1
|
Kato H. Dr. Jekyll and Mr. Hyde in sand fly saliva. Parasitol Int 2025; 105:102998. [PMID: 39581305 DOI: 10.1016/j.parint.2024.102998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/28/2024] [Accepted: 11/20/2024] [Indexed: 11/26/2024]
Abstract
Phlebotomine sand flies are very small hematophagous insects, and some species transmit human pathogens, such as Leishmania protozoa. Similar to other hematophagous insects, sand flies possess unique bioactive substances in their saliva to facilitate blood feeding. Active transcriptome and proteome analyses revealed that sand flies have unique molecules in their saliva that are structurally different from those of other arthropods. These components exert anticoagulant, antiplatelet, vasodilator, and anti-inflammatory effects on the host, and the unique bioactivities of each molecule are currently being characterized. Several bioactivities of salivary components have been associated with the exacerbation of Leishmania infection, and investigations on the molecular mechanisms responsible are underway. On the other hand, host immunity to some salivary components has been shown to confer protection against Leishmania infection, suggesting the potential of salivary components as vaccine candidates. Although some negative effects of protection by sand fly saliva have been reported, the identification of suitable immunogens and elucidation of appropriate protective immunity are expected for the development of a sand fly saliva vaccine against Leishmania infection.
Collapse
Affiliation(s)
- Hirotomo Kato
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke city, Tochigi 329-0498, Japan.
| |
Collapse
|
2
|
Chaves MM. Neutrophils and purinergic signaling: Partners in the crime against Leishmania parasites? Biochimie 2025; 232:43-53. [PMID: 39855456 DOI: 10.1016/j.biochi.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 12/18/2024] [Accepted: 01/17/2025] [Indexed: 01/27/2025]
Abstract
The parasite of the genus Leishmania is the causative agent of diseases that affect humans called leishmaniasis. These diseases affect millions of people worldwide and the currently existing drugs are either very toxic or the parasites acquire resistance. Therefore, new elimination mechanisms need to be elucidated so that new therapeutic strategies can be developed. Much has already been discussed about the role of neutrophils in Leishmania infection, and their participation is still controversial. A recent study showed that receptors present in the neutrophil membrane, the purinergic receptors, can control the infection when activated, but the triggering mechanism has not been elucidated. In this review, we will address the possible participation of purinergic receptors expressed in the neutrophil extracellular membrane that may be participating in the detection of Leishmania infection and their possible effects during parasitism.
Collapse
Affiliation(s)
- Mariana M Chaves
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil; Bio-Manguinhos, Oswaldo Cruz Foundation, Brazilian Ministry of Health, Rio de Janeiro, Brazil.
| |
Collapse
|
3
|
Pala ZR, Alves E Silva TL, Minai M, Crews B, Patino-Martinez E, Carmona-Rivera C, Valenzuela Leon PC, Martin-Martin I, Flores-Garcia Y, Cachau RE, Muslinkina L, Gittis AG, Srivastava N, Garboczi DN, Alves DA, Kaplan MJ, Fischer E, Calvo E, Vega-Rodriguez J. Mosquito salivary apyrase regulates blood meal hemostasis and facilitates malaria parasite transmission. Nat Commun 2024; 15:8194. [PMID: 39294191 PMCID: PMC11410810 DOI: 10.1038/s41467-024-52502-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 09/10/2024] [Indexed: 09/20/2024] Open
Abstract
The evolution of hematophagy involves a series of adaptations that allow blood-feeding insects to access and consume blood efficiently while managing and circumventing the host's hemostatic and immune responses. Mosquito, and other insects, utilize salivary proteins to regulate these responses at the bite site during and after blood feeding. We investigated the function of Anopheles gambiae salivary apyrase (AgApyrase) in regulating hemostasis in the mosquito blood meal and in Plasmodium transmission. Our results demonstrate that salivary apyrase, a known inhibitor of platelet aggregation, interacts with and activates tissue plasminogen activator, facilitating the conversion of plasminogen to plasmin, a human protease that degrades fibrin and facilitates Plasmodium transmission. We show that mosquitoes ingest a substantial amount of apyrase during blood feeding, which reduces coagulation in the blood meal by enhancing fibrin degradation and inhibiting platelet aggregation. AgApyrase significantly enhanced Plasmodium infection in the mosquito midgut, whereas AgApyrase immunization inhibited Plasmodium mosquito infection and sporozoite transmission. This study highlights a pivotal role for mosquito salivary apyrase for regulation of hemostasis in the mosquito blood meal and for Plasmodium transmission to mosquitoes and to the mammalian host, underscoring the potential for strategies to prevent malaria transmission.
Collapse
Affiliation(s)
- Zarna Rajeshkumar Pala
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Thiago Luiz Alves E Silva
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Mahnaz Minai
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Benjamin Crews
- Microscopy Unit, Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, 59840, USA
| | - Eduardo Patino-Martinez
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Carmelo Carmona-Rivera
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Paola Carolina Valenzuela Leon
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Ines Martin-Martin
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
- Laboratory of Medical Entomology, National Center for Microbiology, Instituto de Salud Carlos III, 28220, Majadahonda, Madrid, Spain
| | - Yevel Flores-Garcia
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Raul E Cachau
- Integrated Data Science Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Liya Muslinkina
- Structural Biology Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Apostolos G Gittis
- Structural Biology Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Naman Srivastava
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - David N Garboczi
- Structural Biology Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Derron A Alves
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Elizabeth Fischer
- Microscopy Unit, Research Technologies Branch, 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
| | - Joel Vega-Rodriguez
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA.
| |
Collapse
|
4
|
Fayaz S, Raz A, Bahrami F, Fard-Esfahani P, Parvizi P, Ajdary S. Molecular identification of Phlebotomus kandelakii apyrase and assessment of the immunogenicity of its recombinant protein in BALB/c mice. Sci Rep 2023; 13:8766. [PMID: 37253833 DOI: 10.1038/s41598-023-36037-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 05/28/2023] [Indexed: 06/01/2023] Open
Abstract
Sand fly salivary proteins have immunomodulatory and anti-inflammatory features; hence, they are proven to perform important roles in the early establishment of Leishmania parasite in the vertebrate host. Among them, salivary apyrase with anti-hemostatic properties has a crucial role during the blood meal process. In the present study, a Genome-Walking method was used to characterize a full-length nucleotide sequence of Phlebotomus (P.) kandelakii apyrase (Pkapy). Bioinformatics analyses revealed that Pkapy is a ~ 36 kDa stable and hydrophilic protein that belongs to the Cimex family of apyrases. Moreover, recombinant proteins of Pkapy and P. papatasi apyrase (Ppapy) were over-expressed in Escherichia coli BL2 (DE3) and their antigenicity in BALB/c mice was evaluated. Dot-blot and ELISA results indicated that both recombinant apyrases could induce antibodies in BALB/c. Moreover, a partial cross-reactivity between Pkapy and Ppapy was found. In vitro stimulation of splenocytes from immunized mice with the recombinant proteins indicated cross-reactive T cell proliferative responses. Cytokine analysis revealed significant production of IFN-γ (p < 0.001) and IL-10 (p < 0.01) in response to Pkapy. In conclusion, the full-length nucleotide sequence and molecular characteristics of Pkapy were identified for the first time. Immunologic analyses indicated that Pkapy and Ppapy are immunogenic in BALB/c mice and show partial cross-reactive responses. The immunity to Pkapy was found to be a Th1-dominant response that highlights its potential as a component for an anti-Leishmania vaccine.
Collapse
Affiliation(s)
- Shima Fayaz
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
- Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran
| | - Abbasali Raz
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Fariborz Bahrami
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | | | - Parviz Parvizi
- Department of Parasitology, Pasteur Institute of Iran, Tehran, Iran
| | - Soheila Ajdary
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
5
|
Pala ZR, Alves e Silva TL, Minai M, Crews B, Patino-Martinez E, Carmona-Rivera C, Valenzuela-Leon PC, Martin-Martin I, Flores-Garcia Y, Cachau RE, Srivastava N, Moore IN, Alves DA, Kaplan MJ, Fischer E, Calvo E, Vega-Rodriguez J. Anopheles salivary apyrase regulates blood meal hemostasis and drives malaria parasite transmission. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.22.541827. [PMID: 37292610 PMCID: PMC10245845 DOI: 10.1101/2023.05.22.541827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Mosquito salivary proteins play a crucial role in regulating hemostatic responses at the bite site during blood feeding. In this study, we investigate the function of Anopheles gambiae salivary apyrase (AgApyrase) in Plasmodium transmission. Our results demonstrate that salivary apyrase interacts with and activates tissue plasminogen activator, facilitating the conversion of plasminogen to plasmin, a human protein previously shown to be required for Plasmodium transmission. Microscopy imaging shows that mosquitoes ingest a substantial amount of apyrase during blood feeding which reduces coagulation in the blood meal by enhancing fibrin degradation and inhibiting platelet aggregation. Supplementation of Plasmodium infected blood with apyrase significantly enhanced Plasmodium infection in the mosquito midgut. In contrast, AgApyrase immunization inhibited Plasmodium mosquito infection and sporozoite transmission. This study highlights a pivotal role for mosquito salivary apyrase for regulation of hemostasis in the mosquito blood meal and for Plasmodium transmission to mosquitoes and to the mammal host, underscoring the potential for new strategies to prevent malaria transmission.
Collapse
Affiliation(s)
- Zarna Rajeshkumar Pala
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Thiago Luiz Alves e Silva
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Mahnaz Minai
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Benjamin Crews
- Microscopy Unit, Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Eduardo Patino-Martinez
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Carmelo Carmona-Rivera
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Paola Carolina Valenzuela-Leon
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Ines Martin-Martin
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
- Current address: Laboratory of Medical Entomology, National Center for Microbiology, Instituto de Salud Carlos III, 28220 Majadahonda, Madrid, Spain
| | - Yevel Flores-Garcia
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Raul E. Cachau
- Integrated Data Science Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Naman Srivastava
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Ian N. Moore
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Derron A. Alves
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Elizabeth Fischer
- Microscopy Unit, Research Technologies Branch, 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
| | - Joel Vega-Rodriguez
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| |
Collapse
|
6
|
Barillas-Mury C, Ribeiro JMC, Valenzuela JG. Understanding pathogen survival and transmission by arthropod vectors to prevent human disease. Science 2022; 377:eabc2757. [PMID: 36173836 DOI: 10.1126/science.abc2757] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Many endemic poverty-associated diseases, such as malaria and leishmaniasis, are transmitted by arthropod vectors. Pathogens must interact with specific molecules in the vector gut, the microbiota, and the vector immune system to survive and be transmitted. The vertebrate host, in turn, is infected when the pathogen and vector-derived factors, such as salivary proteins, are delivered into the skin by a vector bite. Here, we review recent progress in our understanding of the biology of pathogen transmission from the human to the vector and back, from the vector to the host. We also highlight recent advances in the biology of vector-borne disease transmission, which have translated into additional strategies to prevent human disease by either reducing vector populations or by disrupting their ability to transmit pathogens.
