire, OX14 3EB, UK
- Australian Research Council Centre of Excellence for Nanoscale Biophotonics, Macquarie University, Sydney, Australia
| | | | - Shi Yan
- Department of Chemistry, University of Natural Resources and Life Sciences, 1190, Vienna, Austria
- Institute of Parasitology, Department of Pathobiology, University of Veterinary Medicine, A-1210, Vienna, Austria
| | - Iain B H Wilson
- Department of Chemistry, University of Natural Resources and Life Sciences, 1190, Vienna, Austria
| | - Katharina Paschinger
- Department of Chemistry, University of Natural Resources and Life Sciences, 1190, Vienna, Austria
| | - Matthew E Rogers
- Department of Disease Control, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | | | - Alvaro Acosta-Serrano
- Department of Vector Biology, Liverpool School of Tropical Medicine, Liverpool, L3 5QA, UK.
| |
Collapse
|
7
|
Veysi A, Mahmoudi AR, Yaghoobi-Ershadi MR, Jeddi-Tehrani M, Rassi Y, Zahraei-Ramazani A, Hosseini-Vasoukolaei N, Zareie B, Khamesipour A, Akhavan AA. Human immune response to Phlebotomus sergenti salivary gland antigens in a leishmaniasis-endemic focus in Iran. Pathog Glob Health 2020; 114:323-332. [PMID: 32643589 DOI: 10.1080/20477724.2020.1789399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Salivary proteins specific antibodies have been shown to be useful biomarkers of exposure to sand fly bites. This study aimed to investigate the level, duration, and dynamics of the human immune response against the SGL of Phlebotomus sergenti Parrot, 1917 (Diptera: Psychodidae), and to assess the immunoreactivity of human sera with SGL components in an endemic area of anthroponotic cutaneous leishmaniasis (ACL) in Iran. The study was carried out in 2-phase; longitudinal and cross-sectional. Sand flies were collected monthly from indoors and outdoors. In the longitudinal study, sera from healthy volunteers were collected monthly, and in the cross-sectional study, sera from healthy volunteers and patients with ACL lesion/s, were collected for immunoassay studies. The level of anti-P. sergenti saliva IgG was detected using the ELISA. Immunoreactivity of individual human sera with saliva components was also assessed by western blotting. Phlebotomus sergenti was the predominant sand fly species in the study area. The maximum and minimum percentages of IgG responses were seen in October (66%) and March (29%), respectively. Additionally, the cross-sectional study showed that 59.3% of the healthy volunteers and 80% of the patients were IgG positive. The antibody response against P. sergenti salivary gland was high during the sand fly active season and declined by the end of the activity of the vectors. Antibody response against the SGL components of P. sergenti was transient and individual-specific. Some individuals shared a strong reaction against certain individual antigens, which could be considered as vector exposure markers for further investigation. LIST OF ABBREVIATIONS ELISA: Enzyme-Linked Immunosorbent Assay; SDS PAGE: Sodium Dodecyl Sulfate-Polyacrylamide Gel Electrophoresis; SGL: Salivary Gland Lysate; ACL: Anthroponotic Cutaneous Leishmaniasis; PBS: Phosphate Buffered Saline; BCA: Bicinchoninic Acid; PBS-T: Phosphate Buffered Saline Tween; FBS: Fetal Bovine Serum; HRP: Horseradish Peroxidase; TMB: 3,3',5,5'-Tetramethylbenzidine; PVDF: Polyvinylidene Difluoride; SGA: Salivary Gland Antigens; OD: Optical Density; KDa: Kilodalton; VL: Visceral Leishmaniasis; CL: Cutaneous Leishmaniasis; SGs: Salivary glands.
Collapse
Affiliation(s)
- Arshad Veysi
- Zoonoses Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences , Sanandaj, Iran.,Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences , Tehran, Iran
| | - Ahmad Reza Mahmoudi
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences , Tehran, Iran
| | - Mohammad Reza Yaghoobi-Ershadi
- Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences , Tehran, Iran
| | - Mahmood Jeddi-Tehrani
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR , Tehran, Iran
| | - Yavar Rassi
- Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences , Tehran, Iran
| | - Alireza Zahraei-Ramazani
- Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences , Tehran, Iran
| | - Nasibeh Hosseini-Vasoukolaei
- Department of Medical Entomology and Vector Control, Health Sciences Research Center, Addiction Institute, Mazandaran University of Medical Sciences , Sari, Iran
| | - Bushra Zareie
- Department of Epidemiology, School of Public Health, Hamadan University of Medical Sciences , Hamadan, Iran
| | - Ali Khamesipour
- Center for Research and Training in Skin Diseases and Leprosy, Tehran University of Medical Sciences , Tehran, Iran
| | - Amir Ahmad Akhavan
- Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences , Tehran, Iran
| |
Collapse
|
8
|
Oliveira F, Giorgobiani E, Guimarães-Costa AB, Abdeladhim M, Oristian J, Tskhvaradze L, Tsertsvadze N, Zakalashvili M, Valenzuela JG, Kamhawi S. Immunity to vector saliva is compromised by short sand fly seasons in endemic regions with temperate climates. Sci Rep 2020; 10:7990. [PMID: 32409684 PMCID: PMC7224377 DOI: 10.1038/s41598-020-64820-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 04/22/2020] [Indexed: 01/26/2023] Open
Abstract
Individuals exposed to sand fly bites develop humoral and cellular immune responses to sand fly salivary proteins. Moreover, cellular immunity to saliva or distinct salivary proteins protects against leishmaniasis in various animal models. In Tbilisi, Georgia, an endemic area for visceral leishmaniasis (VL), sand flies are abundant for a short period of ≤3 months. Here, we demonstrate that humans and dogs residing in Tbilisi have little immunological memory to saliva of P. kandelakii, the principal vector of VL. Only 30% of humans and 50% of dogs displayed a weak antibody response to saliva after the end of the sand fly season. Likewise, their peripheral blood mononuclear cells mounted a negligible cellular immune response after stimulation with saliva. RNA seq analysis of wild-caught P. kandelakii salivary glands established the presence of a typical salivary repertoire that included proteins commonly found in other sand fly species such as the yellow, SP15 and apyrase protein families. This indicates that the absence of immunity to P. kandelakii saliva in humans and dogs from Tbilisi is probably caused by insufficient exposure to sand fly bites. This absence of immunity to vector saliva will influence the dynamics of VL transmission in Tbilisi and other endemic areas with brief sand fly seasons.
Collapse
Affiliation(s)
- Fabiano Oliveira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, 20852, USA
| | - Ekaterina Giorgobiani
- R. G. Lugar Center for Public Health Research, National Center for Disease Control and Public Health (NCDC), Kakheti Highway 99, 0198, Tbilisi, Georgia
| | - Anderson B Guimarães-Costa
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, 20852, USA
| | - Maha Abdeladhim
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, 20852, USA
| | - James Oristian
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, 20852, USA
| | - Lamzira Tskhvaradze
- R. G. Lugar Center for Public Health Research, National Center for Disease Control and Public Health (NCDC), Kakheti Highway 99, 0198, Tbilisi, Georgia
| | - Nikoloz Tsertsvadze
- R. G. Lugar Center for Public Health Research, National Center for Disease Control and Public Health (NCDC), Kakheti Highway 99, 0198, Tbilisi, Georgia
| | - Mariam Zakalashvili
- R. G. Lugar Center for Public Health Research, National Center for Disease Control and Public Health (NCDC), Kakheti Highway 99, 0198, Tbilisi, Georgia
| | - Jesus G Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, 20852, USA.
| | - Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, 20852, USA.
| |
Collapse
|
9
|
Polanska N, Ishemgulova A, Volfova V, Flegontov P, Votypka J, Yurchenko V, Volf P. Sergentomyia schwetzi: Salivary gland transcriptome, proteome and enzymatic activities in two lineages adapted to different blood sources. PLoS One 2020; 15:e0230537. [PMID: 32208452 PMCID: PMC7092997 DOI: 10.1371/journal.pone.0230537] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 03/02/2020] [Indexed: 11/18/2022] Open
Abstract
During the blood feeding, sand fly females inject saliva containing immunomodulatory and anti-haemostatic molecules into their vertebrate hosts. The saliva composition is species-specific, likely due to an adaptation to particular haemostatic pathways of their preferred host. Research on sand fly saliva is limited to the representatives of two best-studied genera, Phlebotomus and Lutzomyia. Although the members of the genus Sergentomyia are highly abundant in many areas in the Old World, their role in human disease transmission remains uncertain. Most Sergentomyia spp. preferentially attack various species of reptiles, but feeding on warm-blooded vertebrates, including humans and domestic animals, has been repeatedly described, especially for Sergentomyia schwetzi, of which salivary gland transcriptome and proteome is analyzed in the current study. Illumina RNA sequencing and de novo assembly of the reads and their annotation revealed 17,293 sequences homologous to other arthropods’ proteins. In the sialome, all proteins typical for sand fly saliva were identified–antigen 5-related, lufaxin, yellow-related, PpSP15-like, D7-related, ParSP25-like, and silk proteins, as well as less frequent salivary proteins included 71kDa-like, ParSP80-like, SP16-like, and ParSP17-like proteins. Salivary enzymes include apyrase, hyaluronidase, endonuclease, amylase, lipase A2, adenosine deaminase, pyrophosphatase, 5’nucleotidase, and ribonuclease. Proteomics analysis of salivary glands identified 631 proteins, 81 of which are likely secreted into the saliva. We also compared two S. schwetzi lineages derived from the same origin. These lineages were adapted for over 40 generations for blood feeding either on mice (S-M) or geckos (S-G), two vertebrate hosts with different haemostatic mechanisms. Altogether, 20 and 40 annotated salivary transcripts were up-regulated in the S-M and S-G lineage, respectively. Proteomic comparison revealed ten salivary proteins more abundant in the lineage S-M, whereas 66 salivary proteins were enriched in the lineage S-G. No difference between lineages was found for apyrase activity; contrarily the hyaluronidase activity was significantly higher in the lineage feeding on mice.
Collapse
Affiliation(s)
- Nikola Polanska
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
- * E-mail:
| | - Aygul Ishemgulova
- Life Science Research Centre, Faculty of Science, University of Ostrava, Ostrava, Czech Republic
| | - Vera Volfova
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Pavel Flegontov
- Life Science Research Centre, Faculty of Science, University of Ostrava, Ostrava, Czech Republic
- Biology Centre, Institute of Parasitology, Czech Academy of Sciences, České Budějovice (Budweis), Czech Republic
| | - Jan Votypka
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Vyacheslav Yurchenko
- Life Science Research Centre, Faculty of Science, University of Ostrava, Ostrava, Czech Republic
- Biology Centre, Institute of Parasitology, Czech Academy of Sciences, České Budějovice (Budweis), Czech Republic
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector Borne Diseases, Sechenov University, Moscow, Russia
| | - Petr Volf
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| |
Collapse
|
10
|
Sumova P, Sima M, Kalouskova B, Polanska N, Vanek O, Oliveira F, Valenzuela JG, Volf P. Amine-binding properties of salivary yellow-related proteins in phlebotomine sand flies. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2019; 115:103245. [PMID: 31604119 DOI: 10.1016/j.ibmb.2019.103245] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 06/10/2023]
Abstract
The amine-binding properties of sand fly salivary yellow-related proteins (YRPs) were described only in Lutzomyia longipalpis sand flies. Here, we experimentally confirmed the kratagonist function of YRPs in the genus Phlebotomus. We utilized microscale thermophoresis technique to determine the amine-binding properties of YRPs in saliva of Phlebotomus perniciosus and P. orientalis, the Old-World vectors of visceral leishmaniases causative agents. Expressed and purified YRPs from three different sand fly species were tested for their interactions with various biogenic amines, including serotonin, histamine and catecholamines. Using the L. longipalpis YRP LJM11 as a control, we have demonstrated the comparability of the microscale thermophoresis method with conventional isothermal titration calorimetry described previously. By homology in silico modeling, we predicted the surface charge and both amino acids and hydrogen bonds of the amine-binding motifs to influence the binding affinities between closely related YRPs. All YRPs tested bound at least two biogenic amines, while the affinities differ both among and within species. Low affinity was observed for histamine. The salivary recombinant proteins rSP03B (P. perniciosus) and rPorASP4 (P. orientalis) showed high-affinity binding of serotonin, suggesting their capability to facilitate inhibition of the blood vessel contraction and platelet aggregation.
