1
|
Chea S, Willen L, Nhek S, Ly P, Tang K, Oristian J, Salas-Carrillo R, Ponce A, Leon PCV, Kong D, Ly S, Sath R, Lon C, Leang R, Huy R, Yek C, Valenzuela JG, Calvo E, Manning JE, Oliveira F. Antibodies to Aedes aegypti D7L salivary proteins as a new serological tool to estimate human exposure to Aedes mosquitoes. Front Immunol 2024; 15:1368066. [PMID: 38751433 PMCID: PMC11094246 DOI: 10.3389/fimmu.2024.1368066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 04/03/2024] [Indexed: 05/18/2024] Open
Abstract
Introduction Aedes spp. are the most prolific mosquito vectors in the world. Found on every continent, they can effectively transmit various arboviruses, including the dengue virus which continues to cause outbreaks worldwide and is spreading into previously non-endemic areas. The lack of widely available dengue vaccines accentuates the importance of targeted vector control strategies to reduce the dengue burden. High-throughput tools to estimate human-mosquito contact and evaluate vector control interventions are lacking. We propose a novel serological tool that allows rapid screening of human cohorts for exposure to potentially infectious mosquitoes. Methods We tested 563 serum samples from a longitudinal pediatric cohort study previously conducted in Cambodia. Children enrolled in the study were dengue-naive at baseline and were followed biannually for dengue incidence for two years. We used Western blotting and enzyme-linked immunosorbent assays to identify immunogenic Aedes aegypti salivary proteins and measure total anti-Ae. aegypti IgG. Results We found a correlation (rs=0.86) between IgG responses against AeD7L1 and AeD7L2 recombinant proteins and those to whole salivary gland homogenate. We observed seasonal fluctuations of AeD7L1+2 IgG responses and no cross-reactivity with Culex quinquefasciatus and Anopheles dirus mosquitoes. The baseline median AeD7L1+2 IgG responses for young children were higher in those who developed asymptomatic versus symptomatic dengue. Discussion The IgG response against AeD7L1+2 recombinant proteins is a highly sensitive and Aedes specific marker of human exposure to Aedes bites that can facilitate standardization of future serosurveys and epidemiological studies by its ability to provide a robust estimation of human-mosquito contact in a high-throughput fashion.
Collapse
Affiliation(s)
- Sophana Chea
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, Phnom Penh, Cambodia
| | - Laura Willen
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Sreynik Nhek
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, Phnom Penh, Cambodia
| | - Piseth Ly
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, Phnom Penh, Cambodia
| | - Kristina Tang
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - James Oristian
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Roberto Salas-Carrillo
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Aiyana Ponce
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Paola Carolina Valenzuela Leon
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Dara Kong
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, Phnom Penh, Cambodia
| | - Sokna Ly
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Ratanak Sath
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Chanthap Lon
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, Phnom Penh, Cambodia
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Rithea Leang
- National Center for Parasitology, Entomology, and Malaria Control, Ministry of Health, Phnom Penh, Cambodia
- National Dengue Control Program, Ministry of Health, Phnom Penh, Cambodia
| | - Rekol Huy
- National Center for Parasitology, Entomology, and Malaria Control, Ministry of Health, Phnom Penh, Cambodia
| | - Christina Yek
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, Phnom Penh, Cambodia
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Jesus G. Valenzuela
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Eric Calvo
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Jessica E. Manning
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, Phnom Penh, Cambodia
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Fabiano Oliveira
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, Phnom Penh, Cambodia
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
2
|
de Araujo FF, Abdeladhim M, Teixeira C, Hummer K, Wilkerson MD, Ressner R, Lakhal-Naouar I, Ellis MW, Meneses C, Nurmukhambetova S, Gomes R, Tolbert WD, Turiansky GW, Pazgier M, Oliveira F, Valenzuela JG, Kamhawi S, Aronson N. Immune response profiles from humans experimentally exposed to Phlebotomus duboscqi bites. Front Immunol 2024; 15:1335307. [PMID: 38633260 PMCID: PMC11021656 DOI: 10.3389/fimmu.2024.1335307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 03/15/2024] [Indexed: 04/19/2024] Open
Abstract
Introduction Cutaneous leishmaniasis is a neglected vector-borne parasitic disease prevalent in 92 countries with approximately one million new infections annually. Interactions between vector saliva and the human host alter the response to infection and outcome of disease. Methods To characterize the human immunological responses developed against saliva of Phlebotomus duboscqi, a Leishmania major (L. major) vector, we repeatedly exposed the arms of 14 healthy U.S volunteers to uninfected P. duboscqi bites. Blood was collected a week after each exposure and used to assess total IgG antibodies against the proteins of P. duboscqi salivary gland homogenate (SGH) and the levels of IFN-gamma and IL-10 from peripheral blood mononuclear cells (PBMCs) stimulated with SGH or recombinant sand fly proteins. We analyzed skin punch biopsies of the human volunteer arms from the insect bite site and control skin site after multiple P. duboscqi exposures (four volunteers) using immunohistochemical staining. Results A variety of immediate insect bite skin reactions were observed. Late skin reactions to insect bites were characterized by macular hyperpigmentation and/or erythematous papules. Hematoxylin and eosin staining showed moderate mononuclear skin infiltrate with eosinophils in those challenged recently (within 2 months), eosinophils were not seen in biopsies with recall challenge (6 month post bites). An increase in plasma antigen-specific IgG responses to SGH was observed over time. Western Blot results showed strong plasma reactivity to five P. duboscqi salivary proteins. Importantly, volunteers developed a cellular immunity characterized by the secretion of IFN-gamma upon PBMC stimulation with P. duboscqi SGH and recombinant antigens. Discussion Our results demonstrate that humans mounted a local and systemic immune response against P. duboscqi salivary proteins. Specifically, PduM02/SP15-like and PduM73/adenosine deaminase recombinant salivary proteins triggered a Th1 type immune response that might be considered in future development of a potential Leishmania vaccine.
Collapse
Affiliation(s)
- Fernanda Fortes de Araujo
- Infectious Disease Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Maha Abdeladhim
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research (LMVR), National Institutes of Allergy and Infectious Diseases, NIH, Rockville, MD, United States
| | - Clarissa Teixeira
- Department of Biotechnology, Laboratory of Immunoparasitology, Oswaldo Cruz Foundation, Eusébio, CE, Brazil
| | - Kelly Hummer
- Infectious Disease Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Matthew D. Wilkerson
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Roseanne Ressner
- Infectious Disease Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Ines Lakhal-Naouar
- Infectious Disease Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | | | - Claudio Meneses
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research (LMVR), National Institutes of Allergy and Infectious Diseases, NIH, Rockville, MD, United States
| | - Saule Nurmukhambetova
- Infectious Disease Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Regis Gomes
- Department of Biotechnology, Laboratory of Immunoparasitology, Oswaldo Cruz Foundation, Eusébio, CE, Brazil
| | - W. David Tolbert
- Infectious Disease Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - George W. Turiansky
- Department of Dermatology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Marzena Pazgier
- Infectious Disease Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Fabiano Oliveira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research (LMVR), National Institutes of Allergy and Infectious Diseases, NIH, Rockville, MD, United States
| | - Jesus G. Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research (LMVR), National Institutes of Allergy and Infectious Diseases, NIH, Rockville, MD, United States
| | - Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research (LMVR), National Institutes of Allergy and Infectious Diseases, NIH, Rockville, MD, United States
| | - Naomi Aronson
- Infectious Disease Division, Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
3
|
Almeida GG, Luehring TAM, Paixão PHDM, Soares RP, de Barros ALB, do Monte-Neto RL, Tafuri WL, Negrão-Corrêa DA, Gonçalves R. The absence of eosinophils is associated with early metastatic lesions in Leishmania amazonensis-infected mice. Mem Inst Oswaldo Cruz 2024; 119:e220242. [PMID: 38198296 PMCID: PMC10777375 DOI: 10.1590/0074-02760220242] [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: 10/13/2022] [Accepted: 12/08/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Eosinophils are granulocytes that rapidly increase frequency in the bloodstream during helminthic infections and allergic responses. They are found in tissue infected by Leishmania during early disease, but their role during infection is not entirely understood. OBJECTIVES We aim to compare the disease due to Leishmania amazonensis in BALB/c and Δdbl-GATA1 mice, which lack eosinophils. METHODS BALB/c and Δdbl-GATA1 mice infected with L. amazonensis were observed for several weeks. The parasite load and dissemination pattern were assessed. FINDINGS The Δdbl-GATA1 mice developed an anticipated dissemination of L. amazonensis and a worsening disease. No differences were found in the lesion development or the parasite load in the footpad among Δdbl-GATA1 mice and BALB/c eight weeks after infection. However, nine weeks after infection, massive growth of metastatic lesions appeared in several parts of the skin in Δdbl-GATA1 mice, weeks earlier than BALB/c. We observed increased parasites in the bloodstream, probably an essential dissemination route. Thirteen weeks after infection, metastatic lesions were found in all Δdbl-GATA1 mice. MAIN CONCLUSION These results suggest a protective role of eosinophils in delaying the disease caused by L. amazonensis, although several limitations of this mice strain must be considered.
Collapse
Affiliation(s)
- Gregório Guilherme Almeida
- Universidade Federal de Minas Gerais, Instituto de Ciências Biológicas, Departamento de Patologia Geral, Belo Horizonte, MG, Brasil
| | | | | | - Rodrigo Pedro Soares
- Fundação Oswaldo Cruz-Fiocruz, Instituto René Rachou, Grupo de Biotecnologia Aplicada a Patógenos, Belo Horizonte, MG, Brasil
| | - André Luís Branco de Barros
- Universidade Federal de Minas Gerais, Faculdade de Farmácia, Departamento de Análises Clínicas e Toxicológicas, Belo Horizonte, MG, Brasil
| | - Rubens Lima do Monte-Neto
- Fundação Oswaldo Cruz-Fiocruz, Instituto René Rachou, Grupo de Biotecnologia Aplicada a Patógenos, Belo Horizonte, MG, Brasil
| | - Wagner Luiz Tafuri
- Universidade Federal de Minas Gerais, Instituto de Ciências Biológicas, Departamento de Patologia Geral, Belo Horizonte, MG, Brasil
| | - Deborah Aparecida Negrão-Corrêa
- Universidade Federal de Minas Gerais, Instituto de Ciências Biológicas, Departamento de Parasitologia, Belo Horizonte, MG, Brasil
| | - Ricardo Gonçalves
- Universidade Federal de Minas Gerais, Instituto de Ciências Biológicas, Departamento de Patologia Geral, Belo Horizonte, MG, Brasil
| |
Collapse
|
4
|
Chea S, Willen L, Nhek S, Ly P, Tang K, Oristian J, Salas-Carrillo R, Ponce A, Leon PCV, Kong D, Ly S, Sath R, Lon C, Leang R, Huy R, Yek C, Valenzuela JG, Calvo E, Manning JE, Oliveira F. Antibodies to Aedes aegypti D7L salivary proteins as a new serological tool to estimate human exposure to Aedes mosquitoes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.22.23300438. [PMID: 38318204 PMCID: PMC10843157 DOI: 10.1101/2023.12.22.23300438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
Introduction Aedes spp. are the most prolific mosquito vectors in the world. Found on every continent, they can effectively transmit various arboviruses, including the dengue virus which continues to cause outbreaks worldwide and is spreading into previously non-endemic areas. The lack of widely available dengue vaccines accentuates the importance of targeted vector control strategies to reduce the dengue burden. High-throughput sensitive tools to estimate human-mosquito contact and evaluate vector control interventions are lacking. We propose a novel serological tool that allows rapid screening of large human cohorts for exposure to potentially infectious mosquitoes and effective targeting of vector control. Methods We tested 563 serum samples from a longitudinal pediatric cohort study previously conducted in Cambodia. Children enrolled in the study were dengue-naïve at baseline and were followed biannually for dengue incidence for two years. We used Western blotting and enzyme-linked immunosorbent assays to identify the most immunogenic Aedes aegypti salivary proteins and measure total anti- Ae. Aegypti IgG. Results We found a strong correlation (r s =0.86) between the combined IgG responses against AeD7L1 and AeD7L2 recombinant proteins and those to whole salivary gland homogenate. We observed seasonal fluctuations of AeD7L1+2 IgG responses, corresponding to Aedes spp. abundance in the region, and no cross-reactivity with Culex quinquefasciatus and Anopheles dirus mosquitoes. The baseline median AeD7L1+2 IgG responses for young children were higher in those who developed asymptomatic dengue versus those who developed symptomatic dengue. Conclusion The IgG response against AeD7L1+2 recombinant proteins is a highly sensitive and Aedes specific marker of human exposure to Aedes bites that can facilitate standardization of future serosurveys and epidemiological studies by its ability to provide a robust estimation of human-mosquito contact in a high-throughput fashion.