Collapse
Affiliation(s)
- Carolina Barillas-Mury
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, MD 20852, USA
| | - José M C Ribeiro
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, MD 20852, USA
| | - Jesus G Valenzuela
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, MD 20852, USA
| |
Collapse
|
7
|
Aoki V, Abdeladhim M, Li N, Cecilio P, Prisayanh P, Diaz LA, Valenzuela JG. Some Good and Some Bad: Sand Fly Salivary Proteins in the Control of Leishmaniasis and in Autoimmunity. Front Cell Infect Microbiol 2022; 12:839932. [PMID: 35281450 PMCID: PMC8913536 DOI: 10.3389/fcimb.2022.839932] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/02/2022] [Indexed: 01/22/2023] Open
Abstract
Sand flies are hematophagous insects responsible for the transmission of vector-borne diseases to humans. Prominent among these diseases is Leishmaniasis that affects the skin and mucous surfaces and organs such as liver and spleen. Importantly, the function of blood-sucking arthropods goes beyond merely transporting pathogens. The saliva of vectors of disease contains pharmacologically active components that facilitate blood feeding and often pathogen establishment. Transcriptomic and proteomic studies have enumerated the repertoire of sand fly salivary proteins and their potential use for the control of Leishmaniasis, either as biomarkers of vector exposure or as anti-Leishmania vaccines. However, a group of specific sand fly salivary proteins triggers formation of cross-reactive antibodies that bind the ectodomain of human desmoglein 1, a member of the epidermal desmosomal cadherins. These cross-reactive antibodies are associated with skin autoimmune blistering diseases, such as pemphigus, in certain immunogenetically predisposed individuals. In this review, we focus on two different aspects of sand fly salivary proteins in the context of human disease: The good, which refers to salivary proteins functioning as biomarkers of exposure or as anti-Leishmania vaccines, and the bad, which refers to salivary proteins as environmental triggers of autoimmune skin diseases.
Collapse
Affiliation(s)
- Valeria Aoki
- Department of Dermatology, Faculdade de Medicina da Universidade de São Paulo (FMUSP), Universidade de Sao Paulo, Sao Paulo, Brazil
- *Correspondence: Valeria Aoki,
| | - 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, United States
| | - Ning Li
- Department of Dermatology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Pedro Cecilio
- Vector Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Phillip Prisayanh
- Department of Dermatology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Luis A. Diaz
- Department of Dermatology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 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
|
8
|
Aronson NE, Oliveira F, Gomes R, Porter WD, Howard RS, Kamhawi S, Valenzuela JG. Antibody Responses to Phlebotomus papatasi Saliva in American Soldiers With Cutaneous Leishmaniasis Versus Controls. FRONTIERS IN TROPICAL DISEASES 2022. [DOI: 10.3389/fitd.2021.766273] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Leishmania major, transmitted in Iraq by the bite of a sand fly Phlebotomus papatasi, causes cutaneous leishmaniasis (CL). The sand fly saliva is immunogenic, with both systemic humoral and cellular human immune responses resulting from natural exposure. 248 Americans who developed L. major infection in Iraq were sex, race/ethnicity, year of Iraq deployment-matched to controls without CL. Using a case-control study design, we compared sand fly saliva-specific human IgG levels and recognized antigens between the two groups. Serologic responses to Ph. papatasi salivary gland homogenate were studied with ELISA and Western blot, using serial samples obtained from before travel, during CL treatment (CL) or at time of return to US (controls), as well as (for CL cases) six to 24 months after return to non-endemic US. The mean change in optical density (MCOD), reflecting the change in sand fly saliva-specific IgG before and after exposure in Iraq, was 0.296 (range -0.138 to 2.057) in cases and 0.151 (range -0.454 to1.085) in controls, p<0.001. Low levels of sand fly saliva specific antibody were noted in CL cases by 7-8 months after return to the US. The most frequently recognized Ph. papatasi salivary antigens were MW30 (PpSP32) and MW64, although other salivary proteins recognized were MW12/14, 15, 18, 28, 32, 36, 42, 44, 46, 52. Logistic regression suggested that MW15, 28 and 42 were associated with the largest effect on the MCOD. MW30 was the most frequently recognized antigen suggesting a role as biomarker for sand fly exposure and CL risk. Anti-Ph. papatasi saliva IgG waned within months of return to the US. We also discuss vector antigenic saliva proteins in the context of CL presentation and identify some salivary antigens that may correlate with less lesion area, ulcer versus papule/plaque, race among those with CL.
Collapse
|
9
|
Tanaka K, Heil M. Damage-Associated Molecular Patterns (DAMPs) in Plant Innate Immunity: Applying the Danger Model and Evolutionary Perspectives. ANNUAL REVIEW OF PHYTOPATHOLOGY 2021; 59:53-75. [PMID: 33900789 DOI: 10.1146/annurev-phyto-082718-100146] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Danger signals trigger immune responses upon perception by a complex surveillance system. Such signals can originate from the infectious nonself or the damaged self, the latter termed damage-associated molecular patterns (DAMPs). Here, we apply Matzinger's danger model to plant innate immunity to discuss the adaptive advantages of DAMPs and their integration into preexisting signaling pathways. Constitutive DAMPs (cDAMPs), e.g., extracellular ATP, histones, and self-DNA, fulfill primary, conserved functions and adopt a signaling role only when cellular damage causes their fragmentation or localization to aberrant compartments. By contrast, immunomodulatory peptides (also known as phytocytokines) exclusively function as signals and, upon damage, are activated as inducible DAMPs (iDAMPs). Dynamic coevolutionary processes between the signals and their emerging receptors and shared co-receptors have likely linked danger recognition to preexisting, conserved downstream pathways.
Collapse
Affiliation(s)
- Kiwamu Tanaka
- Department of Plant Pathology, Washington State University, Pullman, Washington 99163, USA;
| | - Martin Heil
- Departamento de Ingeniería Genética, CINVESTAV, 36821 Irapuato, Guanajuato, México
| |
Collapse
|
10
|
Demarta-Gatsi C, Mécheri S. Vector saliva controlled inflammatory response of the host may represent the Achilles heel during pathogen transmission. J Venom Anim Toxins Incl Trop Dis 2021; 27:e20200155. [PMID: 34035796 PMCID: PMC8128132 DOI: 10.1590/1678-9199-jvatitd-2020-0155] [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] [Indexed: 12/11/2022] Open
Abstract
Infection with vector-borne pathogens starts with the inoculation of these pathogens during blood feeding. In endemic regions, the population is regularly bitten by naive vectors, implicating a permanent stimulation of the immune system by the vector saliva itself (pre-immune context). Comparatively, the number of bites received by exposed individuals from non-infected vectors is much higher than the bites from infected ones. Therefore, vector saliva and the immunological response in the skin may play an important role, so far underestimated, in the establishment of anti-pathogen immunity in endemic areas. Hence, the parasite biology and the disease pathogenesis in “saliva-primed” and “saliva-unprimed” individuals must be different. This integrated view on how the pathogen evolves within the host together with vector salivary components, which are known to be endowed with a variety of pharmacological and immunological properties, must remain the focus of any investigational study dealing with vector-borne diseases. Considering this three-way partnership, the host skin (immune system), the pathogen, and the vector saliva, the approach that consists in the validation of vector saliva as a source of molecular entities with anti-disease vaccine potential has been recently a subject of active and fruitful investigation. As an example, the vaccination with maxadilan, a potent vasodilator peptide extracted from the saliva of the sand fly Lutzomyia longipalpis, was able to protect against infection with various leishmanial parasites. More interestingly, a universal mosquito saliva vaccine that may potentially protect against a range of mosquito-borne infections including malaria, dengue, Zika, chikungunya and yellow fever. In this review, we highlight the key role played by the immunobiology of vector saliva in shaping the outcome of vector-borne diseases and discuss the value of studying diseases in the light of intimate cross talk among the pathogen, the vector saliva, and the host immune mechanisms.
Collapse
Affiliation(s)
- Claudia Demarta-Gatsi
- Institut Pasteur, Unité de Biologie des Interactions Hôte Parasites, Paris, France.,CNRS ERL9195, Paris, France.,INSERM U1201, Paris, France.,Medicines for Malaria Venture (MMV), Geneva, Switzerland.,Institut Pasteur, Unité de Biologie des Interactions Hôte Parasites, Paris, France
| | - Salah Mécheri
- Institut Pasteur, Unité de Biologie des Interactions Hôte Parasites, Paris, France.,CNRS ERL9195, Paris, France.,INSERM U1201, Paris, France
| |
Collapse
|
11
|
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: 0.8] [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
|
12
|
Jablonka W, Kim IH, Alvarenga PH, Valenzuela JG, Ribeiro JMC, Andersen JF. Functional and structural similarities of D7 proteins in the independently-evolved salivary secretions of sand flies and mosquitoes. Sci Rep 2019; 9:5340. [PMID: 30926880 PMCID: PMC6440969 DOI: 10.1038/s41598-019-41848-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 03/18/2019] [Indexed: 11/09/2022] Open
Abstract
The habit of blood feeding evolved independently in many insect orders of families. Sand flies and mosquitoes belong to separate lineages of blood-feeding Diptera and are thus considered to have evolved the trait independently. Because of this, sand fly salivary proteins differ structurally from those of mosquitoes, and orthologous groups are nearly impossible to define. An exception is the long-form D7-like proteins that show conservation with their mosquito counterparts of numerous residues associated with the N-terminal domain binding pocket. In mosquitoes, this pocket is responsible for the scavenging of proinflammatory cysteinyl leukotrienes and thromboxanes at the feeding site. Here we show that long-form D7 proteins AGE83092 and ABI15936 from the sand fly species, Phlebotomus papatasi and P. duboscqi, respectively, inhibit the activation of platelets by collagen and the thromboxane A2 analog U46619. Using isothermal titration calorimetry, we also demonstrate direct binding of U46619 and cysteinyl leukotrienes C4, D4 and E4 to the P. papatasi protein. The crystal structure of P. duboscqi ABI15936 was determined and found to contain two domains oriented similarly to those of the mosquito proteins. The N-terminal domain contains an apparent eicosanoid binding pocket. The C-terminal domain is smaller in overall size than in the mosquito D7s and is missing some helical elements. Consequently, it does not contain an obvious internal binding pocket for small-molecule ligands that bind to many mosquito D7s. Structural similarities indicate that mosquito and sand fly D7 proteins have evolved from similar progenitors, but phylogenetics and differences in intron/exon structure suggest that they may have acquired the ability to bind vertebrate eicosanoids independently, indicating a convergent evolution scenario.