Collapse
Affiliation(s)
- Petra Sumova
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic.
| | - Michal Sima
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Barbora Kalouskova
- Department of Biochemistry, Faculty of Science, Charles University, Prague, Czech Republic
| | - Nikola Polanska
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Ondrej Vanek
- Department of Biochemistry, Faculty of Science, Charles University, Prague, Czech Republic
| | - Fabiano Oliveira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Jesus G Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Petr Volf
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| |
Collapse
|
11
|
An Evolutionary Perspective of Dopachrome Tautomerase Enzymes in Metazoans. Genes (Basel) 2019; 10:genes10070495. [PMID: 31261784 PMCID: PMC6678240 DOI: 10.3390/genes10070495] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/19/2019] [Accepted: 06/24/2019] [Indexed: 12/19/2022] Open
Abstract
Melanin plays a pivotal role in the cellular processes of several metazoans. The final step of the enzymically-regulated melanin biogenesis is the conversion of dopachrome into dihydroxyindoles, a reaction catalyzed by a class of enzymes called dopachrome tautomerases. We traced dopachrome tautomerase (DCT) and dopachrome converting enzyme (DCE) genes throughout metazoans and we could show that only one class is present in most of the phyla. While DCTs are typically found in deuterostomes, DCEs are present in several protostome phyla, including arthropods and mollusks. The respective DCEs belong to the yellow gene family, previously reported to be taxonomically restricted to insects, bacteria and fungi. Mining genomic and transcriptomic data of metazoans, we updated the distribution of DCE/yellow genes, demonstrating their presence and active expression in most of the lophotrochozoan phyla as well as in copepods (Crustacea). We have traced one intronless DCE/yellow gene through most of the analyzed lophotrochozoan genomes and we could show that it was subjected to genomic diversification in some species, while it is conserved in other species. DCE/yellow was expressed in most phyla, although it showed tissue specific expression patterns. In the parasitic copepod Mytilicola intestinalis DCE/yellow even belonged to the 100 most expressed genes. Both tissue specificity and high expression suggests that diverse functions of this gene family also evolved in other phyla apart from insects.
Collapse
|
12
|
Al-Salem WS, Solórzano C, Weedall GD, Dyer NA, Kelly-Hope L, Casas-Sánchez A, Alraey Y, Alyamani EJ, Halliday A, Balghonaim SM, Alsohibany KS, Alzeyadi Z, Alzahrani MH, Al-Shahrani AM, Assiri AM, Memish Z, Acosta-Serrano Á. Old World cutaneous leishmaniasis treatment response varies depending on parasite species, geographical location and development of secondary infection. Parasit Vectors 2019; 12:195. [PMID: 31046820 PMCID: PMC6498568 DOI: 10.1186/s13071-019-3453-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/20/2019] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND In the Kingdom of Saudi Arabia (KSA), Leishmania major and L. tropica are the main causative agents of Old World cutaneous leishmaniasis (CL). The national CL treatment regimen consists of topical 1% clotrimazole/2% fusidic acid cream followed by 1-2 courses of intralesional sodium stibogluconate (SSG); however, treatment efficacy is highly variable and the reasons for this are not well understood. In this study, we present a complete epidemiological map of CL and determined the efficacy of the standard CL treatment regime in several endemic regions of KSA. RESULTS Overall, three quarters of patients in all CL-endemic areas studied responded satisfactorily to the current treatment regime, with the remaining requiring only an extra course of SSG. The majority of unresponsive cases were infected with L. tropica. Furthermore, the development of secondary infections (SI) around or within the CL lesion significantly favoured the treatment response of L. major patients but had no effect on L. tropica cases. CONCLUSIONS The response of CL patients to a national treatment protocol appears to depend on several factors, including Leishmania parasite species, geographical location and occurrences of SI. Our findings suggest there is a need to implement alternative CL treatment protocols based on these parameters.
Collapse
Affiliation(s)
- Waleed S. Al-Salem
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
- Present Address: National Centre for Tropical Diseases, Saudi Ministry of Health, Riyadh, Kingdom of Saudi Arabia
| | - Carla Solórzano
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Gareth D. Weedall
- Faculty of Sciences, Liverpool John Moores University, Liverpool, UK
| | - Naomi A. Dyer
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Louise Kelly-Hope
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Aitor Casas-Sánchez
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Yasser Alraey
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Essam J. Alyamani
- National Center for Biotechnology, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Alice Halliday
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | | | | | - Zeyad Alzeyadi
- Antimicrobial Research Centre, University of Leeds, Leeds, UK
| | | | | | | | - Ziad Memish
- Saudi Ministry of Health, Riyadh, Kingdom of Saudi Arabia
| | - Álvaro Acosta-Serrano
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
- Department of Vector Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| |
Collapse
|
13
|
Sumova P, Sima M, Spitzova T, Osman ME, Guimaraes-Costa AB, Oliveira F, Elnaiem DEA, Hailu A, Warburg A, Valenzuela JG, Volf P. Human antibody reaction against recombinant salivary proteins of Phlebotomus orientalis in Eastern Africa. PLoS Negl Trop Dis 2018; 12:e0006981. [PMID: 30513081 PMCID: PMC6279015 DOI: 10.1371/journal.pntd.0006981] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/07/2018] [Indexed: 01/01/2023] Open
Abstract
Background Phlebotomus orientalis is a vector of Leishmania donovani, the causative agent of life threatening visceral leishmaniasis spread in Eastern Africa. During blood-feeding, sand fly females salivate into the skin of the host. Sand fly saliva contains a large variety of proteins, some of which elicit specific antibody responses in the bitten hosts. To evaluate the exposure to sand fly bites in human populations from disease endemic areas, we tested the antibody reactions of volunteers' sera against recombinant P. orientalis salivary antigens. Methodology/Principal findings Recombinant proteins derived from sequence data on P. orientalis secreted salivary proteins, were produced using either bacterial (five proteins) or mammalian (four proteins) expression systems and tested as antigens applicable for detection of anti-P. orientalis IgG in human sera. Using these recombinant proteins, human sera from Sudan and Ethiopia, countries endemic for visceral leishmaniasis, were screened by ELISA and immunoblotting to identify the potential markers of exposure to P. orientalis bites. Two recombinant proteins; mAG5 and mYEL1, were identified as the most promising antigens showing high correlation coefficients as well as good specificity in comparison to the whole sand fly salivary gland homogenate. Combination of both proteins led to a further increase of correlation coefficients as well as both positive and negative predictive values of P. orientalis exposure. Conclusions/Significance This is the first report of screening human sera for anti-P. orientalis antibodies using recombinant salivary proteins. The recombinant salivary proteins mYEL1 and mAG5 proved to be valid antigens for screening human sera from both Sudan and Ethiopia for exposure to P. orientalis bites. The utilization of equal amounts of these two proteins significantly increased the capability to detect anti-P. orientalis antibody responses. Hosts repeatedly bitten by phlebotomine sand flies develop species-specific antibody responses against certain sand fly salivary antigens. Salivary gland homogenate (SGH) is frequently used to evaluate the levels of this antibody response in host. However, SGH is less suitable for large-scale studies, since obtaining sufficient numbers of salivary glands is labor intensive and requires expertise in dissection. To replace SGH as antigen to screen for exposure to sand fly bites, specific recombinant salivary antigens were utilized. Our study assessed the human antibody reactions against recombinant salivary proteins of Phlebotomus orientalis. This sand fly species is a vector of Leishmania donovani, the causative agent of severe visceral leishmaniasis in Eastern Africa. To identify valid markers of exposure to P. orientalis in humans, we screened for anti-P. orientalis antibody responses in serum samples from individuals residing in Sudan and Ethiopia. We tested nine recombinant salivary antigens and found a combination of yellow-related protein (mYEL1) and antigen 5-related protein (mAG5) the best marker of exposure, accurately correlating with the levels of exposure to P. orientalis bites as determined using SGH. Thus the combination mYEL1+ mAG5 can comprise a useful epidemiological tool to determine levels of exposure to P. orientalis in populations living in endemic areas of Eastern Africa, which could help in monitoring the distribution of P. orientalis and therefore assessing suitable anti-vector campaigns.
Collapse
Affiliation(s)
- Petra Sumova
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
- * E-mail:
| | - Michal Sima
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Tatiana Spitzova
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Maha E. Osman
- Commission for Biotechnology and Genetic Engineering, National Centre for Research, Khartoum, Sudan
| | - Anderson B. Guimaraes-Costa
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Fabiano Oliveira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Dia-Eldin A. Elnaiem
- Department of Natural Sciences, University of Maryland Eastern Shore, Princess Anne, Maryland, United States of America
| | - Asrat Hailu
- Department of Microbiology, Immunology and Parasitology, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Alon Warburg
- Department of Microbiology and Molecular Genetics, The Kuvin Centre for the Study of Infectious and Tropical Diseases, The Hebrew University—Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jesus G. Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Petr Volf
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| |
Collapse
|
14
|
Volfova V, Volf P. The salivary hyaluronidase and apyrase of the sand fly Sergentomyia schwetzi (Diptera, Psychodidae). INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2018; 102:67-74. [PMID: 30273670 DOI: 10.1016/j.ibmb.2018.09.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 09/26/2018] [Accepted: 09/27/2018] [Indexed: 06/08/2023]
Abstract
Current knowledge of sand fly salivary components has been based solely on Lutzomyia and Phlebotomus species which feed mainly on mammals; their hyaluronidases and apyrases were demonstrated to significantly affect blood meal intake and transmission of vector-borne pathogens. Members of the third sand fly genus Sergentomyia preferentially feed on reptiles but some of them are considered as Leishmania and arboviruses vectors; however, nothing is known about their salivary components that might be relevant for pathogens transmission. Here, marked hyaluronidase and apyrase activities were demonstrated in the saliva of a Sergentomyia schwetzi colony maintained on geckos. Hyaluronidase of S. schwetzi cleaved hyaluronan as the prominent substrate, and was active over a broad pH range from 4.0 to 8.0, with a sharp peak at pH 5.0. SDS PAGE zymography demonstrated the monomeric character of the enzyme, which remained active in reducing conditions. The apparent molecular weight of 43 kDa was substantially lower than in any sand fly species tested so far and may indicate relatively low grade of the glycosylation of the enzyme. The apyrase of S. schwetzi was typical strictly Ca2+ dependent Cimex-family apyrase. It was active over a pH range from 6.5 to 9.0, with a peak of activity at pH 8.5, and had an ATPase/ADPase ratio of 0.9. The apyrase activity increased during the first 3 days post-emergence, then reached a plateau and remained relatively constant until day 8. In comparison with a majority of Phlebotomus and Lutzomyia species tested to date, both the hyaluronidase and apyrase activities of S. schwetzi were relatively low, which may reflect an adaptation of this sand fly to blood feeding on non-mammalian hosts.
