1
|
Huang Q, Gavor E, Tulsian NK, Fan J, Lin Q, Mok YK, Kini RM, Sivaraman J. Structural and functional characterization of Aedes aegypti pupal cuticle protein that controls dengue virus infection. Protein Sci 2023; 32:e4761. [PMID: 37593853 PMCID: PMC10510476 DOI: 10.1002/pro.4761] [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: 06/08/2023] [Revised: 08/13/2023] [Accepted: 08/14/2023] [Indexed: 08/19/2023]
Abstract
The pupal cuticle protein from Aedes aegypti (AaPC) inhibits dengue virus (DENV) infection; however, the underlying mechanism of this inhibition remains unknown. Here, we report that AaPC is an intrinsically disordered protein and interacts with domain I/II of the DENV envelope protein via residues Asp59, Asp61, Glu71, Asp73, Ser75, and Asp80. AaPC can directly bind to and cause the aggregation of DENV, which in turn blocks virus infection during the virus-cell fusion stage. AaPC may also influence viral recognition and attachment by interacting with human immune receptors DC-SIGN and CD4. These findings enhance our understanding of the role of AaPC in mitigating viral infection and suggest that AaPC is a potential target for developing inhibitors or antibodies to control dengue virus infection.
Collapse
Affiliation(s)
- Qingqing Huang
- Department of Biological SciencesNational University of SingaporeSingaporeSingapore
| | - Edem Gavor
- Department of Biological SciencesNational University of SingaporeSingaporeSingapore
| | - Nikhil Kumar Tulsian
- Department of Biological SciencesNational University of SingaporeSingaporeSingapore
- Department of BiochemistryNational University of SingaporeSingaporeSingapore
| | - Jingsong Fan
- Department of Biological SciencesNational University of SingaporeSingaporeSingapore
| | - Qingsong Lin
- Department of Biological SciencesNational University of SingaporeSingaporeSingapore
| | - Yu Keung Mok
- Department of Biological SciencesNational University of SingaporeSingaporeSingapore
| | - R. Manjunatha Kini
- Department of Biological SciencesNational University of SingaporeSingaporeSingapore
- Department of Pharmacology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - J. Sivaraman
- Department of Biological SciencesNational University of SingaporeSingaporeSingapore
| |
Collapse
|
2
|
Mancini MV, Tandavanitj R, Ant TH, Murdochy SM, Gingell DD, Setthapramote C, Natsrita P, Kohl A, Sinkins SP, Patel AH, De Lorenzo G. Evaluation of an Engineered Zika Virus-Like Particle Vaccine Candidate in a Mosquito-Mouse Transmission Model. mSphere 2023; 8:e0056422. [PMID: 36840596 PMCID: PMC10117074 DOI: 10.1128/msphere.00564-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 01/31/2023] [Indexed: 02/25/2023] Open
Abstract
The primary route of Zika virus (ZIKV) transmission is through the bite of an infected Aedes mosquito, when it probes the skin of a vertebrate host during a blood meal. Viral particles are injected into the bite site together with mosquito saliva and a complex mixture of other components. Some of them are known to play a key role in the augmentation of the arbovirus infection in the host, with increased viremia and/or morbidity. This vector-derived contribution to the infection is not usually considered when vaccine candidates are tested in preclinical animal models. In this study, we performed a preclinical validation of a promising ZIKV vaccine candidate in a mosquito-mouse transmission model using both Asian and African ZIKV lineages. Mice were immunized with engineered ZIKV virus-like particles and subsequently infected through the bite of ZIKV-infected Aedes aegypti mosquitoes. Despite a mild increase in viremia in mosquito-infected mice compared to those infected through traditional needle injection, the vaccine protected the animals from developing the disease and strongly reduced viremia. In addition, during peak viremia, naive mosquitoes were allowed to feed on infected vaccinated and nonvaccinated mice. Our analysis of viral titers in mosquitos showed that the vaccine was able to inhibit virus transmission from the host to the vector. IMPORTANCE Zika is a mosquito-borne viral disease, causing acute debilitating symptoms and complications in infected individuals and irreversible neuronal abnormalities in newborn children. The primary vectors of ZIKV are Aedes aegypti mosquitoes. Despite representing a significant public health burden with a widespread transmission in many regions of the world, Zika remains a neglected disease with no effective antiviral therapies or approved vaccines. It is known that components of the mosquito bite lead to an enhancement of viral infection and spread, but this aspect is often overlooked when vaccine candidates undergo preclinical validation. In this study, we included mosquitoes as viral vectors, demonstrating the ability of a promising vaccine candidate to protect animals against ZIKV infections after the bite of an infected mosquito and to also prevent its further transmission. These findings represent an additional crucial step for the development of an effective prevention tool for clinical use.