Collapse
Affiliation(s)
- Willy Jablonka
- The Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, Maryland, 20852, USA
| | - Il Hwan Kim
- The Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, Maryland, 20852, USA
| | - Patricia H Alvarenga
- The Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, Maryland, 20852, USA
| | - Jesus G Valenzuela
- The Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, Maryland, 20852, USA
| | - Jose M C Ribeiro
- The Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, Maryland, 20852, USA
| | - John F Andersen
- The Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, Maryland, 20852, USA.
| |
Collapse
|
13
|
Pandey RK, Prajapati VK. Exploring sand fly salivary proteins to design multiepitope subunit vaccine to fight against visceral leishmaniasis. J Cell Biochem 2019; 120:1141-1155. [PMID: 29377223 DOI: 10.1002/jcb.26719] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/24/2018] [Indexed: 01/24/2023]
Abstract
Visceral leishmaniasis (VL) is caused by the parasites of Leishmania donovani complex, leads to the death of 20 000 to 40 000 people from 56 affected countries, worldwide. Till date, there is not a single available vaccine candidate to prevent the VL infection, and treatment only relies upon expensive and toxic chemotherapeutic options. Consequently, immunoinformatics approach was applied to design a multiepitope-based subunit vaccine to enhance the humoral as well as cell-mediated immunity. Constructed vaccine candidate was further subjected to evaluation on allergenicity and antigenicity and physiochemical parameters. Later on, disulfide engineering was performed to increase the stability of vaccine construct. Also, molecular docking and molecular dynamics simulation study were performed to check the binding affinity and stability of toll-like receptor-4 to vaccine construct complex. Finally, codon optimization and in silico cloning were performed to ensure the expression of proposed vaccine construct in a microbial expression system.
Collapse
Affiliation(s)
- Rajan Kumar Pandey
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, India
| |
Collapse
|
14
|
Merchant A, Yu T, Shi J, Zhou X. Development of a Diagnostic Marker for Phlebotomus papatasi to Initiate a Potential Vector Surveillance Program in North America. INSECTS 2018; 9:insects9040162. [PMID: 30424532 PMCID: PMC6315872 DOI: 10.3390/insects9040162] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/07/2018] [Accepted: 11/09/2018] [Indexed: 11/16/2022]
Abstract
Phlebotomus papatasi, an Old World sand fly species, is primarily responsible for the transmission of leishmaniasis, a highly infectious and potentially lethal disease. International travel, especially military rotations, between domestic locations and P. papatasi-prevalent regions in the Middle East poses an imminent threat to the public health of US citizens. Because of its small size and cryptic morphology, identification of P. papatasi is challenging and labor-intensive. Here, we developed a ribosomal DNA-polymerase chain reaction (PCR)-based diagnostic assay that is capable of detecting P. papatasi genomic DNA from mixed samples containing multiple sand flies native to the Americas. Serial dilution of P. papatasi samples demonstrated that this diagnostic assay could detect one P. papatasi from up to 255 non-target sand flies. Due to its simplicity, sensitivity and specificity, this rapid identification tool is suited for a long-term surveillance program to screen for the presence of P. papatasi in the continental United States and to reveal geographical regions potentially vulnerable to sand fly-borne diseases.
Collapse
Affiliation(s)
- Austin Merchant
- Department of Entomology, University of Kentucky, Lexington, KY 40546, USA.
| | - Tian Yu
- Department of Entomology, University of Kentucky, Lexington, KY 40546, USA.
| | - Jizhe Shi
- Department of Entomology, University of Kentucky, Lexington, KY 40546, USA.
| | - Xuguo Zhou
- Department of Entomology, University of Kentucky, Lexington, KY 40546, USA.
| |
Collapse
|
15
|
Tripathi D, Tanaka K. A crosstalk between extracellular ATP and jasmonate signaling pathways for plant defense. PLANT SIGNALING & BEHAVIOR 2018; 13:e1432229. [PMID: 29370573 PMCID: PMC6103277 DOI: 10.1080/15592324.2018.1432229] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 01/22/2018] [Indexed: 05/24/2023]
Abstract
Damage-associated molecular patterns (DAMPs), such as extracellular ATP, act as danger signals in response to biotic and abiotic stresses. Extracellular ATP is perceived by a plant purinoceptor, P2 receptor kinase 1 (P2K1), inducing downstream signaling for defense responses. How ATP induces these defense responses has not been well studied. A recent study by Tripathi et al. (Plant Physiology, 176: 511-523, 2018) revealed a synergistic interaction between extracellular ATP and jasmonate (JA) signaling during plant defense responses. This signaling crosstalk requires the formation of secondary messengers, i.e., cytosolic calcium, reactive oxygen species, and nitric oxide. This finding has given a new direction towards understanding the defense signals activated by DAMPs. In this addendum, we discuss possible insights into how extracellular ATP signaling interacts with the JA signaling pathway for plant defense responses.
Collapse
Affiliation(s)
- Diwaker Tripathi
- Department of Plant Pathology, Washington State University, Pullman, WA, USA
| | - Kiwamu Tanaka
- Department of Plant Pathology, Washington State University, Pullman, WA, USA
| |
Collapse
|
16
|
Nevoa JC, Mendes MT, da Silva MV, Soares SC, Oliveira CJF, Ribeiro JMC. An insight into the salivary gland and fat body transcriptome of Panstrongylus lignarius (Hemiptera: Heteroptera), the main vector of Chagas disease in Peru. PLoS Negl Trop Dis 2018; 12:e0006243. [PMID: 29462134 PMCID: PMC5834209 DOI: 10.1371/journal.pntd.0006243] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 03/02/2018] [Accepted: 01/17/2018] [Indexed: 12/31/2022] Open
Abstract
Triatomines are hematophagous arthropod vectors of Trypanosoma cruzi, the causative agent of Chagas Disease. Panstrongylus lignarius, also known as Panstrongylus herreri, is considered one of the most versatile triatomines because it can parasitize different hosts, it is found in different habitats and countries, it has sylvatic, peridomestic and domestic behavior and it is a very important vector of Chagas disease, especially in Peru. Molecules produced and secreted by salivary glands and fat body are considered of important adaptational value for triatomines because, among other functions, they subvert the host haemostatic, inflammatory and immune systems and detoxify or protect them against environmental aggressors. In this context, the elucidation of the molecules produced by these tissues is highly valuable to understanding the ability of this species to adapt and transmit pathogens. Here, we use high-throughput sequencing techniques to assemble and describe the coding sequences resulting from the transcriptome of the fat body and salivary glands of P. lignarius. The final assembly of both transcriptomes together resulted in a total of 11,507 coding sequences (CDS), which were mapped from a total of 164,676,091 reads. The CDS were subdivided according to their 10 folds overexpression on salivary glands (513 CDS) or fat body (2073 CDS). Among the families of proteins found in the salivary glands, lipocalins were the most abundant. Other ubiquitous families of proteins present in other sialomes were also present in P. lignarius, including serine protease inhibitors, apyrase and antigen-5. The unique transcriptome of fat body showed proteins related to the metabolic function of this organ. Remarkably, nearly 20% of all reads mapped to transcripts coded by Triatoma virus. The data presented in this study improve the understanding on triatomines' salivary glands and fat body function and reveal important molecules used in the interplay between vectors and vertebrate hosts.
Collapse
Affiliation(s)
- Jessica C. Nevoa
- Institute of Natural and Biological Sciences, Laboratory of Immunology, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Maria T. Mendes
- University of Texas at El Paso, El Paso, Texas, United States of America
| | - Marcos V. da Silva
- Institute of Natural and Biological Sciences, Laboratory of Immunology, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Siomar C. Soares
- Institute of Natural and Biological Sciences, Laboratory of Immunology, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Carlo J. F. Oliveira
- Institute of Natural and Biological Sciences, Laboratory of Immunology, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - José M. C. Ribeiro
- National Institute of Allergy and Infectious Diseases (NIAID), Laboratory of Malaria and Vector Research (LMVR), Rockville, Maryland, United States of America
- * E-mail:
| |
Collapse
|
17
|
Santiago PB, de Araújo CN, Charneau S, Bastos IMD, Assumpção TCF, Queiroz RML, Praça YR, Cordeiro TDM, Garcia CHS, da Silva IG, Raiol T, Motta FN, de Araújo Oliveira JV, de Sousa MV, Ribeiro JMC, de Santana JM. Exploring the molecular complexity of Triatoma dimidiata sialome. J Proteomics 2017; 174:47-60. [PMID: 29288089 DOI: 10.1016/j.jprot.2017.12.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/20/2017] [Accepted: 12/22/2017] [Indexed: 01/19/2023]
Abstract
Triatoma dimidiata, a Chagas disease vector widely distributed along Central America, has great capability for domestic adaptation as the majority of specimens caught inside human dwellings or in peridomestic areas fed human blood. Exploring the salivary compounds that overcome host haemostatic and immune responses is of great scientific interest. Here, we provide a deeper insight into its salivary gland molecules. We used high-throughput RNA sequencing to examine in depth the T. dimidiata salivary gland transcriptome. From >51 million reads assembled, 92.21% are related to putative secreted proteins. Lipocalin is the most abundant gene family, confirming it is an expanded family in Triatoma genus salivary repertoire. Other putatively secreted members include phosphatases, odorant binding protein, hemolysin, proteases, protease inhibitors, antigen-5 and antimicrobial peptides. This work expands the previous set of functionally annotated sequences from T. dimidiata salivary glands available in NCBI from 388 to 3815. Additionally, we complemented the salivary analysis through proteomics (available data via ProteomeXchange with identifier PXD008510), disclosing the set complexity of 119 secreted proteins and validating the transcriptomic results. Our large-scale approach enriches the pharmacologically active molecules database and improves our knowledge about the complexity of salivary compounds from haematophagous vectors and their biological interactions. SIGNIFICANCE Several haematophagous triatomine species can transmit Trypanosoma cruzi, the etiological agent of Chagas disease. Due to the reemergence of this disease, new drugs for its prevention and treatment are considered priorities. For this reason, the knowledge of vector saliva emerges as relevant biological finding, contributing to the design of different strategies for vector control and disease transmission. Here we report the transcriptomic and proteomic compositions of the salivary glands (sialome) of the reduviid bug Triatoma dimidiata, a relevant Chagas disease vector in Central America. Our results are robust and disclosed unprecedented insights into the notable diversity of its salivary glands content, revealing relevant anti-haemostatic salivary gene families. Our work expands almost ten times the previous set of functionally annotated sequences from T. dimidiata salivary glands available in NCBI. Moreover, using an integrated transcriptomic and proteomic approach, we showed a correlation pattern of transcription and translation processes for the main gene families found, an important contribution to the research of triatomine sialomes. Furthermore, data generated here reinforces the secreted proteins encountered can greatly contribute for haematophagic habit, Trypanosoma cruzi transmission and development of therapeutic agent studies.