Collapse
Affiliation(s)
- Vera Volfova
- Department of Parasitology, Faculty of Science, Charles University, Vinicna 7, Prague 2, 128 44, Czech Republic
| | - Petr Volf
- Department of Parasitology, Faculty of Science, Charles University, Vinicna 7, Prague 2, 128 44, Czech Republic.
| |
Collapse
|
15
|
Martin-Martin I, Aryan A, Meneses C, Adelman ZN, Calvo E. Optimization of sand fly embryo microinjection for gene editing by CRISPR/Cas9. PLoS Negl Trop Dis 2018; 12:e0006769. [PMID: 30180160 PMCID: PMC6150542 DOI: 10.1371/journal.pntd.0006769] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 09/21/2018] [Accepted: 08/20/2018] [Indexed: 11/26/2022] Open
Abstract
Background Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9 technology has rapidly emerged as a very effective tool for gene editing. Although great advances on gene editing in the medical entomology field have arisen, no attempts of gene editing have been reported in sand flies, the vectors of Leishmaniasis. Methodology/Principal findings Here, we described a detailed protocol for sand fly embryo microinjection taking into consideration the sand fly life cycle, and manipulation and oviposition requirements of this non-model organism. Following our microinjection protocol, a hatching rate of injected embryos of 11.90%-14.22% was achieved, a rate consistent with other non-model organism dipterans such as mosquitoes. Essential factors for the adaptation of CRISPR/Cas9 technology to the sand fly field were addressed including the selection of a target gene and the design and production of sgRNA. An in vitro cleavage assay was optimized to test the activity of each sgRNA and a protocol for Streptococcus pyogenes Cas9 (spCas9) protein expression and purification was described. Relevant considerations for a successful gene editing in the sand fly such as specifics of embryology and double-stranded break DNA repair mechanisms were discussed. Conclusion and significance The step-by-step methodology reported in this article will be of significant use for setting up a sand fly embryo microinjection station for the incorporation of CRISPR/Cas9 technology in the sand fly field. Gene editing strategies used in mosquitoes and other model insects have been adapted to work with sand flies, providing the tools and relevant information for adapting gene editing techniques to the vectors of Leishmaniasis. Gene editing in sand flies will provide essential information on the biology of these vectors of medical and veterinary relevance and will rise a better understanding of vector-parasite-host interactions. The CRISPR/Cas9 system, based on the adaptive immune system in bacteria and archaea against viral infections, has been adapted and has rapidly emerged as a very effective genetic engineering tool in many organisms. Although great advances on gene editing in the medical entomology field have arisen, no attempts have been reported in sand flies, the vectors of Leishmania spp. Leishmaniasis is one of the most neglected parasitic diseases with twelve million people affected worldwide. Despite their importance as disease vectors, sand fly genetics and molecular studies are limited when compared to other insects. In this article, gene editing strategies used in mosquitoes and other model insects have been adapted to work with sand flies, providing the tools and relevant information for adapting embryo microinjection techniques to sand flies, an essential step in a successful gene editing experiment. We believe gene editing in sand flies will provide essential information of medical and veterinary relevance on the biology of these vectors, and will further a better understanding of vector-parasite-host interactions.
Collapse
Affiliation(s)
- Ines Martin-Martin
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Azadeh Aryan
- Department of Entomology and Fralin Life Science Institute, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Claudio Meneses
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Zach N. Adelman
- Department of Entomology and Agrilife Research, Texas A&M University, College Station, Texas, United States of America
| | - Eric Calvo
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
- * E-mail:
| |
Collapse
|
16
|
Coutinho-Abreu IV, Valenzuela JG. Comparative Evolution of Sand Fly Salivary Protein Families and Implications for Biomarkers of Vector Exposure and Salivary Vaccine Candidates. Front Cell Infect Microbiol 2018; 8:290. [PMID: 30211125 PMCID: PMC6123390 DOI: 10.3389/fcimb.2018.00290] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 07/30/2018] [Indexed: 11/25/2022] Open
Abstract
Sand fly salivary proteins that produce a specific antibody response in humans and animal reservoirs have been shown to be promising biomarkers of sand fly exposure. Furthermore, immunity to sand fly salivary proteins were shown to protect rodents and non-human primates against Leishmania infection. We are missing critical information regarding the divergence amongst sand fly salivary proteins from different sand fly vectors, a knowledge that will support the search of broad or specific salivary biomarkers of vector exposure and those for vaccines components against leishmaniasis. Here, we compare the molecular evolution of the salivary protein families in New World and Old World sand flies from 14 different sand fly vectors. We found that the protein families unique to OW sand flies are more conserved than those unique to NW sand flies regarding both sequence polymorphisms and copy number variation. In addition, the protein families unique to OW sand flies do not display as many conserved cysteine residues as the one unique to the NW group (28.5% in OW vs. 62.5% in NW). Moreover, the expression of specific protein families is restricted to the salivary glands of unique sand fly taxon. For instance, the ParSP15 family is unique to the Larroussius subgenus whereas phospholipase A2 is only expressed in member of Larroussius and Adlerius subgenera. The SP2.5-like family is only expressed in members of the Phlebotomus and Paraphlebotomus subgenera. The sequences shared between OW and NW sand flies have diverged at similar rates (38.7 and 45.3% amino acid divergence, respectively), yet differences in gene copy number were evident across protein families and sand fly species. Overall, this comparative analysis sheds light on the different modes of sand fly salivary protein family divergence. Also, it informs which protein families are unique and conserved within taxon for the choice of taxon-specific biomarkers of vector exposure, as well as those families more conserved across taxa to be used as pan-specific vaccines for leishmaniasis.
Collapse
Affiliation(s)
- Iliano V Coutinho-Abreu
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Jesus G Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| |
Collapse
|
17
|
Immunity to LuloHya and Lundep, the salivary spreading factors from Lutzomyia longipalpis, protects against Leishmania major infection. PLoS Pathog 2018; 14:e1007006. [PMID: 29723281 PMCID: PMC5953502 DOI: 10.1371/journal.ppat.1007006] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 05/15/2018] [Accepted: 04/03/2018] [Indexed: 01/04/2023] Open
Abstract
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.
Collapse
|
18
|
Ribeiro JMC, Martin-Martin I, Moreira FR, Bernard KA, Calvo E. A deep insight into the male and female sialotranscriptome of adult Culex tarsalis mosquitoes. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2018; 95:1-9. [PMID: 29526772 PMCID: PMC5927831 DOI: 10.1016/j.ibmb.2018.03.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 02/20/2018] [Accepted: 03/01/2018] [Indexed: 06/09/2023]
Abstract
Previously, a Sanger-based sialotranscriptome analysis of adult female Culex tarsalis was published based on ∼2000 ESTs. During the elapsed 7.5 years, pyrosequencing has been discontinued and Illumina sequences have increased considerable in size and decreased in price. We here report an Illumina-based sialotranscriptome that allowed finding the missing apyrase from the salivary transcriptome of C. tarsalis, to determine several full-length members of the 34-62 kDa family, when a single EST has been found previously, in addition to identifying many salivary families with lower expression levels that were not detected previously. The use of multiple libraries including salivary glands and carcasses from male and female organisms allowed for an unprecedented insight into the tissue specificity of transcripts, and in this particular case permitting identification of transcripts putatively associated with blood feeding, when exclusive of female salivary glands, or associated with sugar feeding, when transcripts are found upregulated in both male and female glands.
Collapse
Affiliation(s)
- José M C Ribeiro
- Section of Vector Biology, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, 12735 Twinbrook Parkway Room 2E32D, Rockville, MD, 20852, United States.
| | - Ines Martin-Martin
- Section of Vector Biology, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, 12735 Twinbrook Parkway Room 2E32D, Rockville, MD, 20852, United States
| | - Fernando R Moreira
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Dr., Madison, WI, 53706, United States
| | - Kristen A Bernard
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Dr., Madison, WI, 53706, United States
| | - Eric Calvo
- Section of Vector Biology, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, 12735 Twinbrook Parkway Room 2E32D, Rockville, MD, 20852, United States
| |
Collapse
|
19
|
Kato H, Jochim RC, Gomez EA, Tsunekawa S, Valenzuela JG, Hashiguchi Y. Salivary gland transcripts of the kissing bug, Panstrongylus chinai, a vector of Chagas disease. Acta Trop 2017; 174:122-129. [PMID: 28690145 DOI: 10.1016/j.actatropica.2017.06.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 06/15/2017] [Accepted: 06/20/2017] [Indexed: 10/19/2022]
Abstract
The saliva of hematophagous arthropods injected during blood feeding contains potent pharmacologically active components to counteract the host hemostatic and inflammatory systems. In the present study, dominant salivary gland transcripts of Panstrongylus chinai, a vector of Chagas disease, were analyzed by sequencing randomly selected clones of the salivary gland cDNA library. This analysis showed that 56.5% of the isolated transcripts coded for putative secreted proteins, of which 73.7% coded for proteins belonging to the lipocalin family. The most abundant transcript of lipocalin family proteins was a homologue of pallidipin 2, an inhibitor of collagen-induced platelet aggregation of Triatoma pallidipennis. In addition, homologues of triafestin, an inhibitor of the kallikrein-kinin system of T. infestans, were identified as the dominant transcript. Other salivary transcripts encoding lipocalin family proteins had homology to triplatin (an inhibitor of platelet aggregation) and others with unknown function. Other than lipocalin family proteins, homologues of a Kazal-type serine protease inhibitor (putative anticoagulant), a hemolysin-like protein (unknown function), inositol polyphosphate 5-related protein (a regulator of membrane phosphoinositide), antigen 5-related protein (unknown function) and apyrase (platelet aggregation inhibitor) were identified.
Collapse
|
20
|
Lestinova T, Rohousova I, Sima M, de Oliveira CI, Volf P. Insights into the sand fly saliva: Blood-feeding and immune interactions between sand flies, hosts, and Leishmania. PLoS Negl Trop Dis 2017; 11:e0005600. [PMID: 28704370 PMCID: PMC5509103 DOI: 10.1371/journal.pntd.0005600] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Leishmaniases are parasitic diseases present worldwide that are transmitted to the vertebrate host by the bite of an infected sand fly during a blood feeding. Phlebotomine sand flies inoculate into the mammalian host Leishmania parasites embedded in promastigote secretory gel (PSG) with saliva, which is composed of a diverse group of molecules with pharmacological and immunomodulatory properties. Methods and findings In this review, we focus on 3 main aspects of sand fly salivary molecules: (1) structure and composition of salivary glands, including the properties of salivary molecules related to hemostasis and blood feeding, (2) immunomodulatory properties of salivary molecules and the diverse impacts of these molecules on leishmaniasis, ranging from disease exacerbation to vaccine development, and (3) use of salivary molecules for field applications, including monitoring host exposure to sand flies and the risk of Leishmania transmission. Studies showed interesting differences between salivary proteins of Phlebotomus and Lutzomyia species, however, no data were ever published on salivary proteins of Sergentomyia species. Conclusions In the last 15 years, numerous studies have characterized sand fly salivary proteins and, in parallel, have addressed the impact of such molecules on the biology of the host–sand fly–parasite interaction. The results obtained shall pave the way for the development of field-application tools that could contribute to the management of leishmaniasis in endemic areas.