Collapse
|
5
|
Souissi C, Marzouki S, Elbini-Dhouib I, Jebali J, Oliveira F, Valenzuela JG, Srairi-Abid N, Kamhawi S, Ben Ahmed M. PpSP32, the Phlebotomus papatasi immunodominant salivary protein, exerts immunomodulatory effects on human monocytes, macrophages, and lymphocytes. Parasit Vectors 2023; 16:1. [PMID: 36593519 PMCID: PMC9806891 DOI: 10.1186/s13071-022-05627-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/19/2022] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND The saliva of sand flies, vectors of Leishmania parasites, contains several components that exert pharmacological activity facilitating the acquisition of blood by the insect and contributing to the establishment of infection. Previously, we demonstrated that PpSP32 is the immunodominant salivary antigen in humans exposed to Phlebotomus papatasi bites and validated its usefulness as a predictive biomarker of disease. PpSP32, whose functions are little known to date, is an intriguing protein due to its involvement in the etiopathogenesis of pemphigus, an auto-immune disease. Herein, we aimed to better decipher its role through the screening of several immunomodulatory activity either on lymphocytes or on monocytes/macrophages. METHODS Peripheral mononuclear cells from healthy volunteers were stimulated with anti-CD3/anti-CD28 antibodies, phytohemagglutinin, phorbol 12-myristate 13-acetate/ionomycin, or lipopolysaccharide in the presence of increasing doses of PpSP32. Cell proliferation was measured after the addition of tritiated thymidine. Monocyte activation was tested by analyzing the expression of CD86 and HLA-DR molecules by flow cytometry. Cytokine production was analyzed in culture supernatants by ELISA. THP-1-derived macrophages were stimulated with LPS in the presence of increasing doses of PpSP32, and cytokine production was analyzed in culture supernatants by ELISA and multiplex technique. The effect of PpSP32 on NF-kB signaling was tested by Western blot. The anti-inflammatory activity of PpSP32 was assessed in vivo in an experimental inflammatory model of carrageenan-induced paw edema in rats. RESULTS Our data showed that PpSP32 down-modulated the expression of activation markers in LPS-stimulated monocytes and THP1-derived macrophages. This protein negatively modulated the secretion of Th1 and Th2 cytokines by human lymphocytes as well as pro-inflammatory cytokines by monocytes, and THP1-derived macrophages. PpSP32 treatment led to a dose-dependent reduction of IκB phosphorylation. When PpSP32 was injected into the paw of carrageenan-injected rats, edema was significantly reduced. CONCLUSIONS Our data indicates that PpSP32 induces a potent immunomodulatory effect on monocytes and THP-1-derived macrophages. This inhibition could be mediated, among others, by the modulation of the NF-kB signaling pathway. The anti-inflammatory activity of PpSP32 was confirmed in vivo in the carrageenan-induced paw edema model in rats.
Collapse
Affiliation(s)
- Cyrine Souissi
- grid.418517.e0000 0001 2298 7385Laboratory of Transmission, Control and Immunobiology of Infections (LTCII), LR11IPT02, Pasteur Institute de Tunis, Tunis, Tunisia
| | - Soumaya Marzouki
- grid.418517.e0000 0001 2298 7385Laboratory of Transmission, Control and Immunobiology of Infections (LTCII), LR11IPT02, Pasteur Institute de Tunis, Tunis, Tunisia
| | - Ines Elbini-Dhouib
- grid.12574.350000000122959819Laboratory of Biomolecules, Venoms and Theranostic Applications, LR20IPT01, Pasteur Institute of Tunis, University of Tunis El Manar, 1002 Tunis, Tunisia
| | - Jed Jebali
- grid.12574.350000000122959819Laboratory of Biomolecules, Venoms and Theranostic Applications, LR20IPT01, Pasteur Institute of Tunis, University of Tunis El Manar, 1002 Tunis, Tunisia
| | - Fabiano Oliveira
- grid.94365.3d0000 0001 2297 5165Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institute of Health, Rockville, MD USA
| | - Jesus G. Valenzuela
- grid.94365.3d0000 0001 2297 5165Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institute of Health, Rockville, MD USA
| | - Najet Srairi-Abid
- grid.12574.350000000122959819Laboratory of Biomolecules, Venoms and Theranostic Applications, LR20IPT01, Pasteur Institute of Tunis, University of Tunis El Manar, 1002 Tunis, Tunisia
| | - Shaden Kamhawi
- grid.94365.3d0000 0001 2297 5165Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institute of Health, Rockville, MD USA
| | - Melika Ben Ahmed
- grid.418517.e0000 0001 2298 7385Laboratory of Transmission, Control and Immunobiology of Infections (LTCII), LR11IPT02, Pasteur Institute de Tunis, Tunis, Tunisia ,grid.12574.350000000122959819Faculty of Medicine de Tunis, University of Tunis El Manar, Tunis, Tunisia
| |
Collapse
|
6
|
Guerrero D, Vo HTM, Lon C, Bohl JA, Nhik S, Chea S, Man S, Sreng S, Pacheco AR, Ly S, Sath R, Lay S, Missé D, Huy R, Leang R, Kry H, Valenzuela JG, Oliveira F, Cantaert T, Manning JE. Evaluation of cutaneous immune response in a controlled human in vivo model of mosquito bites. Nat Commun 2022; 13:7036. [PMID: 36396947 PMCID: PMC9672097 DOI: 10.1038/s41467-022-34534-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 10/27/2022] [Indexed: 11/18/2022] Open
Abstract
Mosquito-borne viruses are a growing global threat. Initial viral inoculation occurs in the skin via the mosquito 'bite', eliciting immune responses that shape the establishment of infection and pathogenesis. Here we assess the cutaneous innate and adaptive immune responses to controlled Aedes aegypti feedings in humans living in Aedes-endemic areas. In this single-arm, cross-sectional interventional study (trial registration #NCT04350905), we enroll 30 healthy adult participants aged 18 to 45 years of age from Cambodia between October 2020 and January 2021. We perform 3-mm skin biopsies at baseline as well as 30 min, 4 h, and 48 h after a controlled feeding by uninfected Aedes aegypti mosquitos. The primary endpoints are measurement of changes in early and late innate responses in bitten vs unbitten skin by gene expression profiling, immunophenotyping, and cytokine profiling. The results reveal induction of neutrophil degranulation and recruitment of skin-resident dendritic cells and M2 macrophages. As the immune reaction progresses T cell priming and regulatory pathways are upregulated along with a shift to Th2-driven responses and CD8+ T cell activation. Stimulation of participants' bitten skin cells with Aedes aegypti salivary gland extract results in reduced pro-inflammatory cytokine production. These results identify key immune genes, cell types, and pathways in the human response to mosquito bites and can be leveraged to inform and develop novel therapeutics and vector-targeted vaccine candidates to interfere with vector-mediated disease.
Collapse
Affiliation(s)
- David Guerrero
- Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Hoa Thi My Vo
- Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Chanthap Lon
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia
| | - Jennifer A Bohl
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Sreynik Nhik
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia
| | - Sophana Chea
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia
| | - Somnang Man
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia
- National Center of Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Sokunthea Sreng
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia
- National Center of Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Andrea R Pacheco
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia
| | - Sokna Ly
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia
| | - Rathanak Sath
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia
| | - Sokchea Lay
- Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Dorothée Missé
- MIVEGEC, Univ. Montpellier, IRD, CNRS, 34000, Montpellier, France
| | - Rekol Huy
- National Center of Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Rithea Leang
- National Center of Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Hok Kry
- Kampong Speu Provincial District, Ministry of Health, Phnom Penh, Cambodia
| | - Jesus G Valenzuela
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Fabiano Oliveira
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Tineke Cantaert
- Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Jessica E Manning
- International Center of Excellence in Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh, Cambodia.
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA.
| |
Collapse
|
7
|
Carvalho AM, Viana SM, Andrade BB, Oliveira F, Valenzuela JG, Carvalho EM, de Oliveira CI. Immune response to LinB13, a Lutzomyia intermedia salivary protein correlates with disease severity in tegumentary leishmaniasis. Clin Infect Dis 2022; 75:1754-1762. [PMID: 35385578 DOI: 10.1093/cid/ciac258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND We have previously shown that seropositivity to rLinB-13, a salivary protein from Lutzomyia intermedia, predicted sand fly exposure and was associated with increased risk of developing cutaneous leishmaniasis (CL). METHODS Herein, we investigated the cellular immune response to saliva from Lu. intermedia, using rLinB-13 as a surrogate antigen in naturally exposed individuals presenting positive serology to LinB-13. We also investigated the response to rLinB-13 in leishmaniasis patients, displaying active ulcers and positive PCR for L. braziliensis. RESULTS Peripheral blood mononuclear cells (PBMCs) stimulated in vitro with rLinB-13 secreted elevated levels of IL-10, IL-4, IL-1β, IL-1α, IL-6 and chemokines (CCL3, CCL4, CCL5 and CXCL5). CL, and disseminated leishmaniasis (DL) patients displayed a significantly higher IgG response to rLinB-13, compared to healthy subjects and anti-rLinB-13 IgG was positively correlated with the number of lesions in DL patients. Positive serology to rLinB-13 was also associated with chemotherapy failure. PBMCs from DL patients stimulated with rLINB-13 secreted significantly higher levels IL-10 and IL-1β compared to CL individuals. CONCLUSIONS In this study, we observed an association between humoral and cellular immune response to the sand fly salivary protein rLinB-13 and disease severity in tegumentary leishmaniasis. This study brings evidence that immunity to rLinB-13 influences disease outcome in L. braziliensis infection and results indicate that positive serology to rLinB-13 IgG can be employed as marker of DL, an emerging and severe form of disease caused by L. braziliensis.
Collapse
Affiliation(s)
- Augusto M Carvalho
- Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Bahia, Brazil.,Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Salvador, Bahia, Brazil
| | | | | | - Fabiano Oliveira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Jesus G Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Edgar M Carvalho
- Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Bahia, Brazil.,Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Salvador, Bahia, Brazil.,Immunology Service of the University Hospital Professor Edgard Santos, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Camila I de Oliveira
- Instituto Gonçalo Moniz, FIOCRUZ, Salvador, Bahia, Brazil.,Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Salvador, Bahia, Brazil
| |
Collapse
|
8
|
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: 1.0] [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
|
9
|
Parkash V, Kaye PM, Layton AM, Lacey CJ. Vaccines against leishmaniasis: using controlled human infection models to accelerate development. Expert Rev Vaccines 2021; 20:1407-1418. [PMID: 34664543 PMCID: PMC9835556 DOI: 10.1080/14760584.2021.1991795] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 10/07/2021] [Indexed: 01/16/2023]
Abstract
INTRODUCTION Leishmaniasis is a neglected tropical disease that is defined by the World Health Organization as vaccine preventable. Although several new candidate vaccines are in development, no vaccine has successfully reached the market for human use. Several species of Leishmania cause human disease and have co-evolved with their respective sand fly vectors. These unique relationships have implications for initiation of infection and vaccine development. An approach to vaccine development for many infectious diseases is the use of controlled human infection models (CHIMs). AREAS COVERED We describe the history and recent development of experimental and deliberate infection using Leishmania in humans and the rationale for developing a new sand fly-initiated CHIM to progress leishmaniasis vaccine development. Examples from other infectious diseases are discussed in the context of the development of a new leishmaniasis CHIM. We also reflect upon the manufacture of the challenge agent, practical considerations, safety, ethics, and regulatory issues. EXPERT OPINION A new cutaneous Leishmania CHIM is being developed to enable testing of vaccines in the development pipeline. Questions remain about the use of such CHIMs to determine effectiveness of vaccines against visceral leishmaniasis. However, such a CHIM will be invaluable in expediting time to market for vaccines.
Collapse
Affiliation(s)
- Vivak Parkash
- York Biomedical Research Institute, Hull York Medical School, University of York, York, UK
- Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals Nhs Foundation Trust, Sheffield, UK
| | - Paul M. Kaye
- York Biomedical Research Institute, Hull York Medical School, University of York, York, UK
| | - Alison M Layton
- York Biomedical Research Institute, Hull York Medical School, University of York, York, UK
| | - Charles J Lacey
- York Biomedical Research Institute, Hull York Medical School, University of York, York, UK
| |
Collapse
|
10
|
Parkash V, Ashwin H, Sadlova J, Vojtkova B, Jones G, Martin N, Greensted E, Allgar V, Kamhawi S, Valenzuela JG, Layton AM, Jaffe CL, Volf P, Kaye PM, Lacey CJN. A clinical study to optimise a sand fly biting protocol for use in a controlled human infection model of cutaneous leishmaniasis (the FLYBITE study). Wellcome Open Res 2021; 6:168. [PMID: 34693027 PMCID: PMC8506224 DOI: 10.12688/wellcomeopenres.16870.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2021] [Indexed: 02/02/2023] Open
Abstract
Background: Leishmaniasis is a globally important yet neglected parasitic disease transmitted by phlebotomine sand flies. With new candidate vaccines in or near the clinic, a controlled human challenge model (CHIM) using natural sand fly challenge would provide a method for early evaluation of prophylactic efficacy. Methods : We evaluated the biting frequency and adverse effects resulting from exposure of human volunteers to bites of either Phlebotomus papatasi or P. duboscqi, two natural vectors of Leishmania major. 12 healthy participants were recruited (mean age 40.2 ± 11.8 years) with no history of significant travel to regions where L. major-transmitting sand flies are prevalent. Participants were assigned to either vector by 1:1 allocation and exposed to five female sand flies for 30 minutes in a custom biting chamber. Bite frequency was recorded to confirm a bloodmeal was taken. Participant responses and safety outcomes were monitored using a visual analogue scale (VAS), clinical examination, and blood biochemistry. Focus groups were subsequently conducted to explore participant acceptability. Results: All participants had at least one successful sand fly bite with none reporting any serious adverse events, with median VAS scores of 0-1/10 out to day 21 post-sand fly bite. Corresponding assessment of sand flies confirmed that for each participant at least 1/5 sand flies had successfully taken a bloodmeal (overall mean 3.67±1.03 bites per participant). There was no significant difference between P. papatasi and P. duboscqi in the number of bites resulting from 5 sand flies applied to human participants (3.3±0.81 vs 3.00±1.27 bites per participant; p=0.56) . In the two focus groups (n=5 per group), themes relating to positive participant-reported experiences of being bitten and the overall study, were identified. Conclusions: These results validate a protocol for achieving successful sand fly bites in humans that is safe, well-tolerated and acceptable for participants. Clinicaltrials.gov registration: NCT03999970 (27/06/2019).