Collapse
Affiliation(s)
- Paula Beatriz Santiago
- Programa Pós-Graduação em Ciências Médicas, Faculty of Medicine, The University of Brasília, Brasília, Brazil
| | - Carla Nunes de Araújo
- Programa Pós-Graduação em Ciências Médicas, Faculty of Medicine, The University of Brasília, Brasília, Brazil; Faculty of Ceilândia, The University of Brasília, Brasília, Brazil.
| | - Sébastien Charneau
- Department of Cell Biology, The University of Brasília, Brasília, Brazil
| | | | - Teresa Cristina F Assumpção
- Vector Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, Rockville, United States
| | | | - Yanna Reis Praça
- Programa Pós-Graduação em Ciências Médicas, Faculty of Medicine, The University of Brasília, Brasília, Brazil
| | | | | | | | - Tainá Raiol
- Department of Cell Biology, The University of Brasília, Brasília, Brazil; Instituto Leônidas e Maria Deane - Fiocruz Amazônia, Manaus, AM, Brazil
| | | | | | | | - José Marcos C Ribeiro
- Vector Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, Rockville, United States
| | - Jaime Martins de Santana
- Programa Pós-Graduação em Ciências Médicas, Faculty of Medicine, The University of Brasília, Brasília, Brazil; Department of Cell Biology, The University of Brasília, Brasília, Brazil
| |
Collapse
|
18
|
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: 106] [Impact Index Per Article: 13.3] [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
|
19
|
Velásquez JJ, Navarro-Vargas JR, Moncada L. Potential pharmacological use of salivary compounds from hematophagous organisms. REVISTA DE LA FACULTAD DE MEDICINA 2017. [DOI: 10.15446/revfacmed.v65n3.52835] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Introducción. La saliva de los artrópodos hematófagos contiene un arsenal de compuestos que les permite acceder a la sangre de sus hospederos vertebrados sin ser detectados.Objetivo. Explorar los compuestos salivares de insectos hematófagos que tienen propiedades vasodilatadoras, anticoagulantes, antiinflamatorias, inmunomoduladoras y anestésicas, las cuales se pueden aprovechar por su alto potencial farmacológico.Materiales y métodos. Se realizó una revisión no sistemática de la literatura mediante búsqueda electrónica en las bases de datos PubMed, EMBASE, OvidSP y ScienceDirect; la búsqueda no se limitó por fecha, idioma ni tipo de artículo. Se buscaron artículos sobre los compuestos salivares de los insectos hematófagos, cuyo tema central fuese los efectos en la hemostasia, inmunomodulación y uso farmacológico. Se encontraron 59 artículos que cumplían con los criterios para ser incluidos en la revisión.Conclusión. La saliva de los insectos hematófagos posee gran variedad de moléculas, lo que ofrece una fuente de investigación y un potencial incalculable para el descubrimiento de compuestos que podrían llegar a tener utilidad farmacológica.
Collapse
|
20
|
Mukbel RM, Khasharmeh RH, Hijjawi NS, Khalifeh MS, Hatmal MM, McDowell MA. Human immune response to salivary proteins of wild-caught Phlebotomus papatasi. Parasitol Res 2016; 115:3345-55. [PMID: 27160331 DOI: 10.1007/s00436-016-5094-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 04/26/2016] [Indexed: 12/15/2022]
Abstract
Phlebotomine sand flies are the known vectors of Leishmania parasites. New approaches in vaccination against Leishmania have investigated the possibility of integrating Phlebotomus papatasi salivary proteins to enhance the immune response and protect against the transmission of the infection. The aim of the present study was to screen human immune responses to wild sand fly saliva and evaluate immunogenic salivary proteins. Blood samples were collected from donors in control and sand fly infested areas. Antibodies specific for sand fly antigens in donor plasma were probed using immunoblotting. In addition, recall proliferation capability of peripheral blood mononuclear cells (PBMC) was tested after sand fly salivary homogenates stimulation. The significant immunogenic salivary proteins (SPs) identified by immunoblotting were SP28, SP32, and SP36. A specific proliferative response of PBMC after stimulation with sand fly salivary homogenates was evident in donors that have antibody responses against sand fly salivary proteins. Individuals with antibody recognition to a higher number of salivary proteins (i.e., 3 or more SP bands) showed lower PBMC proliferative responses after in vitro stimulation with salivary gland homogenates (SGH) only in the sand fly infested, leishmaniasis free area. Interestingly, the presence of a humoral immune response to many SP antigens inversely correlates with a strong cell-mediated immune response (CMI). It was also noticed that some other heavily expressed antigens, in sand fly salivary homogenate, lack or have weak humoral immune reactivity in exposed individuals. Therefore, considering these antigens alone as CMI activators, without including the immunodominant humoral immune response proteins, needs future investigation.
Collapse
Affiliation(s)
- Rami M Mukbel
- Department of Basic Veterinary Medical Science, Faculty of Veterinary Medicine, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan.
| | - Rehab H Khasharmeh
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, The Hashemite University, PO Box 150459, Zarqa, 13115, Jordan
| | - Nawal S Hijjawi
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, The Hashemite University, PO Box 150459, Zarqa, 13115, Jordan
| | - Mohammed S Khalifeh
- Department of Basic Veterinary Medical Science, Faculty of Veterinary Medicine, Jordan University of Science and Technology, P.O. Box 3030, Irbid, 22110, Jordan
| | - Ma'mon M Hatmal
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, The Hashemite University, PO Box 150459, Zarqa, 13115, Jordan
| | - Mary Ann McDowell
- Eck Institute for Global Health, Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, 46556, USA
| |
Collapse
|
21
|
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: 3.1] [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
|
22
|
Walker AA, Weirauch C, Fry BG, King GF. Venoms of Heteropteran Insects: A Treasure Trove of Diverse Pharmacological Toolkits. Toxins (Basel) 2016; 8:43. [PMID: 26907342 PMCID: PMC4773796 DOI: 10.3390/toxins8020043] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 01/25/2016] [Accepted: 01/26/2016] [Indexed: 11/16/2022] Open
Abstract
The piercing-sucking mouthparts of the true bugs (Insecta: Hemiptera: Heteroptera) have allowed diversification from a plant-feeding ancestor into a wide range of trophic strategies that include predation and blood-feeding. Crucial to the success of each of these strategies is the injection of venom. Here we review the current state of knowledge with regard to heteropteran venoms. Predaceous species produce venoms that induce rapid paralysis and liquefaction. These venoms are powerfully insecticidal, and may cause paralysis or death when injected into vertebrates. Disulfide-rich peptides, bioactive phospholipids, small molecules such as N,N-dimethylaniline and 1,2,5-trithiepane, and toxic enzymes such as phospholipase A2, have been reported in predatory venoms. However, the detailed composition and molecular targets of predatory venoms are largely unknown. In contrast, recent research into blood-feeding heteropterans has revealed the structure and function of many protein and non-protein components that facilitate acquisition of blood meals. Blood-feeding venoms lack paralytic or liquefying activity but instead are cocktails of pharmacological modulators that disable the host haemostatic systems simultaneously at multiple points. The multiple ways venom is used by heteropterans suggests that further study will reveal heteropteran venom components with a wide range of bioactivities that may be recruited for use as bioinsecticides, human therapeutics, and pharmacological tools.
Collapse
Affiliation(s)
- Andrew A Walker
- Institute for Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia.
| | - Christiane Weirauch
- Department of Entomology, University of California, Riverside, CA 92521, USA.
| | - Bryan G Fry
- School of Biological Sciences, The University of Queensland, St Lucia, QLD 4072, Australia.
| | - Glenn F King
- Institute for Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia.
| |
Collapse
|
23
|
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: 2.2] [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
|
24
|
Phillips CD, Baker RJ. Secretory Gene Recruitments in Vampire Bat Salivary Adaptation and Potential Convergences With Sanguivorous Leeches. Front Ecol Evol 2015. [DOI: 10.3389/fevo.2015.00122] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
25
|
Ribeiro JMC, Schwarz A, Francischetti IMB. A Deep Insight Into the Sialotranscriptome of the Chagas Disease Vector, Panstrongylus megistus (Hemiptera: Heteroptera). JOURNAL OF MEDICAL ENTOMOLOGY 2015; 52:351-358. [PMID: 26334808 PMCID: PMC4581482 DOI: 10.1093/jme/tjv023] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 02/05/2015] [Indexed: 05/28/2023]
Abstract
Saliva of blood-sucking arthropods contains a complex cocktail of pharmacologically active compounds that assists feeding by counteracting their hosts' hemostatic and inflammatory reactions. Panstrongylus megistus (Burmeister) is an important vector of Chagas disease in South America, but despite its importance there is only one salivary protein sequence publicly deposited in GenBank. In the present work, we used Illumina technology to disclose and publicly deposit 3,703 coding sequences obtained from the assembly of >70 million reads. These sequences should assist proteomic experiments aimed at identifying pharmacologically active proteins and immunological markers of vector exposure. A supplemental file of the transcriptome and deducted protein sequences can be obtained from http://exon.niaid.nih.gov/transcriptome/P_megistus/Pmeg-web.xlsx.
Collapse
Affiliation(s)
- José M C Ribeiro
- National Institute of Allergy and Infectious Diseases, Laboratory of Malaria and Vector Research, 12735 Twinbrook Parkway, MD 20852.
| | - Alexandra Schwarz
- Institute of Parasitology, Biology Centre of the Academy of Sciences of Czech Republic, CZ-370 05 Ceske Budejovice, Czech Republic
| | - Ivo M B Francischetti
- National Institute of Allergy and Infectious Diseases, Laboratory of Malaria and Vector Research, 12735 Twinbrook Parkway, MD 20852
| |
Collapse
|
26
|
Kato H, Gomez EA, Fujita M, Ishimaru Y, Uezato H, Mimori T, Iwata H, Hashiguchi Y. Ayadualin, a novel RGD peptide with dual antihemostatic activities from the sand fly Lutzomyia ayacuchensis, a vector of Andean-type cutaneous leishmaniasis. Biochimie 2015; 112:49-56. [DOI: 10.1016/j.biochi.2015.02.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 02/16/2015] [Indexed: 01/07/2023]
|
27
|
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: 7.5] [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
|
28
|
Vasconcelos CO, Coêlho ZCB, Chaves CDS, Teixeira CR, Pompeu MML, Teixeira MJ. Distinct cellular migration induced by Leishmania infantum chagasi and saliva from Lutzomyia longipalpis in a hemorrhagic pool model. Rev Inst Med Trop Sao Paulo 2014; 56:21-7. [PMID: 24553604 PMCID: PMC4085829 DOI: 10.1590/s0036-46652014000100003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 05/06/2013] [Indexed: 11/22/2022] Open
Abstract
Recruitment of a specific cell population after Leishmania infection can influence the outcome of the disease. Cellular migration in response to Leishmania or vector saliva has been reported in air pouch model, however, cellular migration induced by Leishmania associated with host's blood and vector saliva in this model has not been described. Herein we investigated cellular migration into air pouch of hamster after stimulation with combination of L. chagasi and host's blood and Lutzomyia longipalpis saliva. Migration induced by saliva was 3-fold more than those induced by L. chagasi alone. Additionally, L. chagasi associated with blood and saliva induced significantly even more leukocytes into air pouch than Leishmania alone. L. chagasi recruited a diverse cell population; however, most of these cells seem to have not migrated to the inflammatory exudate, remaining in the pouch lining tissue. These results indicate that L. chagasi can reduce leukocyte accumulation to the initial site of infection, and when associated with vector saliva in the presence of blood components, increase the influx of more neutrophils than macrophages, suggesting that the parasite has developed a strategy to minimize the initial inflammatory response, allowing an unlimited progression within the host. This work reinforces the importance of studies on the salivary components of sand fly vectors of leishmaniasis in the transmission process and the establishment of the infection.