Collapse
Affiliation(s)
- Tereza Lestinova
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
- * E-mail:
| | - Iva Rohousova
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Michal Sima
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | | | - Petr Volf
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| |
Collapse
|
21
|
Sima M, Novotny M, Pravda L, Sumova P, Rohousova I, Volf P. The Diversity of Yellow-Related Proteins in Sand Flies (Diptera: Psychodidae). PLoS One 2016; 11:e0166191. [PMID: 27812196 PMCID: PMC5094789 DOI: 10.1371/journal.pone.0166191] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 10/24/2016] [Indexed: 01/06/2023] Open
Abstract
Yellow-related proteins (YRPs) present in sand fly saliva act as affinity binders of bioamines, and help the fly to complete a bloodmeal by scavenging the physiological signals of damaged cells. They are also the main antigens in sand fly saliva and their recombinant form is used as a marker of host exposure to sand flies. Moreover, several salivary proteins and plasmids coding these proteins induce strong immune response in hosts bitten by sand flies and are being used to design protecting vaccines against Leishmania parasites. In this study, thirty two 3D models of different yellow-related proteins from thirteen sand fly species of two genera were constructed based on the known protein structure from Lutzomyia longipalpis. We also studied evolutionary relationships among species based on protein sequences as well as sequence and structural variability of their ligand-binding site. All of these 33 sand fly YRPs shared a similar structure, including a unique tunnel that connects the ligand-binding site with the solvent by two independent paths. However, intraspecific modifications found among these proteins affects the charges of the entrances to the tunnel, the length of the tunnel and its hydrophobicity. We suggest that these structural and sequential differences influence the ligand-binding abilities of these proteins and provide sand flies with a greater number of YRP paralogs with more nuanced answers to bioamines. All these characteristics allow us to better evaluate these proteins with respect to their potential use as part of anti-Leishmania vaccines or as an antigen to measure host exposure to sand flies.
Collapse
Affiliation(s)
- Michal Sima
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
- * E-mail:
| | - Marian Novotny
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Lukas Pravda
- CEITEC—Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Petra Sumova
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Iva Rohousova
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Petr Volf
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| |
Collapse
|
22
|
Molecular Diversity between Salivary Proteins from New World and Old World Sand Flies with Emphasis on Bichromomyia olmeca, the Sand Fly Vector of Leishmania mexicana in Mesoamerica. PLoS Negl Trop Dis 2016; 10:e0004771. [PMID: 27409591 PMCID: PMC4943706 DOI: 10.1371/journal.pntd.0004771] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 05/21/2016] [Indexed: 01/30/2023] Open
Abstract
Background Sand fly saliva has been shown to have proteins with potent biological activities, salivary proteins that can be used as biomarkers of vector exposure, and salivary proteins that are candidate vaccines against different forms of leishmaniasis. Sand fly salivary gland transcriptomic approach has contributed significantly to the identification and characterization of many of these salivary proteins from important Leishmania vectors; however, sand fly vectors in some regions of the world are still neglected, as Bichromomyia olmeca (formerly known as Lutzomyia olmeca olmeca), a proven vector of Leishmania mexicana in Mexico and Central America. Despite the importance of this vector in transmitting Leishmania parasite in Mesoamerica there is no information on the repertoire of B. olmeca salivary proteins and their relationship to salivary proteins from other sand fly species. Methods and Findings A cDNA library of the salivary glands of wild-caught B. olmeca was constructed, sequenced, and analyzed. We identified transcripts encoding for novel salivary proteins from this sand fly species and performed a comparative analysis between B. olmeca salivary proteins and those from other sand fly species. With this new information we present an updated catalog of the salivary proteins specific to New World sand flies and salivary proteins common to all sand fly species. We also report in this work the anti-Factor Xa activity of Lofaxin, a salivary anticoagulant protein present in this sand fly species. Conclusions This study provides information on the first transcriptome of a sand fly from Mesoamerica and adds information to the limited repertoire of salivary transcriptomes from the Americas. This comparative analysis also shows a fast degree of evolution in salivary proteins from New World sand flies as compared with Old World sand flies. Leishmaniasis is a neglected disease caused by a parasite transmitted to the host by the bite of an infected sand fly. Sand fly saliva contains biologically active components that allow the sand fly to take a blood meal and also the parasite to spread in the host by countering the host immune mechanisms that fights the parasite. Research on sand fly saliva has allowed us to understand the biological functions of some of these proteins, to identify salivary proteins producing an immune response in different hosts and to select potential salivary vaccine that could be used to protect the host against the parasite. However, vectors transmitting different species of Leishmania in diverse regions of the world are still neglected. The present work focuses on the identification of the secreted proteins from the saliva of B. olmeca, a vector of Leishmania mexicana in North and Central America. We catalogued these proteins with those previously identified in other sand fly species from Old and New World. We showed here how conserved or divergent are these proteins families when comparing different sand fly species. We also report the anti-Factor Xa activity of Lofaxin, a salivary anticoagulant protein identified in the saliva of this sand fly species.
Collapse
|
23
|
Volfova V, Tothova V, Volf P. Hyaluronidase activity in the salivary glands of tabanid flies. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2016; 73:38-46. [PMID: 27045753 DOI: 10.1016/j.ibmb.2016.03.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 03/23/2016] [Accepted: 03/23/2016] [Indexed: 06/05/2023]
Abstract
Tabanids are haematophagous insects that act as biological and mechanical vectors of various diseases, including viruses, bacteria and parasites. The saliva of these insects contains strong anticoagulant and vasodilatory activities as well as immunoregulatory peptides. Here we demonstrate pronounced hyaluronidase (hyase) activity in ten tabanid species of the genera Chrysops, Haematopota, Hybomitra and Tabanus. Compared to other haematophagous insects, the ability of tabanid hyases to hydrolyze hyaluronic acid (HA) is extremely high, for example the enzyme activity of Hybomitra muehlfeldi was found to be 32-fold higher than the salivary hyase activity of the sand fly Phlebotomus papatasi. Hyases of all ten tabanid species tested also cleaved chondroitin sulfate A, another glycosaminoglycan present in the extracellular matrix of vertebrates. The pH optimum of the enzyme activity was measured in eight tabanid species; the hyase of Haemopota pluvialis was the only one with optimum at pH 4.0, while in the other seven species the activity optimum was at 5.0. SDS PAGE zymography showed the monomeric character of the enzymes in all tabanid species tested. Under non-reducing conditions the activities were visible as single bands with estimated MW between 35 and 52 kDa. The very high hyaluronidase activity in tabanid saliva might be related to their aggressive biting behavior as well as to their high efficiency as mechanical vectors. As they are supposedly involved in the enlargement of feeding hematomas, hyases might contribute to the mechanical transmission of pathogens. Pathogens present in vector mouthparts are co-inoculated into the vertebrate host together with saliva and may benefit from increased tissue permeability and the immunomodulatory activity of the salivary hyase.
Collapse
Affiliation(s)
- Vera Volfova
- Department of Parasitology, Faculty of Science, Charles University in Prague, Vinicna 7, Prague 2, 128 44, Czech republic
| | - Viktorie Tothova
- Department of Parasitology, Faculty of Science, Charles University in Prague, Vinicna 7, Prague 2, 128 44, Czech republic
| | - Petr Volf
- Department of Parasitology, Faculty of Science, Charles University in Prague, Vinicna 7, Prague 2, 128 44, Czech republic.
| |
Collapse
|
24
|
Rádrová J, Vlková M, Volfová V, Sumová P, Cêtre-Sossah C, Carpenter S, Darpel K, Rakotoarivony I, Allène X, Votýpka J, Volf P. Hyaluronidase Activity in Saliva of European Culicoides (Diptera: Ceratopogonidae). JOURNAL OF MEDICAL ENTOMOLOGY 2016; 53:212-216. [PMID: 26487248 PMCID: PMC4710844 DOI: 10.1093/jme/tjv147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 09/09/2015] [Indexed: 06/05/2023]
Abstract
Biting midges of the genus Culicoides transmit pathogens of veterinary importance such as bluetongue virus (Reoviridae: Orbivirus). The saliva of Culicoides is known to contain bioactive molecules including peptides and proteins with vasodilatory and immunomodulative properties. In this study, we detected activity of enzyme hyaluronidase in six Culicoides species that commonly occur in Europe and that are putative vectors of arboviruses. Hyaluronidase was present in all species studied, although its molecular size, sensitivity to SDS, and substrate specificity differed between species. Further studies on the potential effect of hyaluronidase activity on the vector competence of Culicoides species for arboviruses would be beneficial.
Collapse
Affiliation(s)
- Jana Rádrová
- Department of Parasitology, Charles University in Prague, Faculty of Science, Czech Republic (; ; ; ; ; ), Both authors contributed equally to this work,
| | - Michaela Vlková
- Department of Parasitology, Charles University in Prague, Faculty of Science, Czech Republic (; ; ; ; ; ), Both authors contributed equally to this work
| | - Věra Volfová
- Department of Parasitology, Charles University in Prague, Faculty of Science, Czech Republic (; ; ; ; ; )
| | - Petra Sumová
- Department of Parasitology, Charles University in Prague, Faculty of Science, Czech Republic (; ; ; ; ; )
| | - Catherine Cêtre-Sossah
- Cirad, UMR15 Contrôle des maladies, Montpellier, France (; ; ), INRA, UMR1309 Contrôle des maladies, Montpellier, France
| | - Simon Carpenter
- Vector-borne Viral Diseases Programme, The Pirbright Institute, Pirbright, Surrey, GU24-0NF, United Kingdom (; ), and
| | - Karin Darpel
- Vector-borne Viral Diseases Programme, The Pirbright Institute, Pirbright, Surrey, GU24-0NF, United Kingdom (; ), and School of Veterinary Medicine, University of Surrey, Guildford GU27AL, United Kingdom
| | - Ignace Rakotoarivony
- Cirad, UMR15 Contrôle des maladies, Montpellier, France (; ; ), INRA, UMR1309 Contrôle des maladies, Montpellier, France
| | - Xavier Allène
- Cirad, UMR15 Contrôle des maladies, Montpellier, France (; ; ), INRA, UMR1309 Contrôle des maladies, Montpellier, France
| | - Jan Votýpka
- Department of Parasitology, Charles University in Prague, Faculty of Science, Czech Republic (; ; ; ; ; )
| | - Petr Volf
- Department of Parasitology, Charles University in Prague, Faculty of Science, Czech Republic (; ; ; ; ; )
| |
Collapse
|
25
|
Guiguet A, Dubreuil G, Harris MO, Appel HM, Schultz JC, Pereira MH, Giron D. Shared weapons of blood- and plant-feeding insects: Surprising commonalities for manipulating hosts. JOURNAL OF INSECT PHYSIOLOGY 2016; 84:4-21. [PMID: 26705897 DOI: 10.1016/j.jinsphys.2015.12.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 12/14/2015] [Accepted: 12/15/2015] [Indexed: 05/04/2023]
Abstract
Insects that reprogram host plants during colonization remind us that the insect side of plant-insect story is just as interesting as the plant side. Insect effectors secreted by the salivary glands play an important role in plant reprogramming. Recent discoveries point to large numbers of salivary effectors being produced by a single herbivore species. Since genetic and functional characterization of effectors is an arduous task, narrowing the field of candidates is useful. We present ideas about types and functions of effectors from research on blood-feeding parasites and their mammalian hosts. Because of their importance for human health, blood-feeding parasites have more tools from genomics and other - omics than plant-feeding parasites. Four themes have emerged: (1) mechanical damage resulting from attack by blood-feeding parasites triggers "early danger signals" in mammalian hosts, which are mediated by eATP, calcium, and hydrogen peroxide, (2) mammalian hosts need to modulate their immune responses to the three "early danger signals" and use apyrases, calreticulins, and peroxiredoxins, respectively, to achieve this, (3) blood-feeding parasites, like their mammalian hosts, rely on some of the same "early danger signals" and modulate their immune responses using the same proteins, and (4) blood-feeding parasites deploy apyrases, calreticulins, and peroxiredoxins in their saliva to manipulate the "danger signals" of their mammalian hosts. We review emerging evidence that plant-feeding insects also interfere with "early danger signals" of their hosts by deploying apyrases, calreticulins and peroxiredoxins in saliva. Given emerging links between these molecules, and plant growth and defense, we propose that these effectors interfere with phytohormone signaling, and therefore have a special importance for gall-inducing and leaf-mining insects, which manipulate host-plants to create better food and shelter.