Collapse
Affiliation(s)
- Vivak Parkash
- York Biomedical Research Institute, Hull York Medical School, University of York, York, N.Yorks, YO10 5DD, UK
- Department of Infection and Tropical Medicine, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Helen Ashwin
- York Biomedical Research Institute, Hull York Medical School, University of York, York, N.Yorks, YO10 5DD, UK
| | - Jovana Sadlova
- Department of Parasitology, Charles University, Prague, Czech Republic
| | - Barbora Vojtkova
- Department of Parasitology, Charles University, Prague, Czech Republic
| | - Georgina Jones
- School of Social Sciences, Leeds Beckett University, Leeds, UK
| | - Nina Martin
- School of Social Sciences, Leeds Beckett University, Leeds, UK
| | - Elizabeth Greensted
- York Biomedical Research Institute, Hull York Medical School, University of York, York, N.Yorks, YO10 5DD, UK
| | - Victoria Allgar
- Peninsula Medical School, University of Plymouth, Plymouth, UK
| | - Shaden Kamhawi
- Laboratory of Malaria and Vector Research, National Institutes of Health, Rockville, MD, USA
| | - Jesus G. Valenzuela
- Laboratory of Malaria and Vector Research, National Institutes of Health, Rockville, MD, USA
| | - Alison M. Layton
- York Biomedical Research Institute, Hull York Medical School, University of York, York, N.Yorks, YO10 5DD, UK
| | - Charles L. Jaffe
- Department of Microbiology and Molecular Genetics, The Hebrew University – Hadassah Medical School, Jerusalem, Israel
| | - Petr Volf
- Department of Parasitology, Charles University, Prague, Czech Republic
| | - Paul M. Kaye
- York Biomedical Research Institute, Hull York Medical School, University of York, York, N.Yorks, YO10 5DD, UK
| | - Charles J. N. Lacey
- York Biomedical Research Institute, Hull York Medical School, University of York, York, N.Yorks, YO10 5DD, UK
| |
Collapse
|
11
|
Lakhal-Naouar I, Mukbel R, DeFraites RF, Mody RM, Massoud LN, Shaw D, Co EM, Sherwood JE, Kamhawi S, Aronson NE. The human immune response to saliva of Phlebotomus alexandri, the vector of visceral leishmaniasis in Iraq, and its relationship to sand fly exposure and infection. PLoS Negl Trop Dis 2021; 15:e0009378. [PMID: 34081700 PMCID: PMC8174707 DOI: 10.1371/journal.pntd.0009378] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 04/10/2021] [Indexed: 11/29/2022] Open
Abstract
Background Sand fly saliva exposure plays an important role in immunity against leishmaniasis where it has mostly been associated with protection. Phlebotomus (Ph.) alexandri transmits Leishmania (L.) infantum, the causative agent of visceral leishmaniasis (VL), in Iraq. Our group recently demonstrated that 20% of Operation Iraqi Freedom (OIF) deployers had asymptomatic VL (AVL) indicative of prior infection by the parasite L. infantum. Little is known about Ph. alexandri saliva, and the human immune response to it has never been investigated. Here, we characterize the humoral and cellular immune response to vector saliva in OIF deployers naturally exposed to bites of Ph. alexandri and characterize their immunological profiles in association to AVL. Methodology/Principal findings The humoral response to Ph. alexandri salivary gland homogenate (SGH) showed that 64% of 200 OIF deployers developed an antibody response. To assess the cellular immune response to saliva, we selected a subcohort of subjects based on their post-travel (median 4 months; range 1–22 months) antibody response (SGH Antibody [Ab] positive or negative) as well as their AVL status; ten never-traveled controls were also included. Banked peripheral blood mononuclear cells (PBMC), collected ~10 years after end of deployment, were stimulated with SGH for 96 hours. The levels of IFN- γ, IL-6, IL-10, IL-13 and IL-17 were determined by ELISA. Our findings indicate that OIF deployers mounted a cellular response to SGH where the anti-SGH+ asymptomatic subjects developed the highest cytokine levels. Further, stimulation with SGH produced a mixture of pro-inflammatory and anti-inflammatory cytokines. Contrary to our hypothesis, we observed no correlation between the cellular immune response to Ph. alexandri SGH and prevention from asymptomatic infection with L. infantum. Conclusions/Significance As we found, although all infected deployers demonstrated persistent disease control years after deployment, this did not correlate with anti-saliva systemic cellular response. More exposure to this vector may facilitate transmission of the L. infantum parasite. Since exposure to saliva of Ph. alexandri may alter the human immune response to bites of this vector, this parameter should be taken into consideration when considering the VL risk. This is the first report of human immune responses to Phlebotomus alexandri. Phlebotomus alexandri is a sand fly found in Southwest Asia and is the vector for transmission of the parasite Leishmania infantum, agent of visceral leishmaniasis. During the bite of this sand fly, a small amount of saliva is injected into the skin. In this study, we report the human immune response to Phlebotomus alexandri saliva. We found that 64% of people who traveled to endemic Iraq developed antibodies directed toward this sand fly’s saliva. This suggests that saliva is very immunogenic and that anti-saliva immune responses could be a good indicator of vector exposure. Additionally, we studied the cellular immune responses in saliva-stimulated white blood cells and found a Th2 biased cytokine profile.
Collapse
Affiliation(s)
- Ines Lakhal-Naouar
- Infectious Diseases Division, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
- * E-mail:
| | - Rami Mukbel
- Department of Basic Veterinary Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Robert F. DeFraites
- Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Rupal M. Mody
- Infectious Diseases Service, William Beaumont Army Medical Center, El Paso, Texas, United States of America
| | - Lina N. Massoud
- Department of Basic Veterinary Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Dutchabong Shaw
- Infectious Diseases Division, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Edgie M. Co
- Infectious Diseases Service, Walter Reed National Military Medical Center, Bethesda Maryland, United States of America
| | - Jeffrey E. Sherwood
- Infectious Diseases Service, William Beaumont Army Medical Center, El Paso, Texas, United States of America
| | - Shaden Kamhawi
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Naomi E. Aronson
- Infectious Diseases Division, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| |
Collapse
|
12
|
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
|
13
|
Ratnapriya S, Keerti, Yadav NK, Dube A, Sahasrabuddhe AA. A Chimera of Th1 Stimulatory Proteins of Leishmania donovani Offers Moderate Immunotherapeutic Efficacy with a Th1-Inclined Immune Response against Visceral Leishmaniasis. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8845826. [PMID: 34095312 PMCID: PMC8164546 DOI: 10.1155/2021/8845826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 01/27/2021] [Accepted: 02/15/2021] [Indexed: 11/17/2022]
Abstract
Immunotherapy, a treatment based on host immune system activation, has been shown to provide a substitute for marginally effective conventional chemotherapy in controlling visceral leishmaniasis (VL), the deadliest form of leishmaniasis. As the majority of endemic inhabitants exhibit either subclinical or asymptomatic infection which often develops into the active disease state, therapeutic intervention seems to be an important avenue for combating infections by stimulating the natural defense system of infected individuals. With this perspective, the present study focuses on two immunodominant Leishmania (L.) donovani antigens (triosephosphate isomerase and enolase) previously proved to be potent prophylactic VL vaccine candidates, for generating a recombinant chimeric antigen. This is based on the premise that in a heterogeneous population, a multivalent antigen vaccine would be required for an effective response against leishmaniasis (a complex parasitic disease). The resulting molecule rLdT-E chimeric protein was evaluated for its immunogenicity and immunotherapeutic efficacy. A Th1 stimulating adjuvant BCG was employed with the protein which showed a remarkable 70% inhibition of splenic parasitic multiplication positively correlated with boosted Th1 dominant immune response against lethal L. donovani challenge in hamsters as evidenced by high IFN-γ and TNF-α and low IL-10. In addition, immunological analysis of antibody subclass presented IgG2-based humoral response besides considerable delayed-type hypersensitivity and lymphocyte proliferative responses in rLdT-E/BCG-treated animals. Our observations indicate the potential of the chimera towards its candidature for an effective vaccine against Leishmania donovani infection.
Collapse
Affiliation(s)
- Sneha Ratnapriya
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Keerti
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Narendra Kumar Yadav
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Anuradha Dube
- Molecular Parasitology and Immunology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Amogh Anant Sahasrabuddhe
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| |
Collapse
|
14
|
Bourdeau P, Rowton E, Petersen C. Impact of different Leishmania reservoirs on sand fly transmission: Perspectives from xenodiagnosis and other one health observations. Vet Parasitol 2020; 287:109237. [PMID: 33160145 PMCID: PMC8035349 DOI: 10.1016/j.vetpar.2020.109237] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/01/2020] [Accepted: 09/02/2020] [Indexed: 12/14/2022]
Abstract
Leishmania has biologically adapted to specific phlebotomine sand flies through long co-evolution. The ability of Leishmania spp. to bind to sand fly midgut allows each Leishmania species to propagate and differentiate into infectious promastigotes and be transmitted. Sand fly feeding upon a mammalian host is the first step towards being infected and a host of Leishmania. Once deposited into the skin, host susceptibility to infection vs. ability to mount a sterilizing immune response predicts which hosts could be reservoirs of different Leishmania spp. Materials, in addition to parasites, are expelled during sand fly during feeding, including salivary antigens and other factors that promote local inflammatory responses. These factors aid visceralization of infection increasing the likelihood that systemic infection is established. Any environmental factor that increases sand fly biting of a particular host increases that host's role in Leishmania transmission. First descriptions of reservoir species were based on association with local human disease and ability to observe infected leukocytes on cytology. This approach was one pathogen for one reservoir host. Advances in sensitive molecular tools greatly increased the breadth of mammals found to host Leishmania infection. Visceralizing species of Leishmania, particularly L. infantum, are now known to have multiple mammalian hosts. L. donovani, long been described as an anthroponotic parasite, was recently identified through molecular and serologic surveys to have additional mammalian hosts. The epidemiological role of these animals as a source of parasites to additional hosts via vector transmission is not known. Current evidence suggests that dogs and other domestic animals either control infection or do not have sufficient skin parasitemia to be a source of L. donovani to P. argentipes. Further xenodiagnosis and characterization of skin parasitemia in these different hosts is required to more broadly understand which Leishmania spp. hosts can be a source of parasites to sand flies and which ones are dead-end hosts.