Collapse
Affiliation(s)
| | - Rapeepat Tandavanitj
- MRC–University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
- Biologicals Research Group, Research and Development Institute, Government Pharmaceutical Organization, Bangkok, Thailand
| | - Thomas H. Ant
- MRC–University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Shivan M. Murdochy
- MRC–University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Daniel D. Gingell
- MRC–University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Chayanee Setthapramote
- MRC–University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
- Department of Clinical Pathology, Faculty of Medicine Vajira Hospital, Navamindradhiraj University, Bangkok, Thailand
| | - Piyatida Natsrita
- MRC–University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Alain Kohl
- MRC–University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Steven P. Sinkins
- MRC–University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Arvind H. Patel
- MRC–University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| | - Giuditta De Lorenzo
- MRC–University of Glasgow Centre for Virus Research, Glasgow, Scotland, United Kingdom
| |
Collapse
|
3
|
Visser I, Koenraadt CJ, Koopmans MP, Rockx B. The significance of mosquito saliva in arbovirus transmission and pathogenesis in the vertebrate host. One Health 2023. [DOI: 10.1016/j.onehlt.2023.100506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023] Open
|
4
|
García-Sancha N, Corchado-Cobos R, Gómez-Vecino A, Jiménez-Navas A, Pérez-Baena MJ, Blanco-Gómez A, Holgado-Madruga M, Mao JH, Cañueto J, Castillo-Lluva S, Mendiburu-Eliçabe M, Pérez-Losada J. Evolutionary Origins of Metabolic Reprogramming in Cancer. Int J Mol Sci 2022; 23:ijms232012063. [PMID: 36292921 PMCID: PMC9603151 DOI: 10.3390/ijms232012063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/29/2022] [Accepted: 10/06/2022] [Indexed: 11/23/2022] Open
Abstract
Metabolic changes that facilitate tumor growth are one of the hallmarks of cancer. These changes are not specific to tumors but also take place during the physiological growth of tissues. Indeed, the cellular and tissue mechanisms present in the tumor have their physiological counterpart in the repair of tissue lesions and wound healing. These molecular mechanisms have been acquired during metazoan evolution, first to eliminate the infection of the tissue injury, then to enter an effective regenerative phase. Cancer itself could be considered a phenomenon of antagonistic pleiotropy of the genes involved in effective tissue repair. Cancer and tissue repair are complex traits that share many intermediate phenotypes at the molecular, cellular, and tissue levels, and all of these are integrated within a Systems Biology structure. Complex traits are influenced by a multitude of common genes, each with a weak effect. This polygenic component of complex traits is mainly unknown and so makes up part of the missing heritability. Here, we try to integrate these different perspectives from the point of view of the metabolic changes observed in cancer.
Collapse
Affiliation(s)
- Natalia García-Sancha
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Roberto Corchado-Cobos
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Aurora Gómez-Vecino
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Alejandro Jiménez-Navas
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Manuel Jesús Pérez-Baena
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Adrián Blanco-Gómez
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Marina Holgado-Madruga
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain
- Departamento de Fisiología y Farmacología, Universidad de Salamanca, 37007 Salamanca, Spain
- Instituto de Neurociencias de Castilla y León (INCyL), 37007 Salamanca, Spain
| | - Jian-Hua Mao
- Lawrence Berkeley National Laboratory, Biological Systems and Engineering Division, Berkeley, CA 94720, USA
- Berkeley Biomedical Data Science Center, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Javier Cañueto
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain
- Departamento de Dermatología, Hospital Universitario de Salamanca, Paseo de San Vicente 58-182, 37007 Salamanca, Spain
| | - Sonia Castillo-Lluva
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense, 28040 Madrid, Spain
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC), 28040 Madrid, Spain
| | - Marina Mendiburu-Eliçabe
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain
- Correspondence: (M.M.-E.); (J.P.-L.)
| | - Jesús Pérez-Losada
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain
- Correspondence: (M.M.-E.); (J.P.-L.)
| |
Collapse
|
5
|
Screening and analysis of immune-related genes of Aedes aegypti infected with DENV2. Acta Trop 2022; 236:106698. [PMID: 36162456 DOI: 10.1016/j.actatropica.2022.106698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/17/2022] [Accepted: 09/18/2022] [Indexed: 01/08/2023]
Abstract
Dengue virus type Ⅱ (DENV2) is a primary serotype responsible for the dengue fever epidemic, and Aedes aegypti is the main DENV2 vector. Understanding the Aedes aegypti immune mechanism against DENV2 is the basis for research on immune blockade in mosquitoes. Some preliminary studies lack validation in the literature, so this study was performed to further study and validate the potential target genes to provide a further basis for screening key target genes. We screened 51 genes possibly related to Aedes aegypti infection and immunity from the literature for further verification. First, bioinformatic methods such as GO, KEGG and PPI analysis were used, and then RT-qPCR was used to detect the changes in mRNA expression in the midguts and salivary glands of Aedes aegypti infected with DENV2.Bioinformatic analysis showed that mostly genes of the glucose metabolism pathway and myoprotein were influenced. In salivary glands, the Gst (xa) and Toll (xb) expression levels were significantly correlated with DENV2 load (y, lg[DENV2 RNA copies]), y = -3436xa+0.2287xb+3.8194 (adjusted R2 = 0.5563, F = 9.148, PF = 0.0045). In midguts, DENV2 load was significantly correlated with the relative Fba(R2 = 0.4381, t = 2.497, p < 0.05, df = 8), UcCr(R2 = 0.4072, t = 2.344, p < 0.05, df = 8) and Gbps1(R2 = 0.4678, t = 2.652, p < 0.05, df = 8) expression levels, but multiple regression did not yield significant results. This study shows that genes related to glucose metabolism and muscle proteins contribute to the interaction between Aedes aegypti and dengue virus. It was confirmed that SAAG-4, histone H4, endoplasmin, catalase and other genes are involved in the regulation of DENV2 infection in Aedes aegypti. It was revealed that GST and Toll in salivary glands may have antagonistic effects on the regulation of DENV2 load. Fba, UcCr and Gbps1 in the midgut may increase DENV2 load. These study results further condensed the potential target gene range of the Aedes aegypti immune mechanism against DENV2 infection and provided basic information for research on the Aedes aegypti in vivo blockade strategy against DENV2.