Collapse
Affiliation(s)
- Antoine Guiguet
- Institut de Recherche sur la Biologie de l'Insecte, UMR 7261 CNRS - Université François-Rabelais de Tours, 37200 Tours, France; Département de Biologie, École Normale Supérieure de Lyon, 69007 Lyon, France
| | - Géraldine Dubreuil
- Institut de Recherche sur la Biologie de l'Insecte, UMR 7261 CNRS - Université François-Rabelais de Tours, 37200 Tours, France
| | - Marion O Harris
- Department of Entomology, North Dakota State University, Fargo, ND 58105, USA; Le Studium Loire Valley Institute for Advanced Studies, 45000 Orléans, France
| | - Heidi M Appel
- Life Science Center, University of Missouri, Columbia, MO 65211, USA
| | - Jack C Schultz
- Life Science Center, University of Missouri, Columbia, MO 65211, USA
| | - Marcos H Pereira
- Le Studium Loire Valley Institute for Advanced Studies, 45000 Orléans, France; Laboratório de Fisiologia de Insectos Hematófagos, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - David Giron
- Institut de Recherche sur la Biologie de l'Insecte, UMR 7261 CNRS - Université François-Rabelais de Tours, 37200 Tours, France.
| |
Collapse
|
26
|
Petrella V, Aceto S, Musacchia F, Colonna V, Robinson M, Benes V, Cicotti G, Bongiorno G, Gradoni L, Volf P, Salvemini M. De novo assembly and sex-specific transcriptome profiling in the sand fly Phlebotomus perniciosus (Diptera, Phlebotominae), a major Old World vector of Leishmania infantum. BMC Genomics 2015; 16:847. [PMID: 26493315 PMCID: PMC4619268 DOI: 10.1186/s12864-015-2088-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 10/15/2015] [Indexed: 12/17/2022] Open
Abstract
Background The phlebotomine sand fly Phlebotomus perniciosus (Diptera: Psychodidae, Phlebotominae) is a major Old World vector of the protozoan Leishmania infantum, the etiological agent of visceral and cutaneous leishmaniases in humans and dogs, a worldwide re-emerging diseases of great public health concern, affecting 101 countries. Despite the growing interest in the study of this sand fly species in the last years, the development of genomic resources has been limited so far. To increase the available sequence data for P. perniciosus and to start studying the molecular basis of the sexual differentiation in sand flies, we performed whole transcriptome Illumina RNA sequencing (RNA-seq) of adult males and females and de novo transcriptome assembly. Results We assembled 55,393 high quality transcripts, of which 29,292 were unique, starting from adult whole body male and female pools. 11,736 transcripts had at least one functional annotation, including full-length low abundance salivary transcripts, 981 transcripts were classified as putative long non-coding RNAs and 244 transcripts encoded for putative novel proteins specific of the Phlebotominae sub-family. Differential expression analysis identified 8590 transcripts significantly biased between sexes. Among them, some show relaxation of selective constraints when compared to their orthologs of the New World sand fly species Lutzomyia longipalpis. Conclusions In this paper, we present a comprehensive transcriptome resource for the sand fly species P. perniciosus built from short-read RNA-seq and we provide insights into sex-specific gene expression at adult stage. Our analysis represents a first step towards the identification of sex-specific genes and pathways and a foundation for forthcoming investigations into this important vector species, including the study of the evolution of sex-biased genes and of the sexual differentiation in phlebotomine sand flies. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-2088-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- V Petrella
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - S Aceto
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - F Musacchia
- Stazione Zoologica "Anton Dohrn", Naples, Italy
| | - V Colonna
- National Research Council, Institute of Genetics and Biophysics, Naples, Italy
| | - M Robinson
- Institute of Molecular Life Science, University of Zurich, Zurich, Switzerland.,SIB-Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - V Benes
- Genomics Core Facility, EMBL, Heidelberg, Germany
| | - G Cicotti
- Institute for High Performance Computing and Networking, ICAR-CNR, Naples, Italy
| | - G Bongiorno
- Department of Infectious, Parasitic and Immunomediated Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - L Gradoni
- Department of Infectious, Parasitic and Immunomediated Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - P Volf
- Department of Parasitology, Charles University, Prague, Czech Republic
| | - M Salvemini
- Department of Biology, University of Naples Federico II, Naples, Italy.
| |
Collapse
|
27
|
Bordon KCF, Wiezel GA, Amorim FG, Arantes EC. Arthropod venom Hyaluronidases: biochemical properties and potential applications in medicine and biotechnology. J Venom Anim Toxins Incl Trop Dis 2015; 21:43. [PMID: 26500679 PMCID: PMC4619011 DOI: 10.1186/s40409-015-0042-7] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 10/08/2015] [Indexed: 01/24/2023] Open
Abstract
Hyaluronidases are enzymes that mainly degrade hyaluronan, the major glycosaminoglycan of the interstitial matrix. They are involved in several pathological and physiological activities including fertilization, wound healing, embryogenesis, angiogenesis, diffusion of toxins and drugs, metastasis, pneumonia, sepsis, bacteremia, meningitis, inflammation and allergy, among others. Hyaluronidases are widely distributed in nature and the enzymes from mammalian spermatozoa, lysosomes and animal venoms belong to the subclass EC 3.2.1.35. To date, only five three-dimensional structures for arthropod venom hyaluronidases (Apis mellifera and Vespula vulgaris) were determined. Additionally, there are four molecular models for hyaluronidases from Mesobuthus martensii, Polybia paulista and Tityus serrulatus venoms. These enzymes are employed as adjuvants to increase the absorption and dispersion of other drugs and have been used in various off-label clinical conditions to reduce tissue edema. Moreover, a PEGylated form of a recombinant human hyaluronidase is currently under clinical trials for the treatment of metastatic pancreatic cancer. This review focuses on the arthropod venom hyaluronidases and provides an overview of their biochemical properties, role in the envenoming, structure/activity relationship, and potential medical and biotechnological applications.
Collapse
Affiliation(s)
- Karla C F Bordon
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Avenida do Café, s/n, Ribeirão Preto, SP 14.040-903 Brazil
| | - Gisele A. Wiezel
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Avenida do Café, s/n, Ribeirão Preto, SP 14.040-903 Brazil
| | - Fernanda G. Amorim
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Avenida do Café, s/n, Ribeirão Preto, SP 14.040-903 Brazil
| | - Eliane C. Arantes
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Avenida do Café, s/n, Ribeirão Preto, SP 14.040-903 Brazil
| |
Collapse
|
28
|
Marzouki S, Kammoun-Rebai W, Bettaieb J, Abdeladhim M, Hadj Kacem S, Abdelkader R, Gritli S, Chemkhi J, Aslan H, Kamhawi S, Ben Salah A, Louzir H, Valenzuela JG, Ben Ahmed M. Validation of Recombinant Salivary Protein PpSP32 as a Suitable Marker of Human Exposure to Phlebotomus papatasi, the Vector of Leishmania major in Tunisia. PLoS Negl Trop Dis 2015; 9:e0003991. [PMID: 26368935 PMCID: PMC4569422 DOI: 10.1371/journal.pntd.0003991] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 07/15/2015] [Indexed: 01/08/2023] Open
Abstract
Background During a blood meal, female sand flies, vectors of Leishmania parasites, inject saliva into the host skin. Sand fly saliva is composed of a large variety of components that exert different pharmacological activities facilitating the acquisition of blood by the insect. Importantly, proteins present in saliva are able to elicit the production of specific anti-saliva antibodies, which can be used as markers for exposure to vector bites. Serological tests using total sand fly salivary gland extracts are challenging due to the difficulty of obtaining reproducible salivary gland preparations. Previously, we demonstrated that PpSP32 is the immunodominant salivary antigen in humans exposed to Phlebotomus papatasi bites and established that humans exposed to P. perniciosus bites do not recognize it. Methodology/Principal Findings Herein, we have validated, in a large cohort of 522 individuals, the use of the Phlebotomus papatasi recombinant salivary protein PpSP32 (rPpSP32) as an alternative method for testing exposure to the bite of this sand fly. We also demonstrated that screening for total anti-rPpSP32 IgG antibodies is sufficient, being comparable in efficacy to the screening for IgG2, IgG4 and IgE antibodies against rPpSP32. Additionally, sera obtained from dogs immunized with saliva of P. perniciosus, a sympatric and widely distributed sand fly in Tunisia, did not recognize rPpSP32 demonstrating its suitability as a marker of exposure to P. papatasi saliva. Conclusions/Significance Our data indicate that rPpSP32 constitutes a useful epidemiological tool to monitor the spatial distribution of P. papatasi in a particular region, to direct control measures against zoonotic cutaneous leishmaniasis, to assess the efficiency of vector control interventions and perhaps to assess the risk of contracting the disease. Leishmaniasis results from an infection by Leishmania parasites that are transmitted through the bites of infected sand flies. This disease affects millions of people worldwide. Zoonotic cutaneous leishmaniasis is widespread in Central Tunisia and constitutes an actual public health problem. Leishmania major, the etiological agent, is transmitted by the sand fly vector Phlebotomus papatasi. Saliva of sand flies contains several pharmacologically active components that play a key role in the acquisition of the blood meal and the establishment of the parasites, thus enhancing the infection. Some of these molecules are able to elicit the production of specific antibodies, which can be used as markers of exposure to the vector’s bite. Herein, using a large cohort of individuals, we have validated the use of P. papatasi recombinant salivary protein PpSP32 (rPpSP32) as an alternative method to standard entomological studies for testing exposure to the bite of this sand fly in humans. rPpSP32 represents a promising epidemiological tool to monitor the spatial distribution of P. papatasi, direct control measures against zoonotic cutaneous leishmaniasis, evaluate the efficiency of vector control interventions and potentially assess the risk of contracting the disease.