Collapse
Affiliation(s)
- Patrick Bourdeau
- Laboratoire de Dermatologie, Parasitologie et Mycologie, ONIRIS, Ecole Nationale Veterinaire, Agroalimentaire et de l'Alimentation Nantes-Atlantique, Nantes, France; Immunology Program, Department of Internal Medicine and Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Edgar Rowton
- Walter Reed Army Institute of Research, Silver Spring, MD, USA; Immunology Program, Department of Internal Medicine and Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Christine Petersen
- Walter Reed Army Institute of Research, Silver Spring, MD, USA; Department of Epidemiology, College of Public Health, USA; Center for Emerging Infectious Diseases, Coralville, IA, 52241, USA; Immunology Program, Department of Internal Medicine and Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
15
|
Manning JE, Oliveira F, Coutinho-Abreu IV, Herbert S, Meneses C, Kamhawi S, Baus HA, Han A, Czajkowski L, Rosas LA, Cervantes-Medina A, Athota R, Reed S, Mateja A, Hunsberger S, James E, Pleguezuelos O, Stoloff G, Valenzuela JG, Memoli MJ. Safety and immunogenicity of a mosquito saliva peptide-based vaccine: a randomised, placebo-controlled, double-blind, phase 1 trial. Lancet 2020; 395:1998-2007. [PMID: 32534628 PMCID: PMC9151349 DOI: 10.1016/s0140-6736(20)31048-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/09/2020] [Accepted: 04/27/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND In animal models, immunity to mosquito salivary proteins protects animals against mosquito-borne disease. These findings provide a rationale to vaccinate against mosquito saliva instead of the pathogen itself. To our knowledge, no vector salivary protein-based vaccine has been tested for safety and immunogenicity in humans. We aimed to assess the safety and immunogenicity of Anopheles gambiae saliva vaccine (AGS-v), a peptide-based vaccine derived from four A gambiae salivary proteins, in humans. METHODS In this randomised, placebo-controlled, double-blind, phase 1 trial, participants were enrolled at the National Institutes of Health Clinical Center in Bethesda, MD, USA. Participants were eligible if they were healthy adults, aged 18-50 years with no history of severe allergic reactions to mosquito bites. Participants were randomly assigned (1:1:1), using block randomisation and a computer-generated randomisation sequence, to treatment with either 200 nmol of AGS-v vaccine alone, 200 nmol of AGS-v with adjuvant (Montanide ISA 51), or sterile water as placebo. Participants and clinicians were masked to treatment assignment. Participants were given a subcutaneous injection of their allocated treatment at day 0 and day 21, followed by exposure to feeding by an uninfected Aedes aegypti mosquito at day 42 to assess subsequent risk to mosquito bites in a controlled setting. The primary endpoints were safety and immunogenicity at day 42 after the first immunisation. Participants who were given at least one dose of assigned treatment were assessed for the primary endpoints and analysis was by intention to treat. The trial was registered with ClinicalTrials.gov, NCT03055000, and is closed for accrual. FINDINGS Between Feb 15 and Sept 10, 2017, we enrolled and randomly assigned 49 healthy adult participants to the adjuvanted vaccine (n=17), vaccine alone (n=16), or placebo group (n=16). Five participants did not complete the two-injection regimen with mosquito feeding at day 42, but were included in the safety analyses. No systemic safety concerns were identified; however, one participant in the adjuvanted vaccine group developed a grade 3 erythematous rash at the injection site. Pain, swelling, erythema, and itching were the most commonly reported local symptoms and were significantly increased in the adjuvanted vaccine group compared with both other treatment groups (nine [53%] of 17 participants in the adjuvanted vaccine group, two [13%] of 16 in the vaccine only group, and one [6%] of 16 in the placebo group; p=0·004). By day 42, participants who were given the adjuvanted vaccine had a significant increase in vaccine-specific total IgG antibodies compared with at baseline than did participants who were give vaccine only (absolute difference of log10-fold change of 0·64 [95% CI 0·39 to 0·89]; p=0·0002) and who were given placebo (0·62 [0·34 to 0·91]; p=0·0001). We saw a significant increase in IFN-γ production by peripheral blood mononuclear cells at day 42 in the adjuvanted vaccine group compared with in the placebo group (absolute difference of log10 ratio of vaccine peptide-stimulated vs negative control 0·17 [95% CI 0·061 to 0·27]; p=0·009) but we saw no difference between the IFN-γ production in the vaccine only group compared with the placebo group (0·022 [-0·072 to 0·116]; p=0·63). INTERPRETATION AGS-v was well tolerated, and, when adjuvanted, immunogenic. These findings suggest that vector-targeted vaccine administration in humans is safe and could be a viable option for the increasing burden of vector-borne disease. FUNDING Office of the Director and the Division of Intramural Research at the National Institute of Allergy and Infectious Diseases, and National Institutes of Health.
Collapse
Affiliation(s)
- Jessica E Manning
- Laboratory of Malaria and Vector Research, National Institutes of Health, Bethesda, MD, USA.
| | - Fabiano Oliveira
- Laboratory of Malaria and Vector Research, National Institutes of Health, Bethesda, MD, USA
| | | | - Samantha Herbert
- Laboratory of Malaria and Vector Research, National Institutes of Health, Bethesda, MD, USA
| | - Claudio Meneses
- Laboratory of Malaria and Vector Research, National Institutes of Health, Bethesda, MD, USA
| | - Shaden Kamhawi
- Laboratory of Malaria and Vector Research, National Institutes of Health, Bethesda, MD, USA
| | - Holly Ann Baus
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alison Han
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Lindsay Czajkowski
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Luz Angela Rosas
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Adriana Cervantes-Medina
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Rani Athota
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Susan Reed
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Allyson Mateja
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, sponsored by the National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Sally Hunsberger
- Biostatistics Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | - Jesus G Valenzuela
- Laboratory of Malaria and Vector Research, National Institutes of Health, Bethesda, MD, USA
| | - Matthew J Memoli
- LID Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
16
|
Oliveira F, Giorgobiani E, Guimarães-Costa AB, Abdeladhim M, Oristian J, Tskhvaradze L, Tsertsvadze N, Zakalashvili M, Valenzuela JG, Kamhawi S. Immunity to vector saliva is compromised by short sand fly seasons in endemic regions with temperate climates. Sci Rep 2020; 10:7990. [PMID: 32409684 PMCID: PMC7224377 DOI: 10.1038/s41598-020-64820-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 04/22/2020] [Indexed: 01/26/2023] Open
Abstract
Individuals exposed to sand fly bites develop humoral and cellular immune responses to sand fly salivary proteins. Moreover, cellular immunity to saliva or distinct salivary proteins protects against leishmaniasis in various animal models. In Tbilisi, Georgia, an endemic area for visceral leishmaniasis (VL), sand flies are abundant for a short period of ≤3 months. Here, we demonstrate that humans and dogs residing in Tbilisi have little immunological memory to saliva of P. kandelakii, the principal vector of VL. Only 30% of humans and 50% of dogs displayed a weak antibody response to saliva after the end of the sand fly season. Likewise, their peripheral blood mononuclear cells mounted a negligible cellular immune response after stimulation with saliva. RNA seq analysis of wild-caught P. kandelakii salivary glands established the presence of a typical salivary repertoire that included proteins commonly found in other sand fly species such as the yellow, SP15 and apyrase protein families. This indicates that the absence of immunity to P. kandelakii saliva in humans and dogs from Tbilisi is probably caused by insufficient exposure to sand fly bites. This absence of immunity to vector saliva will influence the dynamics of VL transmission in Tbilisi and other endemic areas with brief sand fly seasons.
Collapse
Affiliation(s)
- Fabiano Oliveira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, 20852, USA
| | - Ekaterina Giorgobiani
- R. G. Lugar Center for Public Health Research, National Center for Disease Control and Public Health (NCDC), Kakheti Highway 99, 0198, Tbilisi, Georgia
| | - Anderson B Guimarães-Costa
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, 20852, USA
| | - Maha Abdeladhim
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, 20852, USA
| | - James Oristian
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, 20852, USA
| | - Lamzira Tskhvaradze
- R. G. Lugar Center for Public Health Research, National Center for Disease Control and Public Health (NCDC), Kakheti Highway 99, 0198, Tbilisi, Georgia
| | - Nikoloz Tsertsvadze
- R. G. Lugar Center for Public Health Research, National Center for Disease Control and Public Health (NCDC), Kakheti Highway 99, 0198, Tbilisi, Georgia
| | - Mariam Zakalashvili
- R. G. Lugar Center for Public Health Research, National Center for Disease Control and Public Health (NCDC), Kakheti Highway 99, 0198, Tbilisi, Georgia
| | - Jesus G Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, 20852, USA.
| | - Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, 20852, USA.
| |
Collapse
|
17
|
Manning JE, Morens DM, Kamhawi S, Valenzuela JG, Memoli M. Mosquito Saliva: The Hope for a Universal Arbovirus Vaccine? J Infect Dis 2019; 218:7-15. [PMID: 29617849 DOI: 10.1093/infdis/jiy179] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 03/28/2018] [Indexed: 02/06/2023] Open
Abstract
Arthropod-borne viruses (arboviruses) are taxonomically diverse causes of significant morbidity and mortality. In recent decades, important mosquito-borne viruses such as West Nile, chikungunya, dengue, and Zika have re-emerged and spread widely, in some cases pandemically, to cause serious public health emergencies. There are no licensed vaccines against most of these viruses, and vaccine development and use has been complicated by the number of different viruses to protect against, by subtype and strain variation, and by the inability to predict when and where outbreaks will occur. A new approach to preventing arboviral diseases is suggested by the observation that arthropod saliva facilitates transmission of pathogens, including leishmania parasites, Borrelia burgdorferi, and some arboviruses. Viruses carried within mosquito saliva may more easily initiate host infection by taking advantage of the host's innate and adaptive immune responses to saliva. This provides a rationale for creating vaccines against mosquito salivary proteins, rather than against only the virus proteins contained within the saliva. As proof of principle, immunization with sand fly salivary antigens to prevent leishmania infection has shown promising results in animal models. A similar approach using salivary proteins of important vector mosquitoes, such as Aedes aegypti, might protect against multiple mosquito-borne viral infections.
Collapse
Affiliation(s)
- Jessica E Manning
- Laboratory of Malaria and Vector Research, National Institutes of Health, Bethesda, Maryland.,Laboratory of Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - David M Morens
- Office of the Director, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Shaden Kamhawi
- Laboratory of Malaria and Vector Research, National Institutes of Health, Bethesda, Maryland
| | - Jesus G Valenzuela
- Laboratory of Malaria and Vector Research, National Institutes of Health, Bethesda, Maryland
| | - Matthew Memoli
- Laboratory of Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
18
|
Al-Salem WS, Solórzano C, Weedall GD, Dyer NA, Kelly-Hope L, Casas-Sánchez A, Alraey Y, Alyamani EJ, Halliday A, Balghonaim SM, Alsohibany KS, Alzeyadi Z, Alzahrani MH, Al-Shahrani AM, Assiri AM, Memish Z, Acosta-Serrano Á. Old World cutaneous leishmaniasis treatment response varies depending on parasite species, geographical location and development of secondary infection. Parasit Vectors 2019; 12:195. [PMID: 31046820 PMCID: PMC6498568 DOI: 10.1186/s13071-019-3453-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/20/2019] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND In the Kingdom of Saudi Arabia (KSA), Leishmania major and L. tropica are the main causative agents of Old World cutaneous leishmaniasis (CL). The national CL treatment regimen consists of topical 1% clotrimazole/2% fusidic acid cream followed by 1-2 courses of intralesional sodium stibogluconate (SSG); however, treatment efficacy is highly variable and the reasons for this are not well understood. In this study, we present a complete epidemiological map of CL and determined the efficacy of the standard CL treatment regime in several endemic regions of KSA. RESULTS Overall, three quarters of patients in all CL-endemic areas studied responded satisfactorily to the current treatment regime, with the remaining requiring only an extra course of SSG. The majority of unresponsive cases were infected with L. tropica. Furthermore, the development of secondary infections (SI) around or within the CL lesion significantly favoured the treatment response of L. major patients but had no effect on L. tropica cases. CONCLUSIONS The response of CL patients to a national treatment protocol appears to depend on several factors, including Leishmania parasite species, geographical location and occurrences of SI. Our findings suggest there is a need to implement alternative CL treatment protocols based on these parameters.
Collapse
Affiliation(s)
- Waleed S. Al-Salem
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
- Present Address: National Centre for Tropical Diseases, Saudi Ministry of Health, Riyadh, Kingdom of Saudi Arabia
| | - Carla Solórzano
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Gareth D. Weedall
- Faculty of Sciences, Liverpool John Moores University, Liverpool, UK
| | - Naomi A. Dyer
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Louise Kelly-Hope
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Aitor Casas-Sánchez
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Yasser Alraey
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Essam J. Alyamani
- National Center for Biotechnology, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Alice Halliday
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | | | | | - Zeyad Alzeyadi
- Antimicrobial Research Centre, University of Leeds, Leeds, UK
| | | | | | | | - Ziad Memish
- Saudi Ministry of Health, Riyadh, Kingdom of Saudi Arabia
| | - Álvaro Acosta-Serrano
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
- Department of Vector Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| |
Collapse
|
19
|
Manning JE, Cantaert T. Time to Micromanage the Pathogen-Host-Vector Interface: Considerations for Vaccine Development. Vaccines (Basel) 2019; 7:E10. [PMID: 30669682 PMCID: PMC6466432 DOI: 10.3390/vaccines7010010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/10/2019] [Accepted: 01/16/2019] [Indexed: 12/18/2022] Open
Abstract
The current increase in vector-borne disease worldwide necessitates novel approaches to vaccine development targeted to pathogens delivered by blood-feeding arthropod vectors into the host skin. A concept that is gaining traction in recent years is the contribution of the vector or vector-derived components, like salivary proteins, to host-pathogen interactions. Indeed, the triad of vector-host-pathogen interactions in the skin microenvironment can influence host innate and adaptive responses alike, providing an advantage to the pathogen to establish infection. A better understanding of this "bite site" microenvironment, along with how host and vector local microbiomes immunomodulate responses to pathogens, is required for future vaccines for vector-borne diseases. Microneedle administration of such vaccines may more closely mimic vector deposition of pathogen and saliva into the skin with the added benefit of near painless vaccine delivery. Focusing on the 'micro'⁻from microenvironments to microbiomes to microneedles⁻may yield an improved generation of vector-borne disease vaccines in today's increasingly complex world.