Collapse
|
6
|
Vander Does A, Labib A, Yosipovitch G. Update on mosquito bite reaction: Itch and hypersensitivity, pathophysiology, prevention, and treatment. Front Immunol 2022; 13:1024559. [PMID: 36211437 PMCID: PMC9532860 DOI: 10.3389/fimmu.2022.1024559] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 09/07/2022] [Indexed: 11/26/2022] Open
Abstract
Mosquito bites are endured by most populations worldwide. Reactions to mosquito bites range from localized wheals and papules with associated pruritus to rare systemic reactions and anaphylaxis in certain populations. The mechanism of itch is due to introduction of mosquito saliva components into the cutaneous tissue, although the exact pathophysiology is unclear. Histamine is thought to be a key player through mosquito saliva itself or through activation of mast cells by IgE or through an IgE-independent pathway. However, other salivary proteins such as tryptase and leukotrienes may induce non-histaminergic itch. Some individuals have a genetic predisposition for mosquito bites, and people with hematologic cancers, HIV, and other conditions are susceptible to robust reactions. Prevention of mosquito bites is key with physical barriers or chemical repellents. Treatment consists of second-generation antihistamines and topical corticosteroids. Further research on topical treatments that target neural-mediated itch is needed.
Collapse
|
7
|
Rajendran KV, Neelakanta G, Sultana H. Sphingomyelinases in a journey to combat arthropod-borne pathogen transmission. FEBS Lett 2021; 595:1622-1638. [PMID: 33960414 DOI: 10.1002/1873-3468.14103] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 12/14/2022]
Abstract
Ixodes scapularis ticks feed on humans and other vertebrate hosts and transmit several pathogens of public health concern. Tick saliva is a complex mixture of bioactive proteins, lipids and immunomodulators, such as I. scapularis sphingomyelinase (IsSMase)-like protein, an ortholog of dermonecrotoxin SMase D found in the venom of Loxosceles spp. of spiders. IsSMase modulates the host immune response towards Th2, which suppresses Th1-mediated cytokines to facilitate pathogen transmission. Arboviruses utilize exosomes for their transmission from tick to the vertebrate host, and exosomes derived from tick saliva/salivary glands suppress C-X-C motif chemokine ligand 12 and interleukin-8 immune response(s) in human skin to delay wound healing and repair processes. IsSMase affects also viral replication and exosome biogenesis, thereby inhibiting tick-to-vertebrate host transmission of pathogenic exosomes. In this review, we elaborate on exosomes and their biogenesis as potential candidates for developing novel control measure(s) to combat tick-borne diseases. Such targets could help with the development of an efficient anti-tick vaccine for preventing the transmission of tick-borne pathogens.
Collapse
Affiliation(s)
- Kundave V Rajendran
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA
| | - Girish Neelakanta
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA.,Center for Molecular Medicine, Old Dominion University, Norfolk, VA, USA
| | - Hameeda Sultana
- Department of Biological Sciences, Old Dominion University, Norfolk, VA, USA.,Center for Molecular Medicine, Old Dominion University, Norfolk, VA, USA.,Department of Medicine, Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, VA, USA
| |
Collapse
|
8
|
Assis JB, Cogliati B, Esteves E, Capurro ML, Fonseca DM, Sá-Nunes A. Aedes aegypti mosquito saliva ameliorates acetaminophen-induced liver injury in mice. PLoS One 2021; 16:e0245788. [PMID: 33556084 PMCID: PMC7869984 DOI: 10.1371/journal.pone.0245788] [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: 05/08/2020] [Accepted: 01/07/2021] [Indexed: 02/07/2023] Open
Abstract
Acetaminophen (N-acetyl-p-aminophenol, APAP) overdose is the most common cause of drug-induced liver injury (DILI). Although the primary hepatic damage is induced by APAP-derived toxic intermediates resulting from cytochrome P450 metabolism, immune components also play an important role in DILI pathophysiology. Aedes aegypti saliva is a source of bioactive molecules with in vitro anti-inflammatory and immunomodulatory activities. However, evidences on the therapeutic use of Ae. aegypti salivary preparations in animal models of relevant clinical conditions are still scarce. Thus, the present study was designed to evaluate the protective role of Ae. aegypti saliva in a murine model of APAP-induced DILI. C57BL/6 mice were exposed to Ae. aegypti bites 2 hours after APAP overdose. Biochemical and immunological parameters were evaluated in blood and liver samples at different time points after APAP administration. Exposure to Ae. aegypti saliva attenuated liver damage, as demonstrated by reduced hepatic necrosis and serum levels of alanine aminotransferase in APAP-overdosed mice. The levels of hepatic CYP2E1, the major enzyme responsible for the bioactivation of APAP, were not changed in Ae. aegypti exposed animals, suggesting no effects in the generation of hepatotoxic metabolites. On the other hand, mice treated with Ae. aegypti saliva following APAP overdose presented lower serum concentration of TNF-α, IL-6, IL-1β and IL-10, as well as reduced frequency of inflammatory cell populations in the liver, such as NKT cells, macrophages and dendritic cells. These findings show that Ae. aegypti saliva has bioactive molecules with therapeutic properties and may represent a prospective source of new compounds in the management of DILI-associated inflammatory disorders and, perhaps, many other inflammatory/autoimmune diseases.