Collapse
Affiliation(s)
- Camila Oliveira Vasconcelos
- Faculdade de Medicina, Universidade Federal do Ceará, 60430-160FortalezaCE, Brazil, Faculdade de Medicina, Universidade Federal do Ceará, Rua Alexandre Baraúna 949, 60430-160 Fortaleza, CE, Brazil
| | - Zirlane C Branco Coêlho
- Faculdade de Medicina, Universidade Federal do Ceará, 60430-160FortalezaCE, Brazil, Faculdade de Medicina, Universidade Federal do Ceará, Rua Alexandre Baraúna 949, 60430-160 Fortaleza, CE, Brazil
| | - Cristina de Souza Chaves
- Faculdade de Medicina, Universidade Federal do Ceará, 60430-160FortalezaCE, Brazil, Faculdade de Medicina, Universidade Federal do Ceará, Rua Alexandre Baraúna 949, 60430-160 Fortaleza, CE, Brazil
| | - Clarissa Romero Teixeira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, RockvilleMaryland, USA, Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Margarida M Lima Pompeu
- Faculdade de Medicina, Universidade Federal do Ceará, 60430-160FortalezaCE, Brazil, Faculdade de Medicina, Universidade Federal do Ceará, Rua Alexandre Baraúna 949, 60430-160 Fortaleza, CE, Brazil
| | - Maria Jania Teixeira
- Faculdade de Medicina, Universidade Federal do Ceará, 60430-160FortalezaCE, Brazil, Faculdade de Medicina, Universidade Federal do Ceará, Rua Alexandre Baraúna 949, 60430-160 Fortaleza, CE, Brazil
| |
Collapse
|
29
|
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: 3.4] [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
|
30
|
Vandermoten S, Harmel N, Mazzucchelli G, De Pauw E, Haubruge E, Francis F. Comparative analyses of salivary proteins from three aphid species. INSECT MOLECULAR BIOLOGY 2014; 23:67-77. [PMID: 24382153 DOI: 10.1111/imb.12061] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Saliva is a critical biochemical interface between aphids and their host plants; however, the biochemical nature and physiological functions of aphid saliva proteins are not fully elucidated. In this study we used a multidisciplinary proteomics approach combining liquid chromatography-electrospray ionization tandem mass spectrometry and two-dimensional differential in-gel electrophoresis/matrix-assisted laser desorption/ionization time-of-flight/mass spectrometry to compare the salivary proteins from three aphid species including Acyrthosiphon pisum, Megoura viciae and Myzus persicae. Comparative analyses revealed variability among aphid salivary proteomes. Among the proteins that varied, 22% were related to DNA-binding, 19% were related to GTP-binding, and 19% had oxidoreductase activity. In addition, we identified a peroxiredoxin enzyme and an ATP-binding protein that may be involved in the modulation of plant defences. Knowledge of salivary components and how they vary among aphid species may reveal how aphids target plant processes and how the aphid and host plant interact.
Collapse
Affiliation(s)
- S Vandermoten
- Gembloux Agro-Bio Tech, Department of Functional and Evolutionary Entomology, University of Liege, Gembloux, Belgium
| | | | | | | | | | | |
Collapse
|
31
|
Tanaka K, Choi J, Cao Y, Stacey G. Extracellular ATP acts as a damage-associated molecular pattern (DAMP) signal in plants. FRONTIERS IN PLANT SCIENCE 2014; 5:446. [PMID: 25232361 PMCID: PMC4153020 DOI: 10.3389/fpls.2014.00446] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 08/19/2014] [Indexed: 05/16/2023]
Abstract
As sessile organisms, plants have evolved effective mechanisms to protect themselves from environmental stresses. Damaged (i.e., wounded) plants recognize a variety of endogenous molecules as danger signals, referred to as damage-associated molecular patterns (DAMPs). ATP is among the molecules that are released by cell damage, and recent evidence suggests that ATP can serve as a DAMP. Although little studied in plants, extracellular ATP is well known for its signaling roles in animals, including acting as a DAMP during the inflammatory response and wound healing. If ATP acts outside the cell, then it is reasonable to expect that it is recognized by a plasma membrane-localized receptor. Recently, DORN1, a lectin receptor kinase, was shown to recognize extracellular ATP in Arabidopsis. DORN1 is the founding member of a new purinoceptor subfamily, P2K (P2 receptor kinase), which is plant-specific. P2K1 (DORN1) is required for ATP-induced cellular responses (e.g., cytosolic Ca(2+) elevation, MAPK phosphorylation, and gene expression). Genetic analysis of loss-of-function mutants and overexpression lines showed that P2K1 participates in the plant wound response, consistent with the role of ATP as a DAMP. In this review, we summarize past research on the roles and mechanisms of extracellular ATP signaling in plants, and discuss the direction of future research on extracellular ATP as a DAMP signal.
Collapse
Affiliation(s)
- Kiwamu Tanaka
- Department of Plant Pathology, Washington State UniversityPullman, WA, USA
- *Correspondence: Kiwamu Tanaka, Department of Plant Pathology, Washington State University, P.O. BOX 646430, Pullman, WA 99164, USA e-mail:
| | - Jeongmin Choi
- Division of Plant Sciences and Biochemistry, Christopher S. Bond Life Sciences Center, University of MissouriColumbia, MO, USA
| | - Yangrong Cao
- Division of Plant Sciences and Biochemistry, Christopher S. Bond Life Sciences Center, University of MissouriColumbia, MO, USA
| | - Gary Stacey
- Division of Plant Sciences and Biochemistry, Christopher S. Bond Life Sciences Center, University of MissouriColumbia, MO, USA
| |
Collapse
|
32
|
Molecular and immunogenic properties of apyrase SP01B and D7-related SP04 recombinant salivary proteins of Phlebotomus perniciosus from Madrid, Spain. BIOMED RESEARCH INTERNATIONAL 2013; 2013:526069. [PMID: 24171166 PMCID: PMC3793307 DOI: 10.1155/2013/526069] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 07/26/2013] [Accepted: 08/16/2013] [Indexed: 11/17/2022]
Abstract
Sand fly salivary proteins are on the spotlight to become vaccine candidates against leishmaniasis and to markers of exposure to sand fly bites due to the host immune responses they elicit. Working with the whole salivary homogenate entails serious drawbacks such as the need for maintaining sand fly colonies and the laborious task of glands dissection. In order to overcome these difficulties, producing recombinant proteins of different vectors has become a major task. In this study, a cDNA library was constructed with the salivary glands of Phlebotomus perniciosus from Madrid, Spain, the most widespread vector of Leishmania infantum in the Mediterranean basin. Analysis of the cDNA sequences showed several polymorphisms among the previously described salivary transcripts. The apyrase SP01B and the D7-related protein SP04 were successfully cloned, expressed in Escherichia coli, and purified. Besides, recombinant proteins were recognized by sera of hamsters and mice previously immunized with saliva through the exposure to uninfected sand fly bites. These results suggest that these two recombinant proteins conserved their immunogenic properties after expression in a prokaryote system. Therefore, this work contributes to expand the knowledge of P. perniciosus saliva that would be eventually used for the development of tools for vector control programs.
Collapse
|
33
|
Collin N, Assumpção TCF, Mizurini DM, Gilmore DC, Dutra-Oliveira A, Kotsyfakis M, Sá-Nunes A, Teixeira C, Ribeiro JMC, Monteiro RQ, Valenzuela JG, Francischetti IMB. Lufaxin, a novel factor Xa inhibitor from the salivary gland of the sand fly Lutzomyia longipalpis blocks protease-activated receptor 2 activation and inhibits inflammation and thrombosis in vivo. Arterioscler Thromb Vasc Biol 2012; 32:2185-98. [PMID: 22796577 DOI: 10.1161/atvbaha.112.253906] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Blood-sucking arthropods' salivary glands contain a remarkable diversity of antihemostatics. The aim of the present study was to identify the unique salivary anticoagulant of the sand fly Lutzomyia longipalpis, which remained elusive for decades. METHODS AND RESULTS Several L. longipalpis salivary proteins were expressed in human embryonic kidney 293 cells and screened for inhibition of blood coagulation. A novel 32.4-kDa molecule, named Lufaxin, was identified as a slow, tight, noncompetitive, and reversible inhibitor of factor Xa (FXa). Notably, Lufaxin's primary sequence does not share similarity to any physiological or salivary inhibitors of coagulation reported to date. Lufaxin is specific for FXa and does not interact with FX, Dansyl-Glu-Gly-Arg-FXa, or 15 other enzymes. In addition, Lufaxin blocks prothrombinase and increases both prothrombin time and activated partial thromboplastin time. Surface plasmon resonance experiments revealed that FXa binds Lufaxin with an equilibrium constant ≈3 nM, and isothermal titration calorimetry determined a stoichiometry of 1:1. Lufaxin also prevents protease-activated receptor 2 activation by FXa in the MDA-MB-231 cell line and abrogates edema formation triggered by injection of FXa in the paw of mice. Moreover, Lufaxin prevents FeCl(3)-induced carotid artery thrombus formation and prolongs activated partial thromboplastin time ex vivo, implying that it works as an anticoagulant in vivo. Finally, salivary gland of sand flies was found to inhibit FXa and to interact with the enzyme. CONCLUSIONS Lufaxin belongs to a novel family of slow-tight FXa inhibitors, which display antithrombotic and anti-inflammatory activities. It is a useful tool to understand FXa structural features and its role in prohemostatic and proinflammatory events.