Collapse
Affiliation(s)
- Jessica E Manning
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Phnom Penh 12201, Cambodia.
| | - Tineke Cantaert
- Immunology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh 12201, Cambodia.
| |
Collapse
|
20
|
Manning JE, Oliveira F, Parker DM, Amaratunga C, Kong D, Man S, Sreng S, Lay S, Nang K, Kimsan S, Sokha L, Kamhawi S, Fay MP, Suon S, Ruhl P, Ackerman H, Huy R, Wellems TE, Valenzuela JG, Leang R. The PAGODAS protocol: pediatric assessment group of dengue and Aedes saliva protocol to investigate vector-borne determinants of Aedes-transmitted arboviral infections in Cambodia. Parasit Vectors 2018; 11:664. [PMID: 30572920 PMCID: PMC6300895 DOI: 10.1186/s13071-018-3224-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 11/20/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Mosquito-borne arboviruses, like dengue virus, continue to cause significant global morbidity and mortality, particularly in Southeast Asia. When the infectious mosquitoes probe into human skin for a blood meal, they deposit saliva containing a myriad of pharmacologically active compounds, some of which alter the immune response and influence host receptivity to infection, and consequently, the establishment of the virus. Previous reports have highlighted the complexity of mosquito vector-derived factors and immunity in the success of infection. Cumulative evidence from animal models and limited data from humans have identified various vector-derived components, including salivary components, that are co-delivered with the pathogen and play an important role in the dissemination of infection. Much about the roles and effects of these vector-derived factors remain to be discovered. METHODS/DESIGN We describe a longitudinal, pagoda (community)-based pediatric cohort study to evaluate the burden of dengue virus infection and document the immune responses to salivary proteins of Aedes aegypti, the mosquito vector of dengue, Zika, and chikungunya viruses. The study includes community-based seroprevalence assessments in the peri-urban town of Chbar Mon in Kampong Speu Province, Cambodia. The study aims to recruit 771 children between the ages of 2 and 9 years for a three year period of longitudinal follow-up, including twice per year (rainy and dry season) serosurveillance for dengue seroconversion and Ae. aegypti salivary gland homogenate antibody intensity determinations by ELISA assays. Diagnostic tests for acute dengue, Zika and chikungunya viral infections will be performed by RT-PCR. DISCUSSION This study will serve as a foundation for further understanding of mosquito saliva immunity and its impact on Aedes-transmitted arboviral diseases endemic to Cambodia. TRIAL REGISTRATION NCT03534245 registered on 23 May 2018.
Collapse
Affiliation(s)
- Jessica E. Manning
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland USA
| | - Fabiano Oliveira
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland USA
| | - Daniel M. Parker
- Department of Population Health and Disease Prevention, University of California, Irvine, California, USA
| | - Chanaki Amaratunga
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland USA
| | - Dara Kong
- National Center of Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Somnang Man
- National Center of Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Sokunthea Sreng
- National Center of Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Sreyngim Lay
- National Center of Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Kimsour Nang
- National Center of Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Soun Kimsan
- National Center of Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Ly Sokha
- National Center of Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Shaden Kamhawi
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland USA
| | - Michael P. Fay
- Biostatistics Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland USA
| | - Seila Suon
- National Center of Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Parker Ruhl
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland USA
| | - Hans Ackerman
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland USA
| | - Rekol Huy
- National Center of Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| | - Thomas E. Wellems
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland USA
| | - Jesus G. Valenzuela
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland USA
| | - Rithea Leang
- National Center of Parasitology, Entomology, and Malaria Control, Phnom Penh, Cambodia
| |
Collapse
|
21
|
Tomiotto-Pellissier F, Bortoleti BTDS, Assolini JP, Gonçalves MD, Carloto ACM, Miranda-Sapla MM, Conchon-Costa I, Bordignon J, Pavanelli WR. Macrophage Polarization in Leishmaniasis: Broadening Horizons. Front Immunol 2018; 9:2529. [PMID: 30429856 PMCID: PMC6220043 DOI: 10.3389/fimmu.2018.02529] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 10/15/2018] [Indexed: 01/14/2023] Open
Abstract
Leishmaniasis is a vector-borne neglected tropical disease that affects more than 700,000 people annually. Leishmania parasites cause the disease, and different species trigger a distinct immune response and clinical manifestations. Macrophages are the final host cells for the proliferation of Leishmania parasites, and these cells are the key to a controlled or exacerbated response that culminates in clinical manifestations. M1 and M2 are the two main macrophage phenotypes. M1 is a pro-inflammatory subtype with microbicidal properties, and M2, or alternatively activated, is an anti-inflammatory/regulatory subtype that is related to inflammation resolution and tissue repair. The present review elucidates the roles of M1 and M2 polarization in leishmaniasis and highlights the role of the salivary components of the vector and the action of the parasite in the macrophage plasticity.
Collapse
Affiliation(s)
- Fernanda Tomiotto-Pellissier
- Biosciences and Biotechnology Postgraduate Program, Carlos Chagas Institute (ICC), Fiocruz, Curitiba, Brazil.,Laboratory of Immunoparasitology, Department of Pathological Sciences, State University of Londrina, Londrina, Brazil
| | - Bruna Taciane da Silva Bortoleti
- Biosciences and Biotechnology Postgraduate Program, Carlos Chagas Institute (ICC), Fiocruz, Curitiba, Brazil.,Laboratory of Immunoparasitology, Department of Pathological Sciences, State University of Londrina, Londrina, Brazil
| | - João Paulo Assolini
- Laboratory of Immunoparasitology, Department of Pathological Sciences, State University of Londrina, Londrina, Brazil
| | - Manoela Daiele Gonçalves
- Laboratory of Biotransformation and Phytochemistry, Department of Chemistry, State University of Londrina, Universitary Hospital, Londrina, Brazil
| | | | | | - Ivete Conchon-Costa
- Laboratory of Immunoparasitology, Department of Pathological Sciences, State University of Londrina, Londrina, Brazil
| | - Juliano Bordignon
- Biosciences and Biotechnology Postgraduate Program, Carlos Chagas Institute (ICC), Fiocruz, Curitiba, Brazil.,Laboratory of Molecular Virology, Carlos Chagas Institute (ICC), Fiocruz, Curitiba, Brazil
| | - Wander Rogério Pavanelli
- Biosciences and Biotechnology Postgraduate Program, Carlos Chagas Institute (ICC), Fiocruz, Curitiba, Brazil.,Laboratory of Immunoparasitology, Department of Pathological Sciences, State University of Londrina, Londrina, Brazil
| |
Collapse
|
22
|
Bahrami F, Harandi AM, Rafati S. Biomarkers of Cutaneous Leishmaniasis. Front Cell Infect Microbiol 2018; 8:222. [PMID: 29998089 PMCID: PMC6029629 DOI: 10.3389/fcimb.2018.00222] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/11/2018] [Indexed: 12/12/2022] Open
Abstract
Cutaneous leishmaniasis (CL) is an immune-mediated skin pathology caused mainly by Leishmania (L.) major, Leishmania tropica, Leishmania braziliensis, L. mexicana, and L. amazonensis. The burden of CL in terms of morbidity and social stigmas are concentrated on certain developing countries in Asia, Africa, and South America. People with asymptomatic CL represent a large proportion of the infected individuals in the endemic areas who exhibit no lesion and can control the infection by as yet not fully understood mechanisms. Currently, there is no approved prophylactic control measure for CL. Discovery of biomarkers of CL infection and immunity can inform the development of more precise diagnostics tools as well as curative or preventive strategies to control CL. Herein, we provide a brief overview of the state-of-the-art for the biomarkers of CL with a special emphasis on the asymptomatic CL biomarkers. Among the identified CL biomarkers so far, direct biomarkers which indicate the actual presence of the infection as well as indirect biomarkers which reflect the host's reaction to the infection, such as alterations in delayed type hypersensitivity, T-cell subpopulations and cytokines, adenosine deaminase, and antibodies against the sand fly saliva proteins are discussed in detail. The future avenues such as the use of systems analysis to identify and characterize novel CL biomarkers are also discussed.
Collapse
Affiliation(s)
- Fariborz Bahrami
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Ali M Harandi
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Sima Rafati
- Department of Immunotherapy and Leishmania Vaccine Research, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
23
|
Teixeira CR, Santos CDS, Prates DB, Dos Santos RT, Araújo-Santos T, de Souza-Neto SM, Borges VM, Barral-Netto M, Brodskyn CI. Lutzomyia longipalpis Saliva Drives Interleukin-17-Induced Neutrophil Recruitment Favoring Leishmania infantum Infection. Front Microbiol 2018; 9:881. [PMID: 29867796 PMCID: PMC5953329 DOI: 10.3389/fmicb.2018.00881] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 04/17/2018] [Indexed: 11/22/2022] Open
Abstract
During bloodfeeding, the presence of sand fly saliva in the hemorrhagic pool where Leishmania is also inoculated modulates the development of host immune mechanisms creating a favorable environment for disease progression. To date, information obtained through experimental models suggests that sand fly saliva induces cellular recruitment and modulates production of eicosanoids. However, the effect of sand fly saliva in the different steps of the inflammatory response triggered by Leishmania remains undefined. Here we further investigate if interaction of Lutzomyia longipalpis salivary gland sonicate (SGS) with different host cells present during the initial inflammatory events regulate Leishmania infantum infectivity. Initially, we observed that incubation of human peripheral blood mononuclear cells (PBMC) with Lu. longipalpis SGS in the presence of L. infantum significantly increased IL-10 but did not alter expression of IFN-γ and TNF-α by CD4+ T cells induced by the parasite alone. Interestingly, incubation of PBMC with Lu. longipalpis SGS alone or in the presence of L. infantum resulted in increased IL-17 production. The presence of IL-17 is related to neutrophil recruitment and plays an important role at the site of infection. Here, we also observed increased migration of neutrophil using an in vitro chemotactic assay following incubation with supernatants from PBMC stimulated with L. infantum and Lu. longipalpis SGS. Neutrophil migration was abrogated following neutralization of IL-17 with specific antibodies. Moreover, culture of human neutrophils with L. infantum in the presence of Lu. longipalpis SGS promoted neutrophil apoptosis resulting in increased parasite viability. Neutrophils operate as the first line of defense in the early stages of infection and later interact with different cells, such as macrophages. The crosstalk between neutrophils and macrophages is critical to determine the type of specific immune response that will develop. Here, we observed that co-culture of human macrophages with autologous neutrophils previously infected in the presence of Lu. longipalpis SGS resulted in a higher infection rate, accompanied by increased production of TGF-β and PGE2. Our results provide new insight into the contribution of Lu. longipalpis SGS to L. infantum-induced regulation of important inflammatory events, creating a favorable environment for parasite survival inside different host cells.
Collapse
Affiliation(s)
| | | | - Deboraci B Prates
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Instituto de Ciências da Saúde da Universidade Federal da Bahia, Departamentos de Biomorfologia e Biointeração, Salvador, Brazil
| | | | - Théo Araújo-Santos
- Centro de Ciências Biológicas e Saúde, Universidade Federal do Oeste da Bahia, Barreiras, Brazil
| | | | - Valéria M Borges
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Manoel Barral-Netto
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Faculdade de Medicina da Universidade Federal da Bahia, Departamento de Patologia e Medicina Legal, Salvador, Brazil.,Instituto de Investigação em Imunologia, iii-INCT, São Paulo, Brazil
| | - Cláudia I Brodskyn
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Instituto de Ciências da Saúde da Universidade Federal da Bahia, Departamentos de Biomorfologia e Biointeração, Salvador, Brazil.,Instituto de Investigação em Imunologia, iii-INCT, São Paulo, Brazil
| |
Collapse
|
24
|
Cunha JM, Abbehusen M, Suarez M, Valenzuela J, Teixeira CR, Brodskyn CI. Immunization with LJM11 salivary protein protects against infection with Leishmania braziliensis in the presence of Lutzomyia longipalpis saliva. Acta Trop 2018; 177:164-170. [PMID: 29037520 DOI: 10.1016/j.actatropica.2017.10.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 08/02/2017] [Accepted: 10/12/2017] [Indexed: 12/31/2022]
Abstract
Leishmania is transmitted in the presence of sand fly saliva. Protective immunity generated by saliva has encouraged identification of a vector salivary-based vaccine. Previous studies have shown that immunization with LJM11, a salivary protein from Lutzomyia longipalpis, is able to induce a Th1 immune response and protect mice against bites of Leishmania major-infected Lutzomyia longipalpis. Here, we further investigate if immunization with LJM11 recombinant protein is able to confer cross-protection against infection with Leishmania braziliensis associated with salivary gland sonicate (SGS) from Lutzomyia intermedia or Lu. longipalpis. Mice immunized with LJM11 protein exhibited an increased production of anti-LJM11 IgG, IgG1 and IgG2a and a DTH response characterized by an inflammatory infiltrate with the presence of CD4+ IFN-γ+ T cells. LJM11-immunized mice were intradermally infected in the ear with L. braziliensis in the presence of Lu. longipalpis or Lu. intermedia SGS. A significant reduction of parasite numbers in the ear and lymph node in the group challenged with L. braziliensis plus Lu. longipalpis SGS was observed, but not when the challenge was performed with L. braziliensis plus Lu. intermedia SGS. A higher specific production of IFN-γ and absence of IL-10 by lymph node cells were only observed in LJM11 immunized mice after infection. After two weeks, a similar frequency of CD4+ IFN-γ+ T cells was detected in LJM11 and BSA groups challenged with L. braziliensis plus Lu. longipalpis SGS, suggesting that early events possibly triggered by immunization are essential for protection against Leishmania infection. Our findings support the specificity of saliva-mediated immune responses and reinforce the importance of identifying cross-protective salivary antigens.