Collapse
Affiliation(s)
- Josiane B. Assis
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Bruno Cogliati
- Departamento de Patologia, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo, Brazil
| | - Eliane Esteves
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Margareth L. Capurro
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Conselho Nacional de Desenvolvimento Científico e Tecnológico (INCT-EM/CNPq), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Denise M. Fonseca
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Anderson Sá-Nunes
- Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Conselho Nacional de Desenvolvimento Científico e Tecnológico (INCT-EM/CNPq), Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
9
|
Guerrero D, Cantaert T, Missé D. Aedes Mosquito Salivary Components and Their Effect on the Immune Response to Arboviruses. Front Cell Infect Microbiol 2020; 10:407. [PMID: 32850501 PMCID: PMC7426362 DOI: 10.3389/fcimb.2020.00407] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 06/30/2020] [Indexed: 12/25/2022] Open
Abstract
Vector-borne diseases are responsible for over a billion infections each year and nearly one million deaths. Mosquito-borne dengue virus, West Nile, Japanese encephalitis, Zika, Chikungunya, and Rift Valley Fever viruses constitute major public health problems in regions with high densities of arthropod vectors. During the initial step of the transmission cycle, vector, host, and virus converge at the bite site, where local immune cells interact with the vector's saliva. Hematophagous mosquito saliva is a mixture of bioactive components known to modulate vertebrate hemostasis, immunity, and inflammation during the insect's feeding process. The capacity of mosquito saliva to modulate the host immune response has been well-studied over the last few decades and has led to the consensus that the presence of saliva is linked to the enhancement of virus transmission, host susceptibility, disease progression, viremia levels, and mortality. We review some of the major aspects of the interactions between mosquito saliva and the host immune response that may be useful for future studies on the control of arboviruses.
Collapse
Affiliation(s)
- David Guerrero
- Immunology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| | - Tineke Cantaert
- Immunology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh, Cambodia
| | - Dorothée Missé
- MIVEGEC, IRD, University of Montpellier, CNRS, Montpellier, France
| |
Collapse
|
10
|
Barros MS, Lara PG, Fonseca MT, Moretti EH, Filgueiras LR, Martins JO, Capurro ML, Steiner AA, Sá-Nunes A. Aedes aegypti saliva impairs M1-associated proinflammatory phenotype without promoting or affecting M2 polarization of murine macrophages. Parasit Vectors 2019; 12:239. [PMID: 31097013 PMCID: PMC6524299 DOI: 10.1186/s13071-019-3487-7] [Citation(s) in RCA: 5] [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: 12/07/2018] [Accepted: 05/06/2019] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND During the feeding process, the mouthparts of hematophagous mosquitoes break the skin barrier and probe the host tissue to find the blood. The saliva inoculated in this microenvironment modulates host hemostasis, inflammation and adaptive immune responses. However, the mechanisms involved in these biological activities remain poorly understood and few studies explored the potential roles of mosquito saliva on the individual cellular components of the immune system. Here, we report the immunomodulatory activities of Aedes aegypti salivary cocktail on murine peritoneal macrophages. RESULTS The salivary gland extract (SGE) of Ae. aegypti inhibited the production of nitric oxide and inflammatory cytokines such as interleukin-6 (IL-6) and IL-12, as well as the expression of inducible nitric oxide synthase and NF-κB by murine macrophages stimulated by lipopolysaccharide (LPS) plus interferon-γ (IFN-γ). The spare respiratory capacity, the phagocytic and microbicidal activities of these macrophages were also reduced by Ae. aegypti SGE. These phenotypic changes are consistent with SGE suppressing the proinflammatory program of M1 macrophages. On the other hand, Ae. aegypti SGE did not influence M2-associated markers (urea production, arginase-1 and mannose receptor-1 expression), either in macrophages alternatively activated by IL-4 or in those classically activated by LPS plus IFN-γ. In addition, Ae. aegypti SGE did not display any cytokine-binding activity, nor did it affect macrophage viability, thus excluding supposed experimental artifacts. CONCLUSIONS Given the importance of macrophages in a number of biological processes, our findings help to enlighten how vector saliva modulates vertebrate host immunity.
Collapse
Affiliation(s)
- Michele S Barros
- Laboratory of Experimental Immunology, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil
| | - Priscila G Lara
- Laboratory of Experimental Immunology, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil
| | - Monique T Fonseca
- Laboratory of Sepsis Neurobiology, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil
| | - Eduardo H Moretti
- Laboratory of Sepsis Neurobiology, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil
| | - Luciano R Filgueiras
- Laboratory of Immunopharmacology, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil
| | - Joilson O Martins
- Laboratory of Immunoendocrinology, Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil
| | - Margareth L Capurro
- Laboratory of Genetically Modified Mosquitoes, Department of Parasitology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil
- National Institute of Science and Technology on Molecular Entomology, National Council for Scientific and Technological Development (INCT-EM/CNPq), Rio de Janeiro, RJ, Brazil
| | - Alexandre A Steiner
- Laboratory of Sepsis Neurobiology, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil
| | - Anderson Sá-Nunes
- Laboratory of Experimental Immunology, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, 05508-000, Brazil.