Collapse
Affiliation(s)
- Nicolas Collin
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Salivary gland transcriptomes and proteomes of Phlebotomus tobbi and Phlebotomus sergenti, vectors of leishmaniasis. PLoS Negl Trop Dis 2012; 6:e1660. [PMID: 22629480 PMCID: PMC3358328 DOI: 10.1371/journal.pntd.0001660] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Accepted: 04/12/2012] [Indexed: 12/22/2022] Open
Abstract
Background Phlebotomus tobbi is a vector of Leishmania infantum, and P. sergenti is a vector of Leishmania tropica. Le. infantum and Le. tropica typically cause visceral or cutaneous leishmaniasis, respectively, but Le. infantum strains transmitted by P. tobbi can cause cutaneous disease. To better understand the components and possible implications of sand fly saliva in leishmaniasis, the transcriptomes of the salivary glands (SGs) of these two sand fly species were sequenced, characterized and compared. Methodology/Principal Findings cDNA libraries of P. tobbi and P. sergenti female SGs were constructed, sequenced, and analyzed. Clones (1,152) were randomly picked from each library, producing 1,142 high-quality sequences from P. tobbi and 1,090 from P. sergenti. The most abundant, secreted putative proteins were categorized as antigen 5-related proteins, apyrases, hyaluronidases, D7-related and PpSP15-like proteins, ParSP25-like proteins, PpSP32-like proteins, yellow-related proteins, the 33-kDa salivary proteins, and the 41.9-kDa superfamily of proteins. Phylogenetic analyses and multiple sequence alignments of putative proteins were used to elucidate molecular evolution and describe conserved domains, active sites, and catalytic residues. Proteomic analyses of P. tobbi and P. sergenti SGs were used to confirm the identification of 35 full-length sequences (18 in P. tobbi and 17 in P. sergenti). To bridge transcriptomics with biology P. tobbi antigens, glycoproteins, and hyaluronidase activity was characterized. Conclusions This analysis of P. sergenti is the first description of the subgenus Paraphlebotomus salivary components. The investigation of the subgenus Larroussius sand fly P. tobbi expands the repertoire of salivary proteins in vectors of Le. infantum. Although P. tobbi transmits a cutaneous form of leishmaniasis, its salivary proteins are most similar to other Larroussius subgenus species transmitting visceral leishmaniasis. These transcriptomic and proteomic analyses provide a better understanding of sand fly salivary proteins across species and subgenera that will be vital in vector-pathogen and vector-host research. Phlebotomine female sand flies require a blood meal for egg development, and it is during the blood feeding that pathogens can be transmitted to a host. Leishmania parasites are among these pathogens and can cause disfiguring cutaneous or even possibly fatal visceral disease. The Leishmania parasites are deposited into the bite wound along with the sand fly saliva. The components of the saliva have many pharmacologic and immune functions important in blood feeding and disease establishment. In this article, the authors identify and investigate the protein components of saliva of two important vectors of leishmaniasis, Phlebotomus tobbi and P. sergenti, by sequencing the transcriptomes of the salivary glands. We then compared the predicted protein sequences of these salivary proteins to those of other bloodsucking insects to elucidate the similarity in composition, structure, and enzymatic activity. Finally, this descriptive analysis of P. tobbi and P. sergenti transcriptomes can aid future research in identifying molecules for epidemiologic assays and in investigating sand fly-host interactions.
Collapse
|
35
|
Ribeiro JMC, Assumpção TCF, Pham VM, Francischetti IMB, Reisenman CE. An insight into the sialotranscriptome of Triatoma rubida (Hemiptera: Heteroptera). JOURNAL OF MEDICAL ENTOMOLOGY 2012; 49:563-72. [PMID: 22679863 PMCID: PMC3544468 DOI: 10.1603/me11243] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
The kissing bug Triatoma rubida (Uhler, 1894) is found in southwestern United States and parts of Mexico where it is found infected with Trypanosoma cruzi, invades human dwellings and causes allergies from their bites. Although the protein salivary composition of several triatomine species is known, not a single salivary protein sequence is known from T. rubida. Furthermore, the salivary diversity of related hematophagous arthropods is very large probably because of the immune pressure from their hosts. Here we report the sialotranscriptome analysis of T. rubida based on the assembly of 1,820 high-quality expressed sequence tags, 51% of which code for putative secreted peptides, including lipocalins, members of the antigen five family, apyrase, hemolysin, and trialysin families. Interestingly, T. rubida lipocalins are at best 40% identical in primary sequence to those of T. protracta, a kissing bug that overlaps its range with T. rubida, indicating the diversity of the salivary lipocalins among species of the same hematophagous genus. We additionally found several expressed sequence tags coding for proteins of clear Trypanosoma spp. origin. This work contributes to the future development of markers of human and pet exposure to T. rubida and to the possible development of desensitization therapies. Supp. Data 1 and 2 (online only) of the transcriptome and deducted protein sequences can be obtained from http://exon.niaid.nih.gov/transcriptome/Trubida/Triru-S1-web.xlsx and http://exon.niaid.nih.gov/transcriptome/Trubida/Triru-S2-web.xlsx.
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, USA.
| | | | | | | | | |
Collapse
|
36
|
Mahamdallie SS, Ready PD. No recent adaptive selection on the apyrase of Mediterranean Phlebotomus: implications for using salivary peptides to vaccinate against canine leishmaniasis. Evol Appl 2012; 5:293-305. [PMID: 25568049 PMCID: PMC3353351 DOI: 10.1111/j.1752-4571.2011.00226.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2011] [Accepted: 11/02/2011] [Indexed: 12/12/2022] Open
Abstract
Vaccine development is informed by a knowledge of genetic variation among antigen alleles, especially the distribution of positive and balancing selection in populations and species. A combined approach using population genetic and phylogenetic methods to detect selective signatures can therefore be informative for identifying vaccine candidates. Parasitic Leishmania species cause the disease leishmaniasis in humans and mammalian reservoir hosts after inoculation by female phlebotomine sandflies. Like other arthropod vectors of disease agents, sandflies use salivary peptides to counteract host haemostatic and immunomodulatory responses during bloodfeeding, and these peptides are vaccine candidates because they can protect against Leishmania infection. We detected no contemporary adaptive selection on one salivary peptide, apyrase, in 20 populations of Phlebotomus ariasi, a European vector of Leishmania infantum. Maximum likelihood branch models on a gene phylogeny showed apyrase to be a single copy in P. ariasi but an ancient duplication event associated with temporary positive selection was observed in its sister group, which contains most Mediterranean vectors of L. infantum. The absence of contemporary adaptive selection on the apyrase of P. ariasi may result from this sandfly's opportunistic feeding behaviour. Our study illustrates how the molecular population genetics of arthropods can help investigate the potential of salivary peptides for disease control and for understanding geographical variation in vector competence.
Collapse
Affiliation(s)
| | - Paul D Ready
- Department of Entomology, Natural History Museum London, UK
| |
Collapse
|
37
|
Grespan R, Lemos HP, Carregaro V, Verri WA, Souto FO, de Oliveira CJF, Teixeira C, Ribeiro JM, Valenzuela JG, Cunha FQ. The protein LJM 111 from Lutzomyia longipalpis salivary gland extract (SGE) accounts for the SGE-inhibitory effects upon inflammatory parameters in experimental arthritis model. Int Immunopharmacol 2012; 12:603-10. [PMID: 22366405 PMCID: PMC3438676 DOI: 10.1016/j.intimp.2012.02.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Revised: 12/12/2011] [Accepted: 02/09/2012] [Indexed: 12/11/2022]
Abstract
Several studies have pointed out the immunomodulatory properties of the Salivary Gland Extract (SGE) from Lutzomyia longipalpis. We aimed to identify the SGE component (s) responsible for its effect on ovalbumin (OVA)-induced neutrophil migration (NM) and to evaluate the effect of SGE and components in the antigen-induced arthritis (AIA) model. We tested the anti-arthritic activities of SGE and the recombinant LJM111 salivary protein (rLJM111) by measuring the mechanical hypernociception and the NM into synovial cavity. Furthermore, we measured IL-17, TNF-α and IFN-γ released by lymph nodes cells stimulated with mBSA or anti-CD3 using enzyme-linked immunosorbent assay (ELISA). Additionally, we tested the effect of SGE and rLJM111 on co-stimulatory molecules expression (MHC-II and CD-86) by flow cytometry, TNF-α and IL-10 production (ELISA) of bone marrow-derived dendritic cells (BMDCs) stimulated with LPS, chemotaxis and actin polymerization from neutrophils. Besides, the effect of SGE on CXCR2 and GRK-2 expression on neutrophils was investigated. We identified one plasmid expressing the protein LJM111 that prevented NM in OVA-challenged immunized mice. Furthermore, both SGE and rLJM111 inhibited NM and pain sensitivity in AIA and reduced IL-17, TNF-α and IFN-γ. SGE and rLJM111 also reduced MHC-II and CD-86 expression and TNF-α whereas increased IL-10 release by LPS-stimulated BMDCs. SGE, but not LJM 111, inhibited neutrophils chemotaxis and actin polymerization. Additionally, SGE reduced neutrophil CXCR2 expression and increased GRK-2. Thus, rLJM111 is partially responsible for SGE mechanisms by diminishing DC function and maturation but not chemoattraction of neutrophils.
Collapse
Affiliation(s)
- Renata Grespan
- Department of Pharmacology, Faculty of Medicine of Ribeirão Preto, University of Sao Paulo, Ribeirão Preto, SP, Brazil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Calì T, Fedrizzi L, Ottolini D, Gomez-Villafuertes R, Mellström B, Naranjo JR, Carafoli E, Brini M. Ca2+-activated nucleotidase 1, a novel target gene for the transcriptional repressor DREAM (downstream regulatory element antagonist modulator), is involved in protein folding and degradation. J Biol Chem 2012; 287:18478-91. [PMID: 22451650 DOI: 10.1074/jbc.m111.304733] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
DREAM is a Ca(2+)-dependent transcriptional repressor highly expressed in neuronal cells. A number of genes have already been identified as the target of its regulation. Targeted analysis performed on cerebella from transgenic mice expressing a dominant active DREAM mutant (daDREAM) showed a drastic reduction of the amount of transcript of Ca(2+)-activated nucleotidase 1 (CANT1), an endoplasmic reticulum (ER)-Golgi resident Ca(2+)-dependent nucleoside diphosphatase that has been suggested to have a role in glucosylation reactions related to the quality control of proteins in the ER and the Golgi apparatus. CANT1 down-regulation was also found in neuroblastoma SH-SY5Y cells stably overexpressing wild type (wt) DREAM or daDREAM, thus providing a simple cell model to investigate the protein maturation pathway. Pulse-chase experiments demonstrated that the down-regulation of CANT1 is associated with reduced protein secretion and increased degradation rates. Importantly, overexpression of wtDREAM or daDREAM augmented the expression of the EDEM1 gene, which encodes a key component of the ER-associated degradation pathway, suggesting an alternative pathway to enhanced protein degradation. Restoring CANT1 levels in neuroblastoma clones recovered the phenotype, thus confirming a key role of CANT1, and of the regulation of its gene by DREAM, in the control of protein synthesis and degradation.