Collapse
Affiliation(s)
- Jurema M Cunha
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, BA, Brazil.
| | - Melissa Abbehusen
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, BA, Brazil.
| | - Martha Suarez
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, BA, Brazil.
| | - Jesus Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA.
| | | | - Cláudia I Brodskyn
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, BA, Brazil; Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, BA, Brazil; Instituto Nacional de Ciência e Tecnologia (INCT) de Investigação em Imunologia, Salvador, BA, Brazil.
| |
Collapse
|
25
|
Tlili A, Marzouki S, Chabaane E, Abdeladhim M, Kammoun-Rebai W, Sakkouhi R, Belhadj Hmida N, Oliveira F, Kamhawi S, Louzir H, Valenzuela JG, Ben Ahmed M. Phlebotomus papatasi Yellow-Related and Apyrase Salivary Proteins Are Candidates for Vaccination against Human Cutaneous Leishmaniasis. J Invest Dermatol 2017; 138:598-606. [PMID: 29054598 DOI: 10.1016/j.jid.2017.09.043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 08/25/2017] [Accepted: 09/06/2017] [Indexed: 10/18/2022]
Abstract
Nowadays, there is no available vaccine for human leishmaniasis. Animal experiments demonstrate that pre-exposure to sand fly saliva confers protection against leishmaniasis. Our preceding work in humans indicates that Phlebotomus papatasi saliva induces the production of IL-10 by CD8+ T lymphocytes. The neutralization of IL-10 enhanced the activation of a T-cell CD4+ population-producing IFN-γ. Herein, we used a biochemical and functional genomics approach to identify the sand fly salivary components that are responsible for the activation of the T helper type 1 immune response in humans, therefore constituting potential vaccine candidates against leishmaniasis. Fractionated P. papatasi salivary extracts were first tested on T lymphocytes of immune donors. We confirmed that the CD4+ lymphocytes proliferate and produce IFN-γ in response to stimulation with the proteins of molecular weight >30 kDa. Peripheral blood mononuclear cells from immune donors were transfected with plasmids coding for the most abundant proteins from the P. papatasi salivary gland cDNA library. Our result showed that the "yellow related proteins," PPTSP42 and PPTSP44, and "apyrase," PPTSP36, are the proteins responsible for the aforementioned cellular immune response and IFN-γ production. Strikingly, PPTSP44 triggered the highest level of lymphocyte proliferation and IFN-γ production. Multiplex cytokine analysis confirmed the T helper type 1-polarized response induced by these proteins. Importantly, recombinant PPTSP44 validated the results observed with the DNA plasmid, further supporting that PPTSP44 constitutes a promising vaccine candidate against human leishmaniasis.
Collapse
Affiliation(s)
- Aymen Tlili
- Laboratory of Transmission, Control and Immunobiology of Infections, LR11IPT02, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Soumaya Marzouki
- Laboratory of Transmission, Control and Immunobiology of Infections, LR11IPT02, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Emna Chabaane
- Laboratory of Transmission, Control and Immunobiology of Infections, LR11IPT02, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Maha Abdeladhim
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Wafa Kammoun-Rebai
- Laboratory of Medical Parasitology, Biotechnologies and Biomolecules, LR11IPT06, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Rahma Sakkouhi
- Laboratory of Transmission, Control and Immunobiology of Infections, LR11IPT02, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Nabil Belhadj Hmida
- Laboratory of Transmission, Control and Immunobiology of Infections, LR11IPT02, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Fabiano Oliveira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Hechmi Louzir
- Laboratory of Transmission, Control and Immunobiology of Infections, LR11IPT02, Institut Pasteur de Tunis, Tunis, Tunisia; Faculté de Médecine de Tunis, Université Tunis El Manar, Tunis, Tunisia
| | - Jesus G Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Mélika Ben Ahmed
- Laboratory of Transmission, Control and Immunobiology of Infections, LR11IPT02, Institut Pasteur de Tunis, Tunis, Tunisia; Faculté de Médecine de Tunis, Université Tunis El Manar, Tunis, Tunisia.
| |
Collapse
|
26
|
Kammoun-Rebai W, Bahi-Jaber N, Naouar I, Toumi A, Ben Salah A, Louzir H, Meddeb-Garnaoui A. Human cellular and humoral immune responses to Phlebotomus papatasi salivary gland antigens in endemic areas differing in prevalence of Leishmania major infection. PLoS Negl Trop Dis 2017; 11:e0005905. [PMID: 29023574 PMCID: PMC5638224 DOI: 10.1371/journal.pntd.0005905] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 08/24/2017] [Indexed: 01/22/2023] Open
Abstract
Background Sand fly saliva compounds are able to elicit specific immune responses that have a significant role in Leishmania parasite establishment and disease outcome. Characterizing anti-saliva immune responses in individuals living in well defined leishmaniasis endemic areas would provide valuable insights regarding their effect on parasite transmission and establishment in humans. Methodology/Principal findings We explored the cellular and humoral immune responses to Phlebotomus (P.) papatasi salivary gland extracts (SGE) in individuals living in cutaneous leishmaniasis (CL) old or emerging foci (OF, EF). OF was characterized by a higher infection prevalence as assessed by higher proportions of leishmanin skin test (LST) positive individuals compared to EF. Subjects were further subdivided into healed, asymptomatic or naïve groups. We showed anti-SGE proliferation in less than 30% of the individuals, regardless of the immune status, in both foci. IFN-γ production was higher in OF and only observed in immune individuals from OF and naïve subjects from EF. Although IL-10 was not detected, addition of anti-human IL-10 antibodies revealed an increase in proliferation and IFN-γ production only in individuals from OF. The percentage of seropositive individuals was similar in immune and naïves groups but was significantly higher in OF. No correlation was observed between anti-saliva immune responses and LST response. High anti-SGE-IgG responses were associated with an increased risk of developing ZCL. No differences were observed for anti-SGE humoral or cellular responses among naïve individuals who converted or not their LST response or developed or not ZCL after the transmission season. Conclusions/Significance These data suggest that individuals living in an old focus characterized by a frequent exposure to sand fly bites and a high prevalence of infection, develop higher anti-saliva IgG responses and IFN-γ levels and a skew towards a Th2-type cellular response, probably in favor of parasite establishment, compared to those living in an emerging focus. During murine experimental leishmaniasis sand fly saliva components modulate the host immune response and facilitate infection while pre-exposition to uninfected sand fly bites is associated with a protective cellular response against subsequent infection. Human anti-saliva immune responses are not well defined in leishmaniasis endemic areas. Here, we report an analysis of anti P. papatasi saliva cellular and humoral responses in individuals residing in endemic foci showing different prevalence rates of L. major infection. Individuals were further subdivided based on LST response and presence of typical CL scars. We showed higher anti-saliva cellular and humoral responses and a skew towards a Th2 response in the old focus characterized by the highest prevalence of infection. No correlation was observed between LST and anti-saliva cellular or humoral response. We showed that high anti-saliva IgG responses constituted a risk factor for the development of CL. Our findings suggest that the anti-P. papatasi saliva cellular and humoral response profiles vary with the level of sand fly exposure and the prevalence of infection in CL endemic areas. Such studies in humans from highly endemic areas could contribute to a better understanding of the immune response to sand fly saliva and its role in leishmaniasis outcome.
Collapse
Affiliation(s)
- Wafa Kammoun-Rebai
- Laboratory of Medical Parasitology, Biotechnologies and Biomolecules, Institut Pasteur de Tunis, Tunis, Tunisia
- Département de Biologie, Université Tunis El Manar, Tunis, Tunisia
| | - Narges Bahi-Jaber
- UPSP EGEAL Institut Polytechnique LaSalle Beauvais, Beauvais, France
| | - Ikbel Naouar
- Département de Biologie, Université Tunis El Manar, Tunis, Tunisia
- Laboratory of Transmission, Control and Immunobiology of Infection, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Amine Toumi
- Laboratory of Transmission, Control and Immunobiology of Infection, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Afif Ben Salah
- Laboratory of Transmission, Control and Immunobiology of Infection, Institut Pasteur de Tunis, Tunis, Tunisia
- Arabian Gulf University, College of Medicine and Medical Sciences, Manama, Bahrain
| | - Hechmi Louzir
- Laboratory of Transmission, Control and Immunobiology of Infection, Institut Pasteur de Tunis, Tunis, Tunisia
- Faculté de Médecine de Tunis, Université Tunis El Manar, Tunis, Tunisia
| | - Amel Meddeb-Garnaoui
- Laboratory of Medical Parasitology, Biotechnologies and Biomolecules, Institut Pasteur de Tunis, Tunis, Tunisia
- * E-mail:
| |
Collapse
|
27
|
Kamiya T, Greischar MA, Mideo N. Epidemiological consequences of immune sensitisation by pre-exposure to vector saliva. PLoS Negl Trop Dis 2017; 11:e0005956. [PMID: 28991904 PMCID: PMC5648264 DOI: 10.1371/journal.pntd.0005956] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 10/19/2017] [Accepted: 09/12/2017] [Indexed: 11/25/2022] Open
Abstract
Blood-feeding arthropods—like mosquitoes, sand flies, and ticks—transmit many diseases that impose serious public health and economic burdens. When a blood-feeding arthropod bites a mammal, it injects saliva containing immunogenic compounds that facilitate feeding. Evidence from Leishmania, Plasmodium and arboviral infections suggests that the immune responses elicited by pre-exposure to arthropod saliva can alter disease progression if the host later becomes infected. Such pre-sensitisation of host immunity has been reported to both exacerbate and limit infection symptoms, depending on the system in question, with potential implications for recovery. To explore if and how immune pre-sensitisation alters the effects of vector control, we develop a general model of vector-borne disease. We show that the abundance of pre-sensitised infected hosts should increase when control efforts moderately increase vector mortality rates. If immune pre-sensitisation leads to more rapid clearance of infection, increasing vector mortality rates may achieve greater than expected disease control. However, when immune pre-sensitisation prolongs the duration of infection, e.g., through mildly symptomatic cases for which treatment is unlikely to be sought, vector control can actually increase the total number of infected hosts. The rising infections may go unnoticed unless active surveillance methods are used to detect such sub-clinical individuals, who could provide long-lasting reservoirs for transmission and suffer long-term health consequences of those sub-clinical infections. Sensitivity analysis suggests that these negative consequences could be mitigated through integrated vector management. While the effect of saliva pre-exposure on acute symptoms is well-studied for leishmaniasis, the immunological and clinical consequences are largely uncharted for other vector-parasite-host combinations. We find a large range of plausible epidemiological outcomes, positive and negative for public health, underscoring the need to quantify how immune pre-sensitisation modulates recovery and transmission rates in vector-borne diseases. Many diseases of health and economic importance are transmitted by arthropod vectors, like mosquitoes, sand flies, and ticks. When a blood-feeding arthropod bites a mammal, it injects saliva containing compounds that facilitate feeding. The immune responses elicited by previous exposure to vector saliva can alter disease severity if the host later becomes infected. Such pre-sensitisation of host immunity has been linked to either exacerbation or mitigation of symptoms in a number of disease systems. We develop a general model of vector-borne disease to examine how vector control efforts alter the frequency of immune pre-sensitisation and thus change the epidemiological impact of control. We show that the abundance of pre-sensitised infected hosts should increase when control efforts moderately increase vector mortality rates. When immune pre-sensitisation leads to longer infections—by generating sub-clinical cases for which treatment is not rapidly sought—killing vectors can lead to unexpected increases in the number of infected hosts. The rising case burden may go unnoticed unless sub-clinical individuals are tested for infection. Conversely, if immune pre-sensitisation leads to more rapid clearance of infection, increasing vector mortality rates may achieve greater than expected disease control. Our findings highlight the need to quantify how immune pre-sensitisation modulates clinical outcomes and parasite transmission in humans.