- National Institute of Science and Technology on Molecular Entomology, National Council for Scientific and Technological Development (INCT-EM/CNPq), Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
11
|
Kotál J, Stergiou N, Buša M, Chlastáková A, Beránková Z, Řezáčová P, Langhansová H, Schwarz A, Calvo E, Kopecký J, Mareš M, Schmitt E, Chmelař J, Kotsyfakis M. The structure and function of Iristatin, a novel immunosuppressive tick salivary cystatin. Cell Mol Life Sci 2019; 76:2003-2013. [PMID: 30747251 PMCID: PMC11105445 DOI: 10.1007/s00018-019-03034-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/14/2019] [Accepted: 01/28/2019] [Indexed: 12/31/2022]
Abstract
To successfully feed, ticks inject pharmacoactive molecules into the vertebrate host including cystatin cysteine protease inhibitors. However, the molecular and cellular events modulated by tick saliva remain largely unknown. Here, we describe and characterize a novel immunomodulatory cystatin, Iristatin, which is upregulated in the salivary glands of feeding Ixodes ricinus ticks. We present the crystal structure of Iristatin at 1.76 Å resolution. Purified recombinant Iristatin inhibited the proteolytic activity of cathepsins L and C and diminished IL-2, IL-4, IL-9, and IFN-γ production by different T-cell populations, IL-6 and IL-9 production by mast cells, and nitric oxide production by macrophages. Furthermore, Iristatin inhibited OVA antigen-induced CD4+ T-cell proliferation and leukocyte recruitment in vivo and in vitro. Our results indicate that Iristatin affects wide range of anti-tick immune responses in the vertebrate host and may be exploitable as an immunotherapeutic.
Collapse
Affiliation(s)
- Jan Kotál
- Laboratory of Genomics and Proteomics of Disease Vectors, Biology Centre CAS, Institute of Parasitology, Branišovská 1160/31, 37005, České Budějovice, Czech Republic
- Department of Medical Biology, Faculty of Science, University of South Bohemia in České Budějovice, Branišovská 1760c, 37005, České Budějovice, Czech Republic
| | - Natascha Stergiou
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, Mainz, 55131, Germany
| | - Michal Buša
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Flemingovo nám. 2, 16610, Prague, Czech Republic
| | - Adéla Chlastáková
- Department of Medical Biology, Faculty of Science, University of South Bohemia in České Budějovice, Branišovská 1760c, 37005, České Budějovice, Czech Republic
| | - Zuzana Beránková
- Department of Medical Biology, Faculty of Science, University of South Bohemia in České Budějovice, Branišovská 1760c, 37005, České Budějovice, Czech Republic
| | - Pavlína Řezáčová
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Flemingovo nám. 2, 16610, Prague, Czech Republic
| | - Helena Langhansová
- Department of Medical Biology, Faculty of Science, University of South Bohemia in České Budějovice, Branišovská 1760c, 37005, České Budějovice, Czech Republic
| | - Alexandra Schwarz
- Laboratory of Genomics and Proteomics of Disease Vectors, Biology Centre CAS, Institute of Parasitology, Branišovská 1160/31, 37005, České Budějovice, Czech Republic
| | - Eric Calvo
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, MD, 20852, USA
| | - Jan Kopecký
- Department of Medical Biology, Faculty of Science, University of South Bohemia in České Budějovice, Branišovská 1760c, 37005, České Budějovice, Czech Republic
| | - Michael Mareš
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Flemingovo nám. 2, 16610, Prague, Czech Republic
| | - Edgar Schmitt
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, Mainz, 55131, Germany
| | - Jindřich Chmelař
- Department of Medical Biology, Faculty of Science, University of South Bohemia in České Budějovice, Branišovská 1760c, 37005, České Budějovice, Czech Republic
| | - Michail Kotsyfakis
- Laboratory of Genomics and Proteomics of Disease Vectors, Biology Centre CAS, Institute of Parasitology, Branišovská 1160/31, 37005, České Budějovice, Czech Republic.