Collapse
Affiliation(s)
- Tito Calì
- Department of Comparative Biomedicine and Food Science, University of Padua, Padua, Italy
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Abdel-Badei NM, Khater EIM, Daba S, Shehata MG. Morphometrics and protein profiles of the salivary glands of Phlebotomus papatasi and Phlebotomus langeroni sand flies. Trans R Soc Trop Med Hyg 2012; 106:235-42. [PMID: 22341687 DOI: 10.1016/j.trstmh.2012.01.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2011] [Revised: 01/13/2012] [Accepted: 01/13/2012] [Indexed: 10/28/2022] Open
Abstract
Sand fly salivary fluid contains numerous proteins that modulate host immune responses to infection and facilitate blood-feeding and establishing Leishmania infection. Salivary proteins are differentially expressed in adaptation to the host, the meal type and ecological factors. We report on the morphometrics and protein composition of salivary glands of colonised Phlebotomus papatasi and P. langeroni sand flies from Egypt. Female glands were dissected at day 1 (D1, unfed), day 2 (D2, sugar-fed), day 3 (D3, blood-fed) and day 7 (D7, blood-digested). The salivary glands are composed of two lobes: heterogeneous in P. papatasi and homogeneous in P. langeroni. Lobe sizes varied considerably with fly age and feeding state; D3 flies had the largest lobe sizes and protein content. The P. papatasi flies had larger lobes and higher protein content than the P. langeroni flies. The P. papatasi D1 flies had 15 protein bands that decreased in the D2, D3 and D7 flies to 10 bands in the Sinai flies and 9 bands in the Alexandria flies. All P. langeroni flies had 12 protein bands but with different intensities. The results reveal inter-specific variation between P. papatasi and P. langeroni, while no intra-specific variation between P. papatasi strains. These results increase our understanding of salivary gland protein composition and blood-feeding behaviour in Old World sand flies with implications for leishmaniasis epidemiology and control.
Collapse
Affiliation(s)
- Noha M Abdel-Badei
- Department of Entomology, Faculty of Science, Ain Shams University, Egypt
| | | | | | | |
Collapse
|
40
|
Manque PA, Woehlbier U, Lara AM, Tenjo F, Alves JM, Buck GA. Identification and characterization of a novel calcium-activated apyrase from Cryptosporidium parasites and its potential role in pathogenesis. PLoS One 2012; 7:e31030. [PMID: 22363541 PMCID: PMC3280346 DOI: 10.1371/journal.pone.0031030] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Accepted: 12/30/2011] [Indexed: 01/20/2023] Open
Abstract
Herein, we report the biochemical and functional characterization of a novel Ca2+-activated nucleoside diphosphatase (apyrase), CApy, of the intracellular gut pathogen Cryptosporidium. The purified recombinant CApy protein displayed activity, substrate specificity and calcium dependency strikingly similar to the previously described human apyrase, SCAN-1 (soluble calcium-activated nucleotidase 1). CApy was found to be expressed in both Cryptosporidium parvum oocysts and sporozoites, and displayed a polar localization in the latter, suggesting a possible co-localization with the apical complex of the parasite. In vitro binding experiments revealed that CApy interacts with the host cell in a dose-dependent fashion, implying the presence of an interacting partner on the surface of the host cell. Antibodies directed against CApy block Cryptosporidium parvum sporozoite invasion of HCT-8 cells, suggesting that CApy may play an active role during the early stages of parasite invasion. Sequence analyses revealed that the capy gene shares a high degree of homology with apyrases identified in other organisms, including parasites, insects and humans. Phylogenetic analysis argues that the capy gene is most likely an ancestral feature that has been lost from most apicomplexan genomes except Cryptosporidium, Neospora and Toxoplasma.
Collapse
Affiliation(s)
- Patricio A. Manque
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Center for the Study of Biological Complexity, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Ute Woehlbier
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Center for the Study of Biological Complexity, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Ana M. Lara
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Center for the Study of Biological Complexity, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Fernando Tenjo
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Center for the Study of Biological Complexity, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Department of Biology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - João M. Alves
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Center for the Study of Biological Complexity, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Gregory A. Buck
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Center for the Study of Biological Complexity, Virginia Commonwealth University, Richmond, Virginia, United States of America
- * E-mail:
| |
Collapse
|
41
|
Chagas AC, Calvo E, Pimenta PFP, Ribeiro JMC. An insight into the sialome of Simulium guianense (DIPTERA:SIMulIIDAE), the main vector of River Blindness Disease in Brazil. BMC Genomics 2011; 12:612. [PMID: 22182526 PMCID: PMC3285218 DOI: 10.1186/1471-2164-12-612] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Accepted: 12/19/2011] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Little is known about the composition and function of the saliva in black flies such as Simulium guianense, the main vector of river blindness disease in Brazil. The complex salivary potion of hematophagous arthropods counteracts their host's hemostasis, inflammation, and immunity. RESULTS Transcriptome analysis revealed ubiquitous salivary protein families--such as the Antigen-5, Yellow, Kunitz domain, and serine proteases--in the S. guianense sialotranscriptome. Insect-specific families were also found. About 63.4% of all secreted products revealed protein families found only in Simulium. Additionally, we found a novel peptide similar to kunitoxin with a structure distantly related to serine protease inhibitors. This study revealed a relative increase of transcripts of the SVEP protein family when compared with Simulium vittatum and S. nigrimanum sialotranscriptomes. We were able to extract coding sequences from 164 proteins associated with blood and sugar feeding, the majority of which were confirmed by proteome analysis. CONCLUSIONS Our results contribute to understanding the role of Simulium saliva in transmission of Onchocerca volvulus and evolution of salivary proteins in black flies. It also consists of a platform for mining novel anti-hemostatic compounds, vaccine candidates against filariasis, and immuno-epidemiologic markers of vector exposure.
Collapse
Affiliation(s)
- Andrezza C Chagas
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, 12735 Twinbrook Parkway, National Institutes of Health, Rockville, Maryland 20892-8132, USA
- Entomology Laboratory, Centro de Pesquisa René Rachou, Belo Horizonte, Minas Gerais, Brazil
| | - Eric Calvo
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, 12735 Twinbrook Parkway, National Institutes of Health, Rockville, Maryland 20892-8132, USA
| | - Paulo FP Pimenta
- Entomology Laboratory, Centro de Pesquisa René Rachou, Belo Horizonte, Minas Gerais, Brazil
| | - José MC Ribeiro
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, 12735 Twinbrook Parkway, National Institutes of Health, Rockville, Maryland 20892-8132, USA
| |
Collapse
|
42
|
An S, Ma D, Wei JF, Yang X, Yang HW, Yang H, Xu X, He S, Lai R. A novel allergen Tab y 1 with inhibitory activity of platelet aggregation from salivary glands of horseflies. Allergy 2011; 66:1420-7. [PMID: 21848516 DOI: 10.1111/j.1398-9995.2011.02683.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Horsefly sting causes allergic reactions in human body. However, our knowledge on horsefly allergens remains poor. OBJECTIVES To identify the novel horsefly allergens and characterize their properties. METHODS A native allergen protein Tab y 1 (apyrase) was purified from the salivary glands of the horsefly Tabanus yao Macquart by gel filtration and ion exchange chromatography. Its sequence was determined by Edman degradation and cDNA cloning. Its allergenicity was assessed by immunoblotting for specific IgE, basophil activation test, skin prick test (SPT), and competitive enzyme-linked immunosorbent assay (ELISA). RESULTS Tab y 1 showed a single diffusion band of 70 kDa on SDS-PAGE. Seventy percent (7/10) of patients with horsefly allergy tested positive to Tab y 1 in SPT; sera from 81% (30/37) of patients reacted to Tab y 1 on western blots. Purified Tab y 1 reduced approximately 42% sera IgE reactivity to horsefly salivary gland extract on a competitive ELISA. Tab y 1 upregulated the expression of CD63 and CCR3 on passively sensitized basophils by up to approximately 4.9-fold. Tab y 1 also showed enzymatic activity to hydrolyze ATP and ADP, and potent antiplatelet aggregation and antithrombotic activities. CONCLUSION The current work identified a novel major allergen of horsefly, Tab y 1, with antiplatelet aggregation and antithrombotic activities, which implicates Tab y 1 in helping horseflies suck host blood, meanwhile causing allergy in their human hosts.
Collapse
Affiliation(s)
- S An
- Biotoxin Units of Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, China
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Fontaine A, Diouf I, Bakkali N, Missé D, Pagès F, Fusai T, Rogier C, Almeras L. Implication of haematophagous arthropod salivary proteins in host-vector interactions. Parasit Vectors 2011; 4:187. [PMID: 21951834 PMCID: PMC3197560 DOI: 10.1186/1756-3305-4-187] [Citation(s) in RCA: 133] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Accepted: 09/28/2011] [Indexed: 01/25/2023] Open
Abstract
The saliva of haematophagous arthropods contains an array of anti-haemostatic, anti-inflammatory and immunomodulatory molecules that contribute to the success of the blood meal. The saliva of haematophagous arthropods is also involved in the transmission and the establishment of pathogens in the host and in allergic responses. This survey provides a comprehensive overview of the pharmacological activity and immunogenic properties of the main salivary proteins characterised in various haematophagous arthropod species. The potential biological and epidemiological applications of these immunogenic salivary molecules will be discussed with an emphasis on their use as biomarkers of exposure to haematophagous arthropod bites or vaccine candidates that are liable to improve host protection against vector-borne diseases.