Collapse
Affiliation(s)
- Tsukushi Kamiya
- Department of Ecology & Evolutionary Biology, University of Toronto, Toronto, Ontario, Canada
| | - Megan A Greischar
- Department of Ecology & Evolutionary Biology, University of Toronto, Toronto, Ontario, Canada
| | - Nicole Mideo
- Department of Ecology & Evolutionary Biology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
28
|
Oliveira F, Rowton E, Aslan H, Gomes R, Castrovinci PA, Alvarenga PH, Abdeladhim M, Teixeira C, Meneses C, Kleeman LT, Guimarães-Costa AB, Rowland TE, Gilmore D, Doumbia S, Reed SG, Lawyer PG, Andersen JF, Kamhawi S, Valenzuela JG. A sand fly salivary protein vaccine shows efficacy against vector-transmitted cutaneous leishmaniasis in nonhuman primates. Sci Transl Med 2016; 7:290ra90. [PMID: 26041707 DOI: 10.1126/scitranslmed.aaa3043] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Currently, there are no commercially available human vaccines against leishmaniasis. In rodents, cellular immunity to salivary proteins of sand fly vectors is associated to protection against leishmaniasis, making them worthy targets for further exploration as vaccines. We demonstrate that nonhuman primates (NHP) exposed to Phlebotomus duboscqi uninfected sand fly bites or immunized with salivary protein PdSP15 are protected against cutaneous leishmaniasis initiated by infected bites. Uninfected sand fly-exposed and 7 of 10 PdSP15-immunized rhesus macaques displayed a significant reduction in disease and parasite burden compared to controls. Protection correlated to the early appearance of Leishmania-specific CD4(+)IFN-γ(+) lymphocytes, suggesting that immunity to saliva or PdSP15 augments the host immune response to the parasites while maintaining minimal pathology. Notably, the 30% unprotected PdSP15-immunized NHP developed neither immunity to PdSP15 nor an accelerated Leishmania-specific immunity. Sera and peripheral blood mononuclear cells from individuals naturally exposed to P. duboscqi bites recognized PdSP15, demonstrating its immunogenicity in humans. PdSP15 sequence and structure show no homology to mammalian proteins, further demonstrating its potential as a component of a vaccine for human leishmaniasis.
Collapse
Affiliation(s)
- Fabiano Oliveira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Edgar Rowton
- Department of Entomology, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Hamide Aslan
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Regis Gomes
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA. Centro de Pesquisas Gonçalo Moniz (CPqGM)-Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Bahia 40296-710, Brazil
| | - Philip A Castrovinci
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Patricia H Alvarenga
- Laboratório de Bioquímica de Resposta ao Estresse, Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil. Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - 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, USA
| | - Clarissa Teixeira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA. Centro de Pesquisas Gonçalo Moniz (CPqGM)-Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Bahia 40296-710, Brazil
| | - Claudio Meneses
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Lindsey T Kleeman
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Anderson B Guimarães-Costa
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Tobin E Rowland
- Department of Entomology, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Dana Gilmore
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Seydou Doumbia
- Faculty of Medicine, Pharmacy and Odontostomatology, University of Bamako, Bamako 1805, Mali
| | - Steven G Reed
- Infectious Disease Research Institute, Seattle, WA 98102, USA
| | - Phillip G Lawyer
- Department of Entomology, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - John F Andersen
- Vector Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA.
| | - Jesus G Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA.
| |
Collapse
|
29
|
Reed SG, Coler RN, Mondal D, Kamhawi S, Valenzuela JG. Leishmania vaccine development: exploiting the host-vector-parasite interface. Expert Rev Vaccines 2015; 15:81-90. [PMID: 26595093 PMCID: PMC6019289 DOI: 10.1586/14760584.2016.1105135] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Visceral leishmaniasis (VL) is a disease transmitted by phlebotomine sand flies, fatal if untreated, and with no available human vaccine. In rodents, cellular immunity to Leishmania parasite proteins as well as salivary proteins of the sand fly is associated with protection, making them worthy targets for further exploration as vaccines. This review discusses the notion that a combination vaccine including Leishmania and vector salivary antigens may improve vaccine efficacy by targeting the parasite at its most vulnerable stage just after transmission. Furthermore, we put forward the notion that better modeling of natural transmission is needed to test efficacy of vaccines. For example, the fact that individuals living in endemic areas are exposed to sand fly bites and will mount an immune response to salivary proteins should be considered in pre-clinical and clinical evaluation of leishmaniasis vaccines. Nevertheless, despite remaining obstacles there is good reason to be optimistic that safe and effective vaccines against leishmaniasis can be developed.
Collapse
Affiliation(s)
- S G Reed
- a Infectious Disease Research Institute , Seattle WA , USA
- b Department of Global Health , University of Washington , Seattle WA , USA
| | - R N Coler
- a Infectious Disease Research Institute , Seattle WA , USA
- b Department of Global Health , University of Washington , Seattle WA , USA
| | - D Mondal
- c International Center for Diarrhoeal Diseases Research, Centre for Nutrition and Food Security , Parasitology Laboratory , Dhaka , Bangladesh
| | - S Kamhawi
- d Vector Molecular Biology Section, LMVR , National Institute of Allergy and Infectious Diseases, NIH , Rockville , MD , USA
| | - J G Valenzuela
- d Vector Molecular Biology Section, LMVR , National Institute of Allergy and Infectious Diseases, NIH , Rockville , MD , USA
| |
Collapse
|
30
|
McDowell MA. Vector-transmitted disease vaccines: targeting salivary proteins in transmission (SPIT). Trends Parasitol 2015; 31:363-72. [PMID: 26003330 DOI: 10.1016/j.pt.2015.04.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 04/20/2015] [Accepted: 04/27/2015] [Indexed: 12/25/2022]
Abstract
More than half the population of the world is at risk for morbidity and mortality from vector-transmitted diseases, and emerging vector-transmitted infections are threatening new populations. Rising insecticide resistance and lack of efficacious vaccines highlight the need for novel control measures. One such approach is targeting the vector-host interface by incorporating vector salivary proteins in anti-pathogen vaccines. Debate remains about whether vector saliva exposure exacerbates or protects against more severe clinical manifestations, induces immunity through natural exposure or extends to all vector species and associated pathogens. Nevertheless, exploiting this unique biology holds promise as a viable strategy for the development of vaccines against vector-transmitted diseases.
Collapse
Affiliation(s)
- Mary Ann McDowell
- Eck Institute for Global Health, Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
31
|
Severity of old world cutaneous leishmaniasis is influenced by previous exposure to sandfly bites in Saudi Arabia. PLoS Negl Trop Dis 2015; 9:e0003449. [PMID: 25646796 PMCID: PMC4315490 DOI: 10.1371/journal.pntd.0003449] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 11/27/2014] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND The sandfly Phlebotomus papatasi is the vector of Leishmania major, the main causative agent of Old World cutaneous leishmaniasis (CL) in Saudi Arabia. Sandflies inject saliva while feeding and the salivary protein PpSP32 was previously shown to be a biomarker for bite exposure. Here we used recombinant PpSP32 to evaluate human exposure to Ph. papatasi bites, and study the association between antibody response to saliva and CL in endemic areas in Saudi Arabia. METHODOLOGY/PRINCIPAL FINDINGS In this observational study, anti-PpSP32 antibodies, as indicators of exposure to sandfly bites, were measured in sera from healthy individuals and patients from endemic regions in Saudi Arabia with active and cured CL. Ph. papatasi was identified as the primary CL vector in the study area. Anti-PpSP32 antibody levels were significantly higher in CL patients presenting active infections from all geographical regions compared to CL cured and healthy individuals. Furthermore, higher anti-PpSP32 antibody levels correlated with the prevalence and type of CL lesions (nodular vs. papular) observed in patients, especially non-local construction workers. CONCLUSIONS Our findings suggest a possible correlation between the type of immunity generated by the exposure to sandfly bites and disease outcome.
Collapse
|
32
|
Carvalho AM, Cristal JR, Muniz AC, Carvalho LP, Gomes R, Miranda JC, Barral A, Carvalho EM, de Oliveira CI. Interleukin 10-Dominant Immune Response and Increased Risk of Cutaneous Leishmaniasis After Natural Exposure to Lutzomyia intermedia Sand Flies. J Infect Dis 2015; 212:157-65. [PMID: 25596303 DOI: 10.1093/infdis/jiv020] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 01/01/2015] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Leishmaniasis is caused by parasites transmitted to the vertebrate host by infected sand flies. During transmission, the vertebrate host is also inoculated with sand fly saliva, which exerts powerful immunomodulatory effects on the host's immune response. METHODS We conducted a prospective cohort analysis to characterize the human immune response to Lutzomyia intermedia saliva in 264 individuals, from an area for cutaneous leishmaniasis (CL) caused by Leishmania braziliensis. RESULTS Antibodies were found in 150 individuals (56.8%); immunoglobulin G1 and G4 were the predominant subclasses. Recall responses to salivary gland sonicate showed elevated production of interleukin 10 (IL-10), interleukin 13, interferon γ, CXCL9, and CCL2 compared with controls. CD4(+)CD25(+) T cells, including Foxp3(+) cells, were the main source of IL-10. L. braziliensis replication was increased (P < .05) in macrophages cocultured with saliva-stimulated lymphocytes from exposed individuals and addition of anti-IL-10 reverted this effect. Positive correlation between antibody response to saliva and cellular response to Leishmania was not found. Importantly, individuals seropositive to saliva are 2.1 times more likely to develop CL (relative risk, 2.1; 95% confidence interval, 1.07-4.2; P < .05). CONCLUSIONS Exposure to L. intermedia sand flies skews the human immune response, facilitating L. braziliensis survival in vitro, and increases the risk of developing CL.
Collapse
Affiliation(s)
| | | | - Aline C Muniz
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia
| | - Lucas P Carvalho
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais
| | - Regis Gomes
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz
| | - José C Miranda
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz
| | - Aldina Barral
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz Instituto de Investigação em Imunologia, Salvador, Bahia, Brazil
| | - Edgar M Carvalho
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais
| | - Camila I de Oliveira
- Fundação Oswaldo Cruz, Centro de Pesquisas Gonçalo Moniz Instituto de Investigação em Imunologia, Salvador, Bahia, Brazil
| |
Collapse
|
33
|
Thakur A, Kaur H, Kaur S. Evaluation of the immunoprophylactic potential of a killed vaccine candidate in combination with different adjuvants against murine visceral leishmaniasis. Parasitol Int 2014; 64:70-8. [PMID: 25316605 DOI: 10.1016/j.parint.2014.10.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 10/02/2014] [Accepted: 10/06/2014] [Indexed: 01/13/2023]
Abstract
Despite a large number of field trials, till date no prophylactic antileishmanial vaccine exists for human use. Killed antigen formulations offer the advantage of being safe but they have limited immunogenicity. Recent research has documented that efforts to develop effective Leishmania vaccine have been limited due to the lack of an appropriate adjuvant. Addition of adjuvants to vaccines boosts and directs the immunogenicity of antigens. So, the present study was done to evaluate the effectiveness of four adjuvants i.e. alum, saponin, cationic liposomes and monophosphoryl lipid-A in combination with Autoclaved Leishmania donovani (ALD) antigen against murine visceral leishmaniasis (VL). BALB/c mice were immunized thrice with respective vaccine formulation. Two weeks after last booster, challenge infection was given. Mice were sacrificed 15 days after last immunization and on 30, 60 and 90 post infection/challenge days. A considerable protective efficacy was shown by all vaccine formulations. It was evident from significant reduction in parasite load, profound delayed type hypersensitivity responses (DTH), increased IgG2a titres and high levels of Th1 cytokines (IFN-γ, IL-12) as compared to the infected controls. However, level of protection varied with the type of adjuvant used. Maximum protection was achieved with the use of liposome encapsulated ALD antigen and it was closely followed by group immunized with ALD+MPL-A. Significant results were also obtained with ALD+saponin, ALD+alum and ALD antigen (alone) but the protective efficacy was reduced as compared to other immunized groups. The present study reveals greater efficacy of two vaccine formulations i.e. ALD+liposome and ALD+MPL-A against murine VL.
Collapse
Affiliation(s)
- Ankita Thakur
- Department of Zoology, Panjab University, Chandigarh 160014, India
| | - Harpreet Kaur
- Department of Zoology, Panjab University, Chandigarh 160014, India
| | - Sukhbir Kaur
- Department of Zoology, Panjab University, Chandigarh 160014, India.
| |
Collapse
|
34
|
Ockenfels B, Michael E, McDowell MA. Meta-analysis of the effects of insect vector saliva on host immune responses and infection of vector-transmitted pathogens: a focus on leishmaniasis. PLoS Negl Trop Dis 2014; 8:e3197. [PMID: 25275509 PMCID: PMC4183472 DOI: 10.1371/journal.pntd.0003197] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 08/18/2014] [Indexed: 11/18/2022] Open
Abstract
A meta-analysis of the effects of vector saliva on the immune response and progression of vector-transmitted disease, specifically with regard to pathology, infection level, and host cytokine levels was conducted. Infection in the absence or presence of saliva in naïve mice was compared. In addition, infection in mice pre-exposed to uninfected vector saliva was compared to infection in unexposed mice. To control for differences in vector and pathogen species, mouse strain, and experimental design, a random effects model was used to compare the ratio of the natural log of the experimental to the control means of the studies. Saliva was demonstrated to enhance pathology, infection level, and the production of Th2 cytokines (IL-4 and IL-10) in naïve mice. This effect was observed across vector/pathogen pairings, whether natural or unnatural, and with single salivary proteins used as a proxy for whole saliva. Saliva pre-exposure was determined to result in less severe leishmaniasis pathology when compared with unexposed mice infected either in the presence or absence of sand fly saliva. The results of further analyses were not significant, but demonstrated trends toward protection and IFN-γ elevation for pre-exposed mice. Arthropod vectors transmit a wide variety of diseases resulting in substantial human morbidity and economic costs worldwide. When hematophagous arthropods blood feed, they release saliva into the host. This saliva elicits a strong immune response and has recently been a focus for vaccine research. There is evidence that the saliva enhances infection in naïve hosts, but that prior exposure to saliva results in less severe infection. This analysis endeavored to determine whether there was a statistically significant enhancement or protective effect with regard to saliva exposure and the progression of disease, and to determine the underlying immune mechanism driving these effects. We found that saliva does indeed enhance infection levels of vector-transmitted pathogens and leishmaniasis pathology in naïve mice and elevates Th2 cytokine levels (IL-4 and IL-10). We also determined that pre-exposure to saliva results in less severe pathology of experimental leishmaniasis in mice. These results are important for vaccine trials and vector control programs, though more studies are needed with regard to pre-exposure.