| |
Collapse
|
12
|
Monteiro VVS, Navegantes-Lima KC, de Lemos AB, da Silva GL, de Souza Gomes R, Reis JF, Rodrigues Junior LC, da Silva OS, Romão PRT, Monteiro MC. Aedes-Chikungunya Virus Interaction: Key Role of Vector Midguts Microbiota and Its Saliva in the Host Infection. Front Microbiol 2019; 10:492. [PMID: 31024463 PMCID: PMC6467098 DOI: 10.3389/fmicb.2019.00492] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 02/26/2019] [Indexed: 01/02/2023] Open
Abstract
Aedes mosquitoes are important vectors for emerging diseases caused by arboviruses, such as chikungunya (CHIKV). These viruses’ main transmitting species are Aedes aegypti and Ae. albopictus, which are present in tropical and temperate climatic areas all over the globe. Knowledge of vector characteristics is fundamentally important to the understanding of virus transmission. Only female mosquitoes are able to transmit CHIKV to the vertebrate host since they are hematophagous. In addition, mosquito microbiota is fundamentally important to virus infection in the mosquito. Microorganisms are able to modulate viral transmission in the mosquito, such as bacteria of the Wolbachia genus, which are capable of preventing viral infection, or protozoans of the Ascogregarina species, which are capable of facilitating virus transmission between mosquitoes and larvae. The competence of the mosquito is also important in the transmission of the virus to the vertebrate host, since their saliva has several substances with biological effects, such as immunomodulators and anticoagulants, which are able to modulate the host’s response to the virus, interfering in its pathogenicity and virulence. Understanding the Aedes vector-chikungunya interaction is fundamentally important since it can enable the search for new methods of combating the virus’ transmission.
Collapse
Affiliation(s)
- Valter Vinícius Silva Monteiro
- Laboratory of Inflammation and Pain, Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Kely Campos Navegantes-Lima
- Graduate Program in Neuroscience and Cellular Biology, Biology Science Institute, Federal University of Pará, Belém, Brazil
| | | | | | - Rafaelli de Souza Gomes
- Graduate Program in Pharmaceutical Science, Health Science Institute, Federal University of Pará, Belém, Brazil
| | - Jordano Ferreira Reis
- School of Pharmacy, Health Science Institute, Federal University of Pará, Belém, Brazil
| | - Luiz Carlos Rodrigues Junior
- Laboratory of Cellular and Molecular Immunology, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil
| | - Onilda Santos da Silva
- Department of Microbiology, Immunology and Parasitology, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Pedro Roosevelt Torres Romão
- Laboratory of Cellular and Molecular Immunology, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil
| | - Marta Chagas Monteiro
- Graduate Program in Neuroscience and Cellular Biology, Biology Science Institute, Federal University of Pará, Belém, Brazil.,Graduate Program in Pharmaceutical Science, Health Science Institute, Federal University of Pará, Belém, Brazil
| |
Collapse
|
13
|
|
14
|
Islam A, Emran TB, Yamamoto DS, Iyori M, Amelia F, Yusuf Y, Yamaguchi R, Alam MS, Silveira H, Yoshida S. Anopheline antiplatelet protein from mosquito saliva regulates blood feeding behavior. Sci Rep 2019; 9:3129. [PMID: 30816309 PMCID: PMC6395645 DOI: 10.1038/s41598-019-39960-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 02/01/2019] [Indexed: 01/23/2023] Open
Abstract
The saliva of hematophagous arthropods is enriched with a complex mixture of antihemostatic molecules, the biological functions of which are largely unknown. Anopheline antiplatelet protein (AAPP) from malaria vector mosquito exhibits strong antiplatelet activity when bound directly to host collagen by its C-terminus and through its N-terminus with Ca2+-binding activity. To investigate the biological functions of AAPP in blood feeding behavior and malaria transmission, we generated transgenic Anopheles stephensi mosquito lines expressing anti-AAPP antibody single-chain fragment (scFv) in their salivary glands. The AAPP-specific collagen-binding activity was completely abolished by AAPP-scFv complex formation in the saliva. Probing and prediuresis time, feeding success, blood meal size, and fecundity, which are all fitness characteristics, were significantly reduced in the transgenic mosquitoes. However, oocysts number in these mosquitoes were not significantly reduced following blood meal intake from Plasmodium berghei-infected mice. These results show that although AAPP plays an important role in mosquito blood feeding, its neutralizing activity did not affect sporogonic development in our laboratory model, but its high fitness cost would pose a survival risk for parasite-infected mosquitoes in nature.
Collapse
Affiliation(s)
- Ashekul Islam
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Talha Bin Emran
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Daisuke S Yamamoto
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, 329-0431, Japan
| | - Mitsuhiro Iyori
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Fitri Amelia
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Yenni Yusuf
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Ririka Yamaguchi
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Md Shah Alam
- Laboratory of Ecology, Graduate School of Natural Science and Technology, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Henrique Silveira
- Laboratory of Vector-borne diseases and Pathogens, Global Health and Tropical Medicine, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Lisbon, 1099-085, Portugal
| | - Shigeto Yoshida
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kakuma-machi, Kanazawa, 920-1192, Japan.
| |
Collapse
|
15
|
Wanelik KM, Begon M, Birtles RJ, Bradley JE, Friberg IM, Jackson JA, Taylor CH, Thomason AG, Turner AK, Paterson S. A candidate tolerance gene identified in a natural population of field voles (Microtus agrestis). Mol Ecol 2018; 27:1044-1052. [PMID: 29290094 DOI: 10.1111/mec.14476] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 11/24/2017] [Accepted: 11/30/2017] [Indexed: 01/27/2023]
Abstract
The animal immune response has hitherto been viewed primarily in the context of resistance only. However, individuals can also employ a tolerance strategy to maintain good health in the face of ongoing infection. To shed light on the genetic and physiological basis of tolerance, we use a natural population of field voles, Microtus agrestis, to search for an association between the expression of the transcription factor Gata3, previously identified as a marker of tolerance in this system, and polymorphism in 84 immune and nonimmune genes. Our results show clear evidence for an association between Gata3 expression and polymorphism in the Fcer1a gene, with the explanatory power of this polymorphism being comparable to that of other nongenetic variables previously identified as important predictors of Gata3 expression. We also uncover the possible mechanism behind this association using an existing protein-protein interaction network for the mouse model rodent, Mus musculus, which we validate using our own expression network for M. agrestis. Our results suggest that the polymorphism in question may be working at the transcriptional level, leading to changes in the expression of the Th2-related genes, Tyrosine-protein kinase BTK and Tyrosine-protein kinase TXK, and hence potentially altering the strength of the Th2 response, of which Gata3 is a mediator. We believe our work has implications for both treatment and control of infectious disease.