Collapse
Affiliation(s)
- Soumaya Marzouki
- Laboratory of Transmission, Control and Immunobiology of Infection, LR11IPT02, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Wafa Kammoun-Rebai
- Laboratory of Medical Parasitology, Biotechnologies and Biomolecules, LR11IPT06, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Jihene Bettaieb
- Laboratory of Transmission, Control and Immunobiology of Infection, LR11IPT02, Institut Pasteur de Tunis, Tunis, Tunisia
- Faculté de Médecine de Tunis, Université Tunis El Manar, Tunis, Tunisia
| | - Maha Abdeladhim
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Saoussen Hadj Kacem
- Laboratory of Transmission, Control and Immunobiology of Infection, LR11IPT02, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Rania Abdelkader
- Laboratory of Transmission, Control and Immunobiology of Infection, LR11IPT02, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Sami Gritli
- Department of Pathology, Charles Nicolle Hospital, Tunis, Tunisia
| | - Jomaa Chemkhi
- Laboratory of Molecular Epidemiology and Experimental Pathology, LR11IPT04, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Hamide Aslan
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Afif Ben Salah
- Laboratory of Transmission, Control and Immunobiology of Infection, LR11IPT02, Institut Pasteur de Tunis, Tunis, Tunisia
- Faculté de Médecine de Tunis, Université Tunis El Manar, Tunis, Tunisia
| | - Hechmi Louzir
- Laboratory of Transmission, Control and Immunobiology of Infection, LR11IPT02, Institut Pasteur de Tunis, Tunis, Tunisia
- Faculté de Médecine de Tunis, Université Tunis El Manar, Tunis, Tunisia
| | - Jesus G. Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Melika Ben Ahmed
- Laboratory of Transmission, Control and Immunobiology of Infection, LR11IPT02, Institut Pasteur de Tunis, Tunis, Tunisia
- Faculté de Médecine de Tunis, Université Tunis El Manar, Tunis, Tunisia
- * E-mail:
| |
Collapse
|
29
|
Cantacessi C, Dantas-Torres F, Nolan MJ, Otranto D. The past, present, and future of Leishmania genomics and transcriptomics. Trends Parasitol 2015; 31:100-8. [PMID: 25638444 PMCID: PMC4356521 DOI: 10.1016/j.pt.2014.12.012] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 12/22/2014] [Accepted: 12/22/2014] [Indexed: 01/31/2023]
Abstract
It has been nearly 10 years since the completion of the first entire genome sequence of a Leishmania parasite. Genomic and transcriptomic analyses have advanced our understanding of the biology of Leishmania, and shed new light on the complex interactions occurring within the parasite-host-vector triangle. Here, we review these advances and examine potential avenues for translation of these discoveries into treatment and control programs. In addition, we argue for a strong need to explore how disease in dogs relates to that in humans, and how an improved understanding in line with the 'One Health' concept may open new avenues for the control of these devastating diseases.
Collapse
Affiliation(s)
- Cinzia Cantacessi
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK.
| | - Filipe Dantas-Torres
- Departamento de Imunologia, Centro de Pesquisas Aggeu Magalhães, Fiocruz-PE, Brazil; Dipartimento di Medicina Veterinaria, Università degli Studi di Bari, Bari, Italy
| | - Matthew J Nolan
- Royal Veterinary College, University of London, North Mymms, UK
| | - Domenico Otranto
- Dipartimento di Medicina Veterinaria, Università degli Studi di Bari, Bari, Italy
| |
Collapse
|
30
|
Severity of old world cutaneous leishmaniasis is influenced by previous exposure to sandfly bites in Saudi Arabia. PLoS Negl Trop Dis 2015; 9:e0003449. [PMID: 25646796 PMCID: PMC4315490 DOI: 10.1371/journal.pntd.0003449] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 11/27/2014] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND The sandfly Phlebotomus papatasi is the vector of Leishmania major, the main causative agent of Old World cutaneous leishmaniasis (CL) in Saudi Arabia. Sandflies inject saliva while feeding and the salivary protein PpSP32 was previously shown to be a biomarker for bite exposure. Here we used recombinant PpSP32 to evaluate human exposure to Ph. papatasi bites, and study the association between antibody response to saliva and CL in endemic areas in Saudi Arabia. METHODOLOGY/PRINCIPAL FINDINGS In this observational study, anti-PpSP32 antibodies, as indicators of exposure to sandfly bites, were measured in sera from healthy individuals and patients from endemic regions in Saudi Arabia with active and cured CL. Ph. papatasi was identified as the primary CL vector in the study area. Anti-PpSP32 antibody levels were significantly higher in CL patients presenting active infections from all geographical regions compared to CL cured and healthy individuals. Furthermore, higher anti-PpSP32 antibody levels correlated with the prevalence and type of CL lesions (nodular vs. papular) observed in patients, especially non-local construction workers. CONCLUSIONS Our findings suggest a possible correlation between the type of immunity generated by the exposure to sandfly bites and disease outcome.
Collapse
|
31
|
Abdeladhim M, Kamhawi S, Valenzuela JG. What's behind a sand fly bite? The profound effect of sand fly saliva on host hemostasis, inflammation and immunity. INFECTION GENETICS AND EVOLUTION 2014; 28:691-703. [PMID: 25117872 DOI: 10.1016/j.meegid.2014.07.028] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Revised: 07/15/2014] [Accepted: 07/24/2014] [Indexed: 10/24/2022]
Abstract
Sand flies are blood-feeding insects and vectors of the Leishmania parasite. For many years, saliva of these insects has represented a gold mine for the discovery of molecules with anti-hemostatic and immuno-modulatory activities. Furthermore, proteins in sand fly saliva have been shown to be a potential vaccine against leishmaniasis and also markers of vector exposure. A bottleneck to progress in these areas of research has been the identification of molecules responsible for the observed activities and properties of saliva. Over the past decade, rapid advances in transcriptomics and proteomics resulted in the completion of a number of sialomes (salivary gland transcriptomes) and the expression of several recombinant salivary proteins from different species of sand fly vectors. This review will provide readers with a comprehensive update of recent advances in the characterization of these salivary molecules and their biological activities and offer insights pertaining to their protective effect against leishmaniasis and their potential as markers of vector exposure.
Collapse
Affiliation(s)
- Maha Abdeladhim
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, United States
| | - Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, United States.
| | - Jesus G Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, United States.
| |
Collapse
|
32
|
Kamhawi S, Aslan H, Valenzuela JG. Vector saliva in vaccines for visceral leishmaniasis: a brief encounter of high consequence? Front Public Health 2014; 2:99. [PMID: 25152872 PMCID: PMC4126209 DOI: 10.3389/fpubh.2014.00099] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 07/14/2014] [Indexed: 11/25/2022] Open
Abstract
Visceral leishmaniasis (VL) is a vector-borne disease transmitted by phlebotomine sand flies and remains the most serious form of the disease with no available human vaccine. Repeatedly, studies have demonstrated the immunogenicity and protective efficacy of a number of sand fly salivary proteins against cutaneous and visceral leishmaniasis. All Leishmania species including agents of VL are co-deposited into the skin together with vector saliva. Generally, the immune response to a protective salivary protein in vaccinated animals is rapid and possibly acts on the parasites soon after delivery into the skin by the bite of an infective sand fly. This is followed by the development of a stronger Leishmania-specific immunity in saliva-vaccinated animals compared to controls. Considering that several of the most efficacious protective molecules were identified from a proven vector of VL, we put forward the notion that a combination vaccine that includes a Leishmania antigen and a vector salivary protein has the potential to improve vaccine efficacy by targeting the parasite at it most vulnerable stage just after transmission.
Collapse
Affiliation(s)
- Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Hamide Aslan
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Jesus G. Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
33
|
Lehiy CJ, Drolet BS. The salivary secretome of the biting midge, Culicoides sonorensis. PeerJ 2014; 2:e426. [PMID: 24949243 PMCID: PMC4060021 DOI: 10.7717/peerj.426] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 05/21/2014] [Indexed: 11/20/2022] Open
Abstract
Culicoides biting midges (Diptera: Ceratopogonidae) are hematophagous insects with over 1400 species distributed throughout the world. Many of these species are of particular agricultural importance as primary vectors of bluetongue and Schmallenberg viruses, yet little is known about Culicoides genomics and proteomics. Detailed studies of members from other blood-feeding Dipteran families, including those of mosquito (Culicidae) and black fly (Simuliidae), have shown that protein components within the insect's saliva facilitate the blood feeding process. To determine the protein components in Culicoides sonorensis midges, secreted saliva was collected for peptide sequencing by tandem mass spectrometry. Forty-five secreted proteins were identified, including members of the D7 odorant binding protein family, Kunitz-like serine protease inhibitors, maltase, trypsin, and six novel proteins unique to C. sonorensis. Identifying the complex myriad of proteins in saliva from blood-feeding Dipteran species is critical for understanding their role in blood feeding, arbovirus transmission, and possibly the resulting disease pathogenesis.