Collapse
Affiliation(s)
- Brittany Ockenfels
- Eck Institute for Global Health, Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Edwin Michael
- Eck Institute for Global Health, Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Mary Ann McDowell
- Eck Institute for Global Health, Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
- * E-mail:
| |
Collapse
|
35
|
HABIBI G, ESMAEIL-NIA K, IZADI H, ATAIE AMARLOIE O, AFSHARI A, BORDBAR N. Immunological analysis of aerobic bioreactor bovine theileriosis vaccine. IRANIAN JOURNAL OF PARASITOLOGY 2014; 9:382-93. [PMID: 25678923 PMCID: PMC4316570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 06/21/2014] [Indexed: 11/07/2022]
Abstract
BACKGROUND In this study, the pilot production of aerobic bioreactor tropical theileriosis vaccine was optimized with the aim of immunological assays for further mass production. METHODS We have shown earlier the delayed type hypersensitivity (DTH) assay could be used for evaluating the immunity and memory cells against specific Theileria antigen in vaccinated animals. In addition, TNF-α is the principle cytokine in modulating the cytotoxic activity of cytotoxic T-lymphocytes (CTL). Immunological analysis of the vaccine was performed by using two cell mediated immunity (CMI) in vitro and in vivo DTH test (Theilerin) and TNF-α assay. RESULTS The results of immune responses of susceptible immunized cattle by bioreactor vaccine in comparison with conventional flask vaccine revealed a significant stimulation of immune cells by transcription of high level of TNF-α and positive reaction against Theileria antigen in Theilerin skin test (DTH). CONCLUSION The equal immunological results achieved in both above mentioned vaccines verified the satisfactory immunity for aerobic bioreactor theileriosis vaccine for advance mass vaccination in the field on a large-scale.
Collapse
Affiliation(s)
- Gholamreza HABIBI
- Department of Parasite Vaccine Research and Production, Razi Vaccine and Serum Research Institute, Karaj, Iran
| | - Kasra ESMAEIL-NIA
- Department of Parasite Vaccine Research and Production, Razi Vaccine and Serum Research Institute, Karaj, Iran
| | - Hasan IZADI
- Department of Foot and Mouth Disease Vaccine production, Razi Vaccine and Serum Research Institute, Karaj, Iran
| | - Orang ATAIE AMARLOIE
- Department of Clinical Sciences, Faculty of Veterinary Sciences, Islamic Azad University, Karaj, Iran
| | - Asghar AFSHARI
- Department of Parasite Vaccine Research and Production, Razi Vaccine and Serum Research Institute, Karaj, Iran
| | - Nadia BORDBAR
- Department of Quality Control of Biological Products, Razi Vaccine and Serum Research Institute, Karaj, Iran
| |
Collapse
|
36
|
Abdeladhim M, Kamhawi S, Valenzuela JG. What's behind a sand fly bite? The profound effect of sand fly saliva on host hemostasis, inflammation and immunity. INFECTION GENETICS AND EVOLUTION 2014; 28:691-703. [PMID: 25117872 DOI: 10.1016/j.meegid.2014.07.028] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Revised: 07/15/2014] [Accepted: 07/24/2014] [Indexed: 10/24/2022]
Abstract
Sand flies are blood-feeding insects and vectors of the Leishmania parasite. For many years, saliva of these insects has represented a gold mine for the discovery of molecules with anti-hemostatic and immuno-modulatory activities. Furthermore, proteins in sand fly saliva have been shown to be a potential vaccine against leishmaniasis and also markers of vector exposure. A bottleneck to progress in these areas of research has been the identification of molecules responsible for the observed activities and properties of saliva. Over the past decade, rapid advances in transcriptomics and proteomics resulted in the completion of a number of sialomes (salivary gland transcriptomes) and the expression of several recombinant salivary proteins from different species of sand fly vectors. This review will provide readers with a comprehensive update of recent advances in the characterization of these salivary molecules and their biological activities and offer insights pertaining to their protective effect against leishmaniasis and their potential as markers of vector exposure.
Collapse
Affiliation(s)
- Maha Abdeladhim
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, United States
| | - Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, United States.
| | - Jesus G Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, United States.
| |
Collapse
|
37
|
Kamhawi S, Aslan H, Valenzuela JG. Vector saliva in vaccines for visceral leishmaniasis: a brief encounter of high consequence? Front Public Health 2014; 2:99. [PMID: 25152872 PMCID: PMC4126209 DOI: 10.3389/fpubh.2014.00099] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 07/14/2014] [Indexed: 11/25/2022] Open
Abstract
Visceral leishmaniasis (VL) is a vector-borne disease transmitted by phlebotomine sand flies and remains the most serious form of the disease with no available human vaccine. Repeatedly, studies have demonstrated the immunogenicity and protective efficacy of a number of sand fly salivary proteins against cutaneous and visceral leishmaniasis. All Leishmania species including agents of VL are co-deposited into the skin together with vector saliva. Generally, the immune response to a protective salivary protein in vaccinated animals is rapid and possibly acts on the parasites soon after delivery into the skin by the bite of an infective sand fly. This is followed by the development of a stronger Leishmania-specific immunity in saliva-vaccinated animals compared to controls. Considering that several of the most efficacious protective molecules were identified from a proven vector of VL, we put forward the notion that a combination vaccine that includes a Leishmania antigen and a vector salivary protein has the potential to improve vaccine efficacy by targeting the parasite at it most vulnerable stage just after transmission.
Collapse
Affiliation(s)
- Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Hamide Aslan
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Jesus G. Valenzuela
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
38
|
Teixeira C, Gomes R, Oliveira F, Meneses C, Gilmore DC, Elnaiem DEA, Valenzuela JG, Kamhawi S. Characterization of the early inflammatory infiltrate at the feeding site of infected sand flies in mice protected from vector-transmitted Leishmania major by exposure to uninfected bites. PLoS Negl Trop Dis 2014; 8:e2781. [PMID: 24762408 PMCID: PMC3998922 DOI: 10.1371/journal.pntd.0002781] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 02/23/2014] [Indexed: 11/18/2022] Open
Abstract
Background Mice exposed to sand fly saliva are protected against vector-transmitted Leishmania major. Although protection has been related to IFN- γ producing T cells, the early inflammatory response orchestrating this outcome has not been defined. Methodology/Principal findings Mice exposed to uninfected P. duboscqi bites and naïve mice were challenged with L. major-infected flies to characterize their early immune response at the bite site. Mostly, chemokine and cytokine transcript expression post-infected bites was amplified in exposed compared to naïve mice. In exposed mice, induced chemokines were mostly involved in leukocyte recruitment and T cell and NK cell activation; IL-4 was expressed at 6 h followed by IFN-γ and iNOS2 as well as IL-5 and IL-10 expression. In naïve animals, the transcript expression following Leishmania-infected sand fly bites was suppressed. Expression profiles translated to an earlier and significantly larger recruitment of leukocytes including neutrophils, macrophages, Gr+ monocytes, NK cells and CD4+ T cells to the bite site of exposed compared to naïve mice post-infected bites. Additionally, up to 48 hours post-infected bites the number of IFN-γ-producing CD4+T cells and NK cells arriving at the bite site was significantly higher in exposed compared to naïve mice. Thereafter, NK cells become cytolytic and persist at the bite site up to a week post-bite. Conclusion/Significance The quiet environment induced by a Leishmania-infected sand fly bite in naïve mice was significantly altered in animals previously exposed to saliva of uninfected flies. We propose that the enhanced recruitment of Gr+ monocytes, NK cells and CD4 Th1 cells observed at the bite site of exposed mice creates an inhospitable environment that counters the establishment of L. major infection. Sand flies transmit Leishmania parasites during bloodfeeding. Salivary molecules are deposited alongside parasites and can reshape the host's immune response to infection. Exposure to uninfected sand fly bites or immunization with salivary molecules protects the host against Leishmania infection. Here we show that mice exposed to bites of uninfected Phlebotomus duboscqi sand flies are protected against P. duboscqi-transmitted L. major and characterize the formerly unknown early cellular infiltrate at the bite site following L.major vector-transmission. The kinetics and nature of the inflammatory response at the bite site of exposed mice were notably different from those of naïve mice showing an amplified expression of cytokines and chemokines after parasite transmission. The transcripts reflected a faster and more robust infiltrate of immune cells to the bite site of exposed mice composed of neutrophils, macrophages, monocytes, NK cells and CD4+ T cells. In addition, there was an increased influx of activated IFN-γ producing CD4+ T cells and Granzyme B-producing mature NK cells in exposed animals. These findings suggest that the observed robust and persistent proinflammatory response in exposed animals restrict parasite multiplication.
Collapse
Affiliation(s)
- Clarissa Teixeira
- 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
- Centro de Pesquisas Gonçalo Moniz, Fiocruz, Bahia, Brazil
| | - Regis Gomes
- 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
- Centro de Pesquisas Gonçalo Moniz, Fiocruz, Bahia, Brazil
| | - Fabiano Oliveira
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Claudio Meneses
- 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
| | - Dana C. Gilmore
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Dia-Eldin A. Elnaiem
- Department of Zoology, University of Maryland Eastern Shore, Princess Anne, 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); (SK)
| | - Shaden Kamhawi
- Vector Molecular Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
- * E-mail: (JGV); (SK)
| |
Collapse
|
39
|
Allergy to Hematophagous Arthropods Bites. CURRENT DERMATOLOGY REPORTS 2014. [DOI: 10.1007/s13671-013-0065-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
40
|
Oliveira F, de Carvalho AM, de Oliveira CI. Sand-fly saliva-leishmania-man: the trigger trio. Front Immunol 2013; 4:375. [PMID: 24312093 PMCID: PMC3832839 DOI: 10.3389/fimmu.2013.00375] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 10/31/2013] [Indexed: 11/23/2022] Open
Abstract
Leishmaniases are worldwide diseases transmitted to the vertebrate host by the bite of an infected sand-fly. Sand-fly biting and parasite inoculation are accompanied by the injection of salivary molecules, whose immunomodulatory properties are actively being studied. This mini review focuses on how the interactions between sand-fly saliva and the immune system may shape the outcome of infection, given its immunomodulatory properties, in experimental models and in the endemic area. Additionally, we approach the recent contributions regarding the identification of individual salivary components and how these are currently being considered as additional components of a vaccine against leishmaniasis.
Collapse
Affiliation(s)
- Fabiano Oliveira
- Vector Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Rockville, MD , USA
| | | | | |
Collapse
|
41
|
Walker DM, Oghumu S, Gupta G, McGwire BS, Drew ME, Satoskar AR. Mechanisms of cellular invasion by intracellular parasites. Cell Mol Life Sci 2013; 71:1245-63. [PMID: 24221133 DOI: 10.1007/s00018-013-1491-1] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 10/04/2013] [Accepted: 10/07/2013] [Indexed: 12/22/2022]
Abstract
Numerous disease-causing parasites must invade host cells in order to prosper. Collectively, such pathogens are responsible for a staggering amount of human sickness and death throughout the world. Leishmaniasis, Chagas disease, toxoplasmosis, and malaria are neglected diseases and therefore are linked to socio-economical and geographical factors, affecting well-over half the world's population. Such obligate intracellular parasites have co-evolved with humans to establish a complexity of specific molecular parasite-host cell interactions, forming the basis of the parasite's cellular tropism. They make use of such interactions to invade host cells as a means to migrate through various tissues, to evade the host immune system, and to undergo intracellular replication. These cellular migration and invasion events are absolutely essential for the completion of the lifecycles of these parasites and lead to their for disease pathogenesis. This review is an overview of the molecular mechanisms of protozoan parasite invasion of host cells and discussion of therapeutic strategies, which could be developed by targeting these invasion pathways. Specifically, we focus on four species of protozoan parasites Leishmania, Trypanosoma cruzi, Plasmodium, and Toxoplasma, which are responsible for significant morbidity and mortality.
Collapse
Affiliation(s)
- Dawn M Walker
- Department of Microbial Infection and Immunity, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA
| | | | | | | | | | | |
Collapse
|