Collapse
Affiliation(s)
- Klara M Wanelik
- Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Michael Begon
- Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Richard J Birtles
- School of Environment and Life Sciences, University of Salford, Salford, UK
| | | | - Ida M Friberg
- School of Environment and Life Sciences, University of Salford, Salford, UK
| | - Joseph A Jackson
- School of Environment and Life Sciences, University of Salford, Salford, UK
| | | | - Anna G Thomason
- School of Environment and Life Sciences, University of Salford, Salford, UK
| | - Andrew K Turner
- Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Steve Paterson
- Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| |
Collapse
|
16
|
Yang S, Fink D, Hulse A, Pratt RD. Regulatory considerations in development of vaccines to prevent disease caused by Chikungunya virus. Vaccine 2017; 35:4851-4858. [PMID: 28760614 DOI: 10.1016/j.vaccine.2017.07.065] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 06/21/2017] [Accepted: 07/19/2017] [Indexed: 12/01/2022]
Abstract
Chikungunya virus (CHIKV) is a mosquito-transmitted alphavirus. Chikungunya disease (CHIK) in humans is characterized by sudden onset of high fever, cutaneous rash, myalgia and debilitating polyarthralgia. Until recently the virus was considered endemic to only Africa and Asia, but since 2004 CHIK has spread to previously non-endemic regions, including Europe and the Americas, thereby emerging as a global health threat. Although a variety of CHIKV vaccine candidates have been tested in animals, and a few have advanced to human clinical trials, no licensed vaccine is currently available for prevention of disease. In this article, we review recent efforts in CHIKV vaccine development and discuss regulatory considerations for CHIKV vaccine licensure under U.S. FDA regulations. Several licensure pathways are available, and the most appropriate licensure pathway for a CHIK vaccine will depend on the type of evidence that can be generated to demonstrate the vaccine's effectiveness. If "traditional approval" following demonstration of direct benefit in adequate and well-controlled clinical disease endpoint studies is not possible, the Accelerated Approval and Animal Rule pathways are potential alternatives. In terms of vaccine safety, the potential for vaccine associated arthralgia and antibody-dependent enhancement of infectivity and disease severity are important issues that should be addressed in both pre-clinical and clinical studies. CHIK vaccine developers are encouraged to communicate with the FDA during all stages of vaccine development.
Collapse
Affiliation(s)
- Sixun Yang
- Division of Vaccines and Related Product Applications, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research (CBER), U.S. Food and Drug Administration (FDA), 10903 New Hampshire Avenue, Silver Spring, MD 20993, United States.
| | - Doran Fink
- Division of Vaccines and Related Product Applications, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research (CBER), U.S. Food and Drug Administration (FDA), 10903 New Hampshire Avenue, Silver Spring, MD 20993, United States
| | - Andrea Hulse
- Division of Vaccines and Related Product Applications, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research (CBER), U.S. Food and Drug Administration (FDA), 10903 New Hampshire Avenue, Silver Spring, MD 20993, United States
| | - R Douglas Pratt
- Division of Vaccines and Related Product Applications, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research (CBER), U.S. Food and Drug Administration (FDA), 10903 New Hampshire Avenue, Silver Spring, MD 20993, United States
| |
Collapse
|
17
|
Jones AV, Tilley M, Gutteridge A, Hyde C, Nagle M, Ziemek D, Gorman D, Fauman EB, Chen X, Miller MR, Tian C, Hu Y, Hinds DA, Cox P, Scollen S. GWAS of self-reported mosquito bite size, itch intensity and attractiveness to mosquitoes implicates immune-related predisposition loci. Hum Mol Genet 2017; 26:1391-1406. [PMID: 28199695 PMCID: PMC5390679 DOI: 10.1093/hmg/ddx036] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 01/23/2017] [Indexed: 12/31/2022] Open
Abstract
Understanding the interaction between humans and mosquitoes is a critical area of study due to the phenomenal burdens on public health from mosquito-transmitted diseases. In this study, we conducted the first genome-wide association studies (GWAS) of self-reported mosquito bite reaction size (n = 84,724), itchiness caused by bites (n = 69,057), and perceived attractiveness to mosquitoes (n = 16,576). In total, 15 independent significant (P < 5×10−8) associations were identified. These loci were enriched for immunity-related genes that are involved in multiple cytokine signalling pathways. We also detected suggestive enrichment of these loci in enhancer regions that are active in stimulated T-cells, as well as within loci previously identified as controlling central memory T-cell levels. Egger regression analysis between the traits suggests that perception of itchiness and attractiveness to mosquitoes is driven, at least in part, by the genetic determinants of bite reaction size. Our findings illustrate the complex genetic and immunological landscapes underpinning human interactions with mosquitoes.