Collapse
Affiliation(s)
- Albin Fontaine
- Unité de Parasitologie - UMR6236 - IFR48, Antenne Marseille de l'Institut de Recherche Biomédicale des Armées (IRBA), Le Pharo, BP 60109, 13 262 Marseille Cedex 07, France
| | - Ibrahima Diouf
- Unité de Parasitologie - UMR6236 - IFR48, Antenne Marseille de l'Institut de Recherche Biomédicale des Armées (IRBA), Le Pharo, BP 60109, 13 262 Marseille Cedex 07, France
| | - Nawal Bakkali
- Unité de Parasitologie - UMR6236 - IFR48, Antenne Marseille de l'Institut de Recherche Biomédicale des Armées (IRBA), Le Pharo, BP 60109, 13 262 Marseille Cedex 07, France
| | - Dorothée Missé
- Laboratoire de Génétique et Evolution des Maladies infectieuses, UMR 2724 CNRS/IRD, Montpellier, France
| | - Frédéric Pagès
- Unité d'Entomologie Médicale, Antenne Marseille de l'Institut de Recherche Biomédicale des Armées (IRBA), Le Pharo, BP 60109, 13 262 Marseille Cedex 07, France
| | - Thierry Fusai
- Unité de Parasitologie - UMR6236 - IFR48, Antenne Marseille de l'Institut de Recherche Biomédicale des Armées (IRBA), Le Pharo, BP 60109, 13 262 Marseille Cedex 07, France
| | - Christophe Rogier
- Unité de Parasitologie - UMR6236 - IFR48, Antenne Marseille de l'Institut de Recherche Biomédicale des Armées (IRBA), Le Pharo, BP 60109, 13 262 Marseille Cedex 07, France
- Institut Pasteur de Madagascar, B.P. 1274, Ambohitrakely, 101 Antananarivo, Madagascar
| | - Lionel Almeras
- Unité de Parasitologie - UMR6236 - IFR48, Antenne Marseille de l'Institut de Recherche Biomédicale des Armées (IRBA), Le Pharo, BP 60109, 13 262 Marseille Cedex 07, France
| |
Collapse
|
44
|
Dong F, Fu Y, Li X, Jiang J, Sun J, Cheng X. Cloning, expression, and characterization of salivary apyrase from Aedes albopictus. Parasitol Res 2011; 110:931-7. [PMID: 21842387 DOI: 10.1007/s00436-011-2579-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Accepted: 07/27/2011] [Indexed: 12/20/2022]
Abstract
Apyrases (ATP diphosphohydrolase) hydrolyze the phosphodiester bonds of nucleoside tri- and diphosphates to orthophosphate and mononucleodides. They can inhibit platelet activation by depletion of adenosine diphosphate. In the current study, the Escherichia coli expression vector pET-19b equipped with an N-terminal histidine tag was applied to express the apyrase of Aedes albopictus. The gene-coding mature apyrase protein was amplified by RT-PCR and cloned into pET-19b. Soluble apyrase protein with high purity was successfully obtained by utilization of the suitable renaturation approach and Ni-NTA purification column. Four monoclonal antibodies to apyrase from A. albopictus were produced in male BALB/c mice immunized with the renatured apyrase. Using immunofluorescence assay and immunoblotting analysis, recombinant apyrase showed fine consistency with native apyrase. From kinetic analysis, it had a K (m) of 11.6 μM and V (max) of 1.02 nM/S/μg protein for adenosine triphosphate. Adenosine diphosphate-induced platelet aggregation was inhibited by approximately 6% when 0.4 μM recombinant apyrase was added and by about 9.5% when the concentration of recombinant apyrase was 0.8 μM. The effect on platelet aggregation was dose dependent. In conclusion, the apyrase of A. albopictus was cloned and expressed highly in the E. coli expression system. Recombinant apyrase protein showed biological activity, and anti-apyrase monoclonal antibody was also prepared.
Collapse
Affiliation(s)
- Fang Dong
- Department of Microbiology and Parasitology, Shanghai Medical College of Fudan University, No. 138 Yixueyuan Road, Shanghai 200032, China
| | | | | | | | | | | |
Collapse
|
45
|
Hewitson JP, Harcus Y, Murray J, van Agtmaal M, Filbey KJ, Grainger JR, Bridgett S, Blaxter ML, Ashton PD, Ashford DA, Curwen RS, Wilson RA, Dowle AA, Maizels RM. Proteomic analysis of secretory products from the model gastrointestinal nematode Heligmosomoides polygyrus reveals dominance of venom allergen-like (VAL) proteins. J Proteomics 2011; 74:1573-94. [PMID: 21722761 DOI: 10.1016/j.jprot.2011.06.002] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2011] [Revised: 05/20/2011] [Accepted: 06/05/2011] [Indexed: 01/25/2023]
Abstract
The intestinal helminth parasite, Heligmosomoides polygyrus bakeri offers a tractable experimental model for human hookworm infections such as Ancylostoma duodenale and veterinary parasites such as Haemonchus contortus. Parasite excretory-secretory (ES) products represent the major focus for immunological and biochemical analyses, and contain immunomodulatory molecules responsible for nematode immune evasion. In a proteomic analysis of adult H. polygyrus secretions (termed HES) matched to an extensive transcriptomic dataset, we identified 374 HES proteins by LC-MS/MS, which were distinct from those in somatic extract HEx, comprising 446 identified proteins, confirming selective export of ES proteins. The predominant secreted protein families were proteases (astacins and other metalloproteases, aspartic, cysteine and serine-type proteases), lysozymes, apyrases and acetylcholinesterases. The most abundant products were members of the highly divergent venom allergen-like (VAL) family, related to Ancylostoma secreted protein (ASP); 25 homologues were identified, with VAL-1 and -2 also shown to be associated with the parasite surface. The dominance of VAL proteins is similar to profiles reported for Ancylostoma and Haemonchus ES products. Overall, this study shows that the secretions of H. polygyrus closely parallel those of clinically important GI nematodes, confirming the value of this parasite as a model of helminth infection.
Collapse
Affiliation(s)
- James P Hewitson
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh EH9 3JT, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
A calcium-activated nucleotidase secreted from Ostertagia ostertagi 4th-stage larvae is a member of the novel salivary apyrases present in blood-feeding arthropods. Parasitology 2011; 138:333-43. [PMID: 20809998 DOI: 10.1017/s0031182010001241] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Apyrases (ATP-diphosphohydrolase) comprise a ubiquitous class of glycosylated nucleotidases that hydrolyse extracellular ATP and ADP to orthophosphate and AMP. One class of newly-described, Ca2+-dependent, salivary apyrases known to counteract blood-clotting, has been identified in haematophagous arthropods. Herein, we have identified a gene (Oos-apy-1) encoding a protein that structurally conforms to the Ca2+-activated apyrase from the bed bug, Cimex lectularius, by immunologically screening an Ostertagia L4 cDNA expression library. The expressed protein (rOos-APY-1) was biochemically functional in the presence of Ca2+ only, with greatest activity on ATP, ADP, UTP and UDP. Host antibodies to the fusion protein appeared as early as 14 days post-infection (p.i.) and increased through 30 days p.i. Immunohistochemical and Western blot analyses demonstrated that the native Oos-APY-1 protein is present in the glandular bulb of the oesophagus and is confined to the L4. A putative signal sequence at the N-terminus and near 100% identity with a Teladorsagia circumcincta L4 secreted protein is consistent with the native protein being secreted at the cellular level. Predicated upon substrate specificity, the native protein may be used by the parasite to control the levels of host extracellular nucleotides released by locally-damaged tissues in an effort to modulate immune intervention and inflammation.
Collapse
|
47
|
Francischetti IMB, Calvo E, Andersen JF, Pham VM, Favreau AJ, Barbian KD, Romero A, Valenzuela JG, Ribeiro JMC. Insight into the Sialome of the Bed Bug, Cimex lectularius. J Proteome Res 2010; 9:3820-31. [PMID: 20441151 DOI: 10.1021/pr1000169] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The evolution of insects to a blood diet leads to the development of a saliva that antagonizes their hosts' hemostasis and inflammation. Hemostasis and inflammation are redundant processes, and thus a complex salivary potion composed of dozens or near 100 different polypeptides is commonly found by transcriptome or proteome analysis of these organisms. Several insect orders or families evolved independently to hematophagy, creating unique salivary potions in the form of novel pharmacological use of endogenous substances and in the form of unique proteins not matching other known proteins, these probably arriving by fast evolution of salivary proteins as they evade their hosts' immune response. In this work we present a preliminary description of the sialome (from the Greek Sialo = saliva) of the common bed bug Cimex lectularius, the first such work from a member of the Cimicidae family. This manuscript is a guide for the supplemental database files http://exon.niaid.nih.gov/transcriptome/C_lectularius/S1/Cimex-S1.zip and http://exon.niaid.nih.gov/transcriptome/C_lectularius/S2/Cimex-S2.xls.
Collapse
Affiliation(s)
- Ivo M B Francischetti
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, Maryland 20852, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Las glándulas salivales de dos flebotominos vectores de Leishmania: Lutzomyia migonei (França) y Lutzomyia ovallesi (Ortiz) (Diptera: Psychodidae). BIOMEDICA 2010. [DOI: 10.7705/biomedica.v30i3.274] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
49
|
Ribeiro JMC, Valenzuela JG, Pham VM, Kleeman L, Barbian KD, Favreau AJ, Eaton DP, Aoki V, Hans-Filho G, Rivitti EA, Diaz LA. An insight into the sialotranscriptome of Simulium nigrimanum, a black fly associated with fogo selvagem in South America. Am J Trop Med Hyg 2010; 82:1060-75. [PMID: 20519601 DOI: 10.4269/ajtmh.2010.09-0769] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Pemphigus foliaceus is a life threatening skin disease that is associated with autoimmunity to desmoglein, a skin protein involved in the adhesion of keratinocytes. This disease is endemic in certain areas of South America, suggesting the mediation of environmental factors triggering autoimmunity. Among the possible environmental factors, exposure to bites of black flies, in particular Simulium nigrimanum has been suggested. In this work, we describe the sialotranscriptome of adult female S. nigrimanum flies. It reveals the complexity of the salivary potion of this insect, comprised by over 70 distinct genes within over 30 protein families, including several novel families, even when compared with the previously described sialotranscriptome of the autogenous black fly, S. vittatum. The uncovering of this sialotranscriptome provides a platform for testing pemphigus patient sera against recombinant salivary proteins from S. nigrimanum and for the discovery of novel pharmacologically active compounds.
Collapse
Affiliation(s)
- José M C Ribeiro
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, MA, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Flores G, Qian Y, Díaz LA. The enigmatic autoimmune response in endemic pemphigus foliaceus. ACTAS DERMO-SIFILIOGRAFICAS 2010; 100 Suppl 2:40-8. [PMID: 20096161 DOI: 10.1016/s0001-7310(09)73377-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Endemic pemphigus foliaceus, known as Fogo Selvagem (FS) in Brazil, is a disease characterized by subcorneal blisters and IgG4 anti-dermoglein 1 (Dsg1) autoantibodies. Epidemiological studies of FS strongly an environmental etiology. A 15-year surveillance of the Limao Verde Amerindian reservation in Brazil has uncovered information on the transition of the autoimmune response from the pre-clinical stage to disease state. This incubation time may evolve over several years. The serological markers of the pre-clinical state of FS are IgM anti-Dsg1, IgE and non-IgG4 autoantibodies against Dsg1. The disease stage of FS is characterized by the rise of pathogenic IgG4 anti-Dsg1 autoantibodies. In this review, the authors reviewed the literature on the relevance of the humoral autoimmune response of FS as well as the possible environmental triggers of anti-Dsg1 autoantibody formation. Based on epidemiological observations, the authors hypothesize that the pathogenic IgG4 response in FS may be triggered by hematophagous insect bites.
Collapse
Affiliation(s)
- G Flores
- Department of Dermatology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| | | | | |
Collapse
|