Collapse
Affiliation(s)
- Christopher J Lehiy
- United States Department of Agriculture, Agricultural Research Service, Arthropod-Borne Animal Diseases Research Unit , Manhattan, KS , USA
| | - Barbara S Drolet
- United States Department of Agriculture, Agricultural Research Service, Arthropod-Borne Animal Diseases Research Unit , Manhattan, KS , USA
| |
Collapse
|
34
|
Martín-Martín I, Molina R, Rohoušová I, Drahota J, Volf P, Jiménez M. High levels of anti-Phlebotomus perniciosus saliva antibodies in different vertebrate hosts from the re-emerging leishmaniosis focus in Madrid, Spain. Vet Parasitol 2014; 202:207-16. [DOI: 10.1016/j.vetpar.2014.02.045] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 02/06/2014] [Accepted: 02/16/2014] [Indexed: 12/28/2022]
|
35
|
Vlkova M, Sima M, Rohousova I, Kostalova T, Sumova P, Volfova V, Jaske EL, Barbian KD, Gebre-Michael T, Hailu A, Warburg A, Ribeiro JMC, Valenzuela JG, Jochim RC, Volf P. Comparative analysis of salivary gland transcriptomes of Phlebotomus orientalis sand flies from endemic and non-endemic foci of visceral leishmaniasis. PLoS Negl Trop Dis 2014; 8:e2709. [PMID: 24587463 PMCID: PMC3937273 DOI: 10.1371/journal.pntd.0002709] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 01/07/2014] [Indexed: 12/20/2022] Open
Abstract
Background In East Africa, Phlebotomus orientalis serves as the main vector of Leishmania donovani, the causative agent of visceral leishmaniasis (VL). Phlebotomus orientalis is present at two distant localities in Ethiopia; Addis Zemen where VL is endemic and Melka Werer where transmission of VL does not occur. To find out whether the difference in epidemiology of VL is due to distant compositions of P. orientalis saliva we established colonies from Addis Zemen and Melka Werer, analyzed and compared the transcriptomes, proteomes and enzymatic activity of the salivary glands. Methodology/Principal Findings Two cDNA libraries were constructed from the female salivary glands of P. orientalis from Addis Zemen and Melka Werer. Clones of each P. orientalis library were randomly selected, sequenced and analyzed. In P. orientalis transcriptomes, we identified members of 13 main protein families. Phylogenetic analysis and multiple sequence alignments were performed to evaluate differences between the P. orientalis colonies and to show the relationship with other sand fly species from the subgenus Larroussius. To further compare both colonies, we investigated the humoral antigenicity and cross-reactivity of the salivary proteins and the activity of salivary apyrase and hyaluronidase. Conclusions This is the first report of the salivary components of P. orientalis, an important vector sand fly. Our study expanded the knowledge of salivary gland compounds of sand fly species in the subgenus Larroussius. Based on the phylogenetic analysis, we showed that P. orientalis is closely related to Phlebotomus tobbi and Phlebotomus perniciosus, whereas Phlebotomus ariasi is evolutionarily more distinct species. We also demonstrated that there is no significant difference between the transcriptomes, proteomes or enzymatic properties of the salivary components of Addis Zemen (endemic area) and Melka Werer (non-endemic area) P. orientalis colonies. Thus, the different epidemiology of VL in these Ethiopian foci cannot be attributed to the salivary gland composition. Phlebotomus orientalis is the vector of visceral leishmaniasis (VL) caused by Leishmania donovani in Northeast Africa. Immunization with sand fly saliva or with individual salivary proteins has been shown to protect against leishmaniasis in different hosts, warranting the intensive study of salivary proteins of sand fly vectors. In our study, we characterize the salivary compounds of P. orientalis, thereby broadening the repertoire of salivary proteins of sand fly species belonging to the subgenus Larroussius. In order to find out whether there is any connection between the composition of P. orientalis saliva and the epidemiology of VL in two distinct Ethiopian foci, Addis Zemen and Melka Werer, we studied the transcriptomes, proteomes, enzymatic activities, and the main salivary antigens in two P. orientalis colonies originating from these areas. We did not detect any significant difference between the saliva of female sand flies originating in Addis Zemen (endemic area) and Melka Werer (non-endemic area). Therefore, the different epidemiology of VL in these Ethiopian foci cannot be related to the distant salivary gland protein composition. Identifying the sand fly salivary gland compounds will be useful for future research focused on characterizing suitable salivary proteins as potential anti-Leishmania vaccine candidates.
Collapse
Affiliation(s)
- Michaela Vlkova
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Michal Sima
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Iva Rohousova
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Tatiana Kostalova
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Petra Sumova
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Vera Volfova
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Erin L. Jaske
- Genomics Unit, Research Technologies Section, Rocky Mountain Laboratories, Hamilton, Montana, United States of America
| | - Kent D. Barbian
- Genomics Unit, Research Technologies Section, Rocky Mountain Laboratories, Hamilton, Montana, United States of America
| | - Teshome Gebre-Michael
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia
| | - Asrat Hailu
- Department of Microbiology, Immunology & Parasitology, Faculty of Medicine, Addis Ababa University, Addis Ababa, Ethiopia
| | - Alon Warburg
- Department of Parasitology, The Kuvin Centre for the Study of Infectious and Tropical Diseases, Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jose M. C. Ribeiro
- Vector Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Jesus G. Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
- * E-mail: (JGV); (RCJ); (PV)
| | - Ryan C. Jochim
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
- * E-mail: (JGV); (RCJ); (PV)
| | - Petr Volf
- Department of Parasitology, Faculty of Science, Charles University, Prague, Czech Republic
- * E-mail: (JGV); (RCJ); (PV)
| |
Collapse
|
36
|
Drahota J, Martin-Martin I, Sumova P, Rohousova I, Jimenez M, Molina R, Volf P. Recombinant antigens from Phlebotomus perniciosus saliva as markers of canine exposure to visceral leishmaniases vector. PLoS Negl Trop Dis 2014; 8:e2597. [PMID: 24392167 PMCID: PMC3879210 DOI: 10.1371/journal.pntd.0002597] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 11/04/2013] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Phlebotomus perniciosus is the main vector in the western Mediterranean area of the protozoan parasite Leishmania infantum, the causative agent of canine and human visceral leishmaniases. Infected dogs serve as a reservoir of the disease, and therefore measuring the exposure of dogs to sand fly bites is important for estimating the risk of L. infantum transmission. In bitten hosts, sand fly saliva elicits a specific antibody response that reflects the intensity of sand fly exposure. As screening of specific anti-saliva antibodies is limited by the availability of salivary gland homogenates, utilization of recombinant salivary proteins is a promising alternative. In this manuscript we show for the first time the use of recombinant salivary proteins as a functional tool for detecting P. perniciosus bites in dogs. METHODOLOGY/PRINCIPAL FINDINGS The reactivity of six bacterially-expressed recombinant salivary proteins of P. perniciosus, yellow-related protein rSP03B, apyrases rSP01B and rSP01, antigen 5-related rSP07, ParSP25-like protein rSP08 and D7-related protein rSP04, were tested with sera of mice and dogs experimentally bitten by this sand fly using immunoblots and ELISA. In the immunoblots, both mice and canine sera gave positive reactions with yellow-related protein, both apyrases and ParSP25-like protein. A similar reaction for recombinant salivary proteins was observed by ELISA, with the reactivity of yellow-related protein and apyrases significantly correlated with the antibody response of mice and dogs against the whole salivary gland homogenate. CONCLUSIONS/SIGNIFICANCE Three recombinant salivary antigens of P. perniciosus, yellow-related protein rSP03B and the apyrases rSP01B and rSP01, were identified as the best candidates for evaluating the exposure of mice and dogs to P. perniciosus bites. Utilization of these proteins, or their combination, would be beneficial for screening canine sera in endemic areas of visceral leishmaniases for vector exposure and for estimating the risk of L. infantum transmission in dogs.
Collapse
Affiliation(s)
- Jan Drahota
- Department of Parasitology, Faculty of Science, Charles University in Prague, Prague, Czech Republic
- * E-mail:
| | - Ines Martin-Martin
- Unidad de Entomología Médica, Servicio de Parasitología, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Petra Sumova
- Department of Parasitology, Faculty of Science, Charles University in Prague, Prague, Czech Republic
| | - Iva Rohousova
- Department of Parasitology, Faculty of Science, Charles University in Prague, Prague, Czech Republic
| | - Maribel Jimenez
- Unidad de Entomología Médica, Servicio de Parasitología, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Ricardo Molina
- Unidad de Entomología Médica, Servicio de Parasitología, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Petr Volf
- Department of Parasitology, Faculty of Science, Charles University in Prague, Prague, Czech Republic
| |
Collapse
|
37
|
Oliveira F, de Carvalho AM, de Oliveira CI. Sand-fly saliva-leishmania-man: the trigger trio. Front Immunol 2013; 4:375. [PMID: 24312093 PMCID: PMC3832839 DOI: 10.3389/fimmu.2013.00375] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 10/31/2013] [Indexed: 11/23/2022] Open
Abstract
Leishmaniases are worldwide diseases transmitted to the vertebrate host by the bite of an infected sand-fly. Sand-fly biting and parasite inoculation are accompanied by the injection of salivary molecules, whose immunomodulatory properties are actively being studied. This mini review focuses on how the interactions between sand-fly saliva and the immune system may shape the outcome of infection, given its immunomodulatory properties, in experimental models and in the endemic area. Additionally, we approach the recent contributions regarding the identification of individual salivary components and how these are currently being considered as additional components of a vaccine against leishmaniasis.
Collapse
Affiliation(s)
- Fabiano Oliveira
- Vector Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Rockville, MD , USA
| | | | | |
Collapse
|
38
|
Martín-Martín I, Molina R, Jiménez M. Identifying salivary antigens of Phlebotomus argentipes by a 2DE approach. Acta Trop 2013; 126:229-39. [PMID: 23422341 DOI: 10.1016/j.actatropica.2013.02.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 01/09/2013] [Accepted: 02/05/2013] [Indexed: 12/21/2022]
Abstract
In the Indian subcontinent visceral leishmaniasis, also known as kala-azar, is caused by the protozoa Leishmania donovani and is transmitted to humans by the bite of infected female sand flies Phlebotomus argentipes in an anthroponotic cycle. Sand fly saliva is known to play an important role in host infection outcome after an infective bite. Immunogenicity of P. argentipes saliva has already been described. However, specific antigens that can contribute to these immunogenic properties are unknown. This work focuses on the identification of antigens present in P. argentipes saliva through the combination of two-dimensional electrophoresis (2DE) and Western blot (WB). Analysis of the salivary protein profile showed a gradual increase of the protein content in relation to the age of sand flies, reaching the complete salivary protein pattern at day five, which marked the minimum age for dissections. The 2DE revealed a reproducible protein profile that matched the classic monodimensional SDS-PAGE pattern (1DE). The resulting salivary proteomic map consisted of at least 30 spots located between 10 and 60 kDa. According to their isoelectric points, spots were mostly distributed around pH ranges: 5-6 and 9-10. In the proteomic maps, the presence of isoforms or posttranslational modifications was also highlighted since several spots were identified as the same protein. Analysis by in silico prediction programs located several potential glycosylation and phosphorylation sites in the aminoacidic sequences. On the other hand, pooled sera of immunized hamsters through the bite of uninfected sand flies showed elevated anti-saliva IgG levels. These sera permitted the detection of 4 protein bands and at least 20 protein spots in 1DE and 2DE respectively, followed by WB. The antigens were identified by MALDI-TOF, MALDI-TOF/TOF and de novo sequencing as D7-related proteins, PpSP15-like proteins, antigen 5-related proteins, apyrases, and several proteins without assigned protein family. Absence of cross-reactivity between P. argentipes and Phlebotomus perniciosus saliva antibodies determined by ELISA and WB was highlighted in this study, confirming that specific salivary antigens from different sand fly vectors need to be sought when designing vector-borne vaccines and markers for vector exposure assays.