Collapse
Affiliation(s)
- Amy V. Jones
- Pfizer WRD, Human Genetics and Computational Biomedicine, The Portway Building, Granta Park, Cambridge CB21 6GS, UK
| | - Mera Tilley
- Pfizer WRD, Pharmatherapeutics Clinical R&D, Precision Medicine, 300 Technology Square Fl #3, Cambridge, MA 02139, USA
| | - Alex Gutteridge
- Pfizer WRD, Computational Sciences CoE, The Portway Building, Granta Park, Cambridge CB21 6GS, UK
| | - Craig Hyde
- Pfizer WRD, Research Statistics, 558 Eastern Point Rd, Groton, CT 06340, USA
| | - Michael Nagle
- Pfizer WRD, Human Genetics and Computational Biomedicine, 610 Main Street S, Cambridge, MA 02139, USA
| | - Daniel Ziemek
- Pfizer WRD, Computational Sciences CoE, Linkstraße 10, 10785 Berlin, Germany
| | - Donal Gorman
- Pfizer WRD, Research Statistics, The Portway Building, Granta Park, Cambridge CB21 6GS, UK
| | - Eric B. Fauman
- Pfizer WRD, Computational Sciences CoE, 610 Main Street S, Cambridge, MA 02139, USA
| | - Xing Chen
- Pfizer WRD, Research Statistics, 558 Eastern Point Rd, Groton, CT 06340, USA
| | - Melissa R. Miller
- Pfizer WRD, Human Genetics and Computational Biomedicine, 610 Main Street S, Cambridge, MA 02139, USA
| | - Chao Tian
- 23andMe, Inc, 899 W Evelyn Avenue, Mountain View, California, CA 94043, USA
| | - Youna Hu
- 23andMe, Inc, 899 W Evelyn Avenue, Mountain View, California, CA 94043, USA
| | - David A. Hinds
- 23andMe, Inc, 899 W Evelyn Avenue, Mountain View, California, CA 94043, USA
| | - Peter Cox
- Pfizer Ltd, Neuroscience and Pain Research Unit, The Portway Building, Granta Park, Cambridge CB21 6GS, UK
| | - Serena Scollen
- Pfizer WRD, Human Genetics and Computational Biomedicine, The Portway Building, Granta Park, Cambridge CB21 6GS, UK
- To whom correspondence should be addressed at: ELIXIR Hub, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK. Tel: +44 1223494322; Fax: +44 (0)1223 484696;
| |
Collapse
|
18
|
Wichit S, Ferraris P, Choumet V, Missé D. The effects of mosquito saliva on dengue virus infectivity in humans. Curr Opin Virol 2016; 21:139-145. [PMID: 27770704 DOI: 10.1016/j.coviro.2016.10.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 10/04/2016] [Accepted: 10/06/2016] [Indexed: 12/14/2022]
Abstract
Arboviruses such as Dengue, Chikungunya, and Zika viruses represent a major public health problem due to globalization and propagation of susceptible vectors worldwide. Arthropod vector-derived salivary factors have the capacity to modulate human cells function by enhancing or suppressing viral replication and, therefore, modify the establishment of local and systemic viral infection. Here, we discuss how mosquito saliva may interfere with Dengue virus (DENV) infection in humans. Identification of saliva factors that enhance infectivity will allow the production of vector-based vaccines and therapeutics that would interfere with viral transmission by targeting arthropod saliva components. Understanding the role of salivary proteins in DENV transmission will provide tools to control not only Dengue but also other arboviral diseases transmitted by the same vectors.
Collapse
Affiliation(s)
| | - Pauline Ferraris
- Laboratory of MIVEGEC, UMR 224 IRD/CNRS/UM1, Montpellier, France
| | - Valérie Choumet
- Environment and Infectious Risks Unit, Pasteur Institute, Paris, France
| | - Dorothée Missé
- Laboratory of MIVEGEC, UMR 224 IRD/CNRS/UM1, Montpellier, France.
| |
Collapse
|
19
|
Londono-Renteria B, Cardenas JC, Troupin A, Colpitts TM. Natural Mosquito-Pathogen Hybrid IgG4 Antibodies in Vector-Borne Diseases: A Hypothesis. Front Immunol 2016; 7:380. [PMID: 27746778 PMCID: PMC5040711 DOI: 10.3389/fimmu.2016.00380] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 09/08/2016] [Indexed: 12/24/2022] Open
Abstract
Chronic exposure to antigens may favor the production of IgG4 antibodies over other antibody types. Recent studies have shown that up to a 30% of normal human IgG4 is bi-specific and is able to recognize two antigens of different nature. A requirement for this specificity is the presence of both eliciting antigens in the same time and at the same place where the immune response is induced. During transmission of most vector-borne diseases, the pathogen is delivered to the vertebrate host along with the arthropod saliva during blood feeding and previous studies have shown the existence of IgG4 antibodies against mosquito salivary allergens. However, there is very little ongoing research or information available regarding IgG4 bi-specificity with regard to infectious disease, particularly during immune responses to vector-borne diseases, such as malaria, filariasis, or dengue virus infection