Collapse
|
39
|
de Moura TR, Oliveira F, Carneiro MW, Miranda JC, Clarêncio J, Barral-Netto M, Brodskyn C, Barral A, Ribeiro JMC, Valenzuela JG, de Oliveira CI. Functional transcriptomics of wild-caught Lutzomyia intermedia salivary glands: identification of a protective salivary protein against Leishmania braziliensis infection. PLoS Negl Trop Dis 2013; 7:e2242. [PMID: 23717705 PMCID: PMC3662654 DOI: 10.1371/journal.pntd.0002242] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 04/16/2013] [Indexed: 01/02/2023] Open
Abstract
Background Leishmania parasites are transmitted in the presence of sand fly saliva. Together with the parasite, the sand fly injects salivary components that change the environment at the feeding site. Mice immunized with Phlebotomus papatasi salivary gland (SG) homogenate are protected against Leishmania major infection, while immunity to Lutzomyia intermedia SG homogenate exacerbated experimental Leishmania braziliensis infection. In humans, antibodies to Lu. intermedia saliva are associated with risk of acquiring L. braziliensis infection. Despite these important findings, there is no information regarding the repertoire of Lu. intermedia salivary proteins. Methods and Findings A cDNA library from the Salivary Glands (SGs) of wild-caught Lu. intermedia was constructed, sequenced, and complemented by a proteomic approach based on 1D SDS PAGE and mass/mass spectrometry to validate the transcripts present in this cDNA library. We identified the most abundant transcripts and proteins reported in other sand fly species as well as novel proteins such as neurotoxin-like proteins, peptides with ML domain, and three small peptides found so far only in this sand fly species. DNA plasmids coding for ten selected transcripts were constructed and used to immunize BALB/c mice to study their immunogenicity. Plasmid Linb-11—coding for a 4.5-kDa protein—induced a cellular immune response and conferred protection against L. braziliensis infection. This protection correlated with a decreased parasite load and an increased frequency of IFN-γ-producing cells. Conclusions We identified the most abundant and novel proteins present in the SGs of Lu. intermedia, a vector of cutaneous leishmaniasis in the Americas. We also show for the first time that immunity to a single salivary protein from Lu. intermedia can protect against cutaneous leishmaniasis caused by L. braziliensis. Sand fly saliva contains potent, biologically active proteins that allow the insect to stop host responses to acquire a blood meal. After repeated exposures, a number of these salivary proteins also induce a response in the host such as antibody production and/or cellular-mediated immunity. In animal models, these immune responses affect Leishmania infection. On one hand, immunity to Phlebotomus papatasi saliva protected animals against cutaneous leishmaniasis, while on the other hand, immunity to Lutzomyia intermedia saliva did not protect but exacerbated this disease. These differences are probably due to the types of proteins present in the saliva of these different sand fly species. The present work focused on isolation and identification of the secreted proteins present in the salivary glands of Lu. intermedia, an important vector of L. braziliensis, the agent of mucocutaneous leishmaniasis. Saliva from this sand fly contains a number of proteins not present in P. papatasi saliva and, with some exceptions; proteins that are homologous between the two species are very divergent. Furthermore, we identified one protein that, after vaccination, induced a cellular immune response able to protect mice against Leishmania braziliensis infection. This is the first evidence that a single salivary protein from Lu. intermedia can protect mice against this cutaneous leishmaniasis.
Collapse
Affiliation(s)
- Tatiana R. de Moura
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Bahia, Brazil
| | - Fabiano Oliveira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Marcia W. Carneiro
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Bahia, Brazil
| | - José Carlos Miranda
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Bahia, Brazil
| | - Jorge Clarêncio
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Bahia, Brazil
| | - Manoel Barral-Netto
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Bahia, Brazil
- Universidade Federal da Bahia, Salvador, Bahia, Brazil
- Instituto Nacional de Ciência e Tecnologia de Investigação em Imunologia (iii-INCT), Salvador, Bahia, Brazil
| | - Cláudia Brodskyn
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Bahia, Brazil
- Universidade Federal da Bahia, Salvador, Bahia, Brazil
- Instituto Nacional de Ciência e Tecnologia de Investigação em Imunologia (iii-INCT), Salvador, Bahia, Brazil
| | - Aldina Barral
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Bahia, Brazil
- Universidade Federal da Bahia, Salvador, Bahia, Brazil
- Instituto Nacional de Ciência e Tecnologia de Investigação em Imunologia (iii-INCT), Salvador, Bahia, Brazil
| | - José M. C. Ribeiro
- Vector Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Jesus G. Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
- * E-mail: (JGV); (CIdO)
| | - Camila I. de Oliveira
- Centro de Pesquisas Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Bahia, Brazil
- Instituto Nacional de Ciência e Tecnologia de Investigação em Imunologia (iii-INCT), Salvador, Bahia, Brazil
- * E-mail: (JGV); (CIdO)
| |
Collapse
|
40
|
Abstract
Leishmaniasis is a vector-borne neglected tropical disease associated with a spectrum of clinical manifestations, ranging from self-healing cutaneous lesions to fatal visceral infections. Among the most important questions in Leishmania research is why some species like L. donovani infect visceral organs, whereas other species like L. major remain in the skin. The determinants of visceral leishmaniasis are still poorly understood, although genomic, immunologic, and animal models are beginning to provide important insight into this disease. In this review, we discuss the vector, host, and pathogen factors that mediate the development of visceral leishmaniasis. We examine the progression of the parasite from the initial site of sand fly bite to the visceral organs and its ability to survive there. The identification of visceral disease determinants is required to understand disease evolution, to understand visceral organ survival mechanisms, and potentially to develop better interventions for this largely neglected disease.
Collapse
|
41
|
Abstract
Leishmaniases are vector-borne parasitic diseases with 0.9 - 1.4 million new human cases each year worldwide. In the vectorial part of the life-cycle, Leishmania development is confined to the digestive tract. During the first few days after blood feeding, natural barriers to Leishmania development include secreted proteolytic enzymes, the peritrophic matrix surrounding the ingested blood meal and sand fly immune reactions. As the blood digestion proceeds, parasites need to bind to the midgut epithelium to avoid being excreted with the blood remnant. This binding is strictly stage-dependent as it is a property of nectomonad and leptomonad forms only. While the attachment in specific vectors (P. papatasi, P. duboscqi and P. sergenti) involves lipophosphoglycan (LPG), this Leishmania molecule is not required for parasite attachment in other sand fly species experimentally permissive for various Leishmania. During late-stage infections, large numbers of parasites accumulate in the anterior midgut and produce filamentous proteophosphoglycan creating a gel-like plug physically obstructing the gut. The parasites attached to the stomodeal valve cause damage to the chitin lining and epithelial cells of the valve, interfering with its function and facilitating reflux of parasites from the midgut. Transformation to metacyclic stages highly infective for the vertebrate host is the other prerequisite for effective transmission. Here, we review the current state of knowledge of molecular interactions occurring in all these distinct phases of parasite colonization of the sand fly gut, highlighting recent discoveries in the field.
Collapse
Affiliation(s)
- Anna Dostálová
- Department of Parasitology, Faculty of Science, Charles University in Prague, Vinicna 7, 12844 Praha 2, Czech Republic
| | | |
Collapse
|
42
|
Marzouki S, Abdeladhim M, Abdessalem CB, Oliveira F, Ferjani B, Gilmore D, Louzir H, Valenzuela JG, Ahmed MB. Salivary antigen SP32 is the immunodominant target of the antibody response to Phlebotomus papatasi bites in humans. PLoS Negl Trop Dis 2012; 6:e1911. [PMID: 23209854 PMCID: PMC3510156 DOI: 10.1371/journal.pntd.0001911] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Accepted: 10/04/2012] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Zoonotic cutaneous leishmaniasis (ZCL) due to Leishmania major is highly prevalent in Tunisia and is transmitted by a hematophagous vector Phlebotomus papatasi (P. papatasi). While probing for a blood meal, the sand fly injects saliva into the host's skin, which contains a variety of compounds that are highly immunogenic. We recently showed that the presence of anti-saliva antibodies was associated with an enhanced risk for leishmaniasis and identified the immunodominant salivary protein of Phlebotomus papatasi as a protein of approximately 30 kDa. METHODOLOGY/PRINCIPAL FINDINGS We cloned and expressed in mammalian cells two salivary proteins PpSP30 and PpSP32 with predicted molecular weights close to 30 kDa from the Tunisian strain of P. papatasi. The two recombinant salivary proteins were purified by two-step HPLC (High-Performance Liquid Chromatography) and tested if these proteins correspond to the immunodominant antigen of 30 kDa previously shown to be recognized by human sera from endemic areas for ZCL and exposed naturally to P. papatasi bites. While recombinant PpSP30 (rPpSP30) was poorly recognized by human sera from endemic areas for ZCL, rPpSP32 was strongly recognized by the tested sera. The binding of human IgG antibodies to native PpSP32 was inhibited by the addition of rPpSP32. Consistently, experiments in mice showed that PpSP32 induced the highest levels of antibodies compared to other P. papatasi salivary molecules while PpSP30 did not induce any detectable levels of antibodies. CONCLUSIONS Our findings demonstrate that PpSP32 is the immunodominant target of the antibody response to P. papatasi saliva. They also indicate that the recombinant form of PpSP32 is similar to the native one and represents a good candidate for large scale testing of human exposure to P. papatasi bites and perhaps for assessing the risk of contracting the disease.
Collapse
Affiliation(s)
- Soumaya Marzouki
- Laboratory of Transmission, Control and Immunobiology of Infection, LR11IPT02, Institut Pasteur de Tunis, Tunis, Tunisie
| | - Maha Abdeladhim
- Laboratory of Transmission, Control and Immunobiology of Infection, LR11IPT02, Institut Pasteur de Tunis, Tunis, Tunisie
| | - Chaouki Ben Abdessalem
- Laboratory of Transmission, Control and Immunobiology of Infection, LR11IPT02, Institut Pasteur de Tunis, Tunis, Tunisie
| | - Fabiano Oliveira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Beya Ferjani
- Laboratory of Transmission, Control and Immunobiology of Infection, LR11IPT02, Institut Pasteur de Tunis, Tunis, Tunisie
| | - Dana Gilmore
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Hechmi Louzir
- Laboratory of Transmission, Control and Immunobiology of Infection, LR11IPT02, Institut Pasteur de Tunis, Tunis, Tunisie
- Faculté de Médecine de Tunis, Université Tunis El Manar, Tunis, Tunisie
| | - Jesus G. Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Mélika Ben Ahmed
- Laboratory of Transmission, Control and Immunobiology of Infection, LR11IPT02, Institut Pasteur de Tunis, Tunis, Tunisie
- Faculté de Médecine de Tunis, Université Tunis El Manar, Tunis, Tunisie
| |
Collapse
|
43
|
Kato H, Jochim RC, Gomez EA, Uezato H, Mimori T, Korenaga M, Sakurai T, Katakura K, Valenzuela JG, Hashiguchi Y. Analysis of salivary gland transcripts of the sand fly Lutzomyia ayacuchensis, a vector of Andean-type cutaneous leishmaniasis. INFECTION GENETICS AND EVOLUTION 2012; 13:56-66. [PMID: 23000112 DOI: 10.1016/j.meegid.2012.08.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 08/30/2012] [Accepted: 08/31/2012] [Indexed: 01/07/2023]
Abstract
The saliva of blood sucking insects contains potent pharmacologically active components that assist them in counteracting the host hemostatic and inflammatory systems during blood feeding. In addition, sand fly salivary proteins affect host immunity and have the potential to be a vaccine against Leishmania infection. In the present study, the salivary gland transcripts of Lutzomyia ayacuchensis, a vector of cutaneous leishmaniasis in Ecuadorian and Peruvian Andes, were analyzed by sequencing randomly selected clones of the salivary gland cDNA library of this sand fly. This resulted in the identification of the most abundant transcripts coding for secreted proteins. These proteins were homologous to the salivary molecules present in other sand flies including the RGD-containing peptide, PpSP15/SL1 family protein, yellow-related protein, putative apyrase, antigen 5-related protein, D7 family protein, and 27 kDa salivary protein. Of note, homologues of maxadilan, an active vasodilator abundantly present in saliva of Lutzomyia longipalpis, were not identified. This analysis is the first description of salivary proteins from a sand fly of the subgenus Helcocyrtomyia and from vector of cutaneous leishmaniasis in the New World. The present analysis will provide further insights into the evolution of salivary components in blood sucking arthropods.
Collapse
Affiliation(s)
- Hirotomo Kato
- Laboratory of Parasitology, Department of Disease Control, Graduate School of Veterinary Medicine, Hokkaido University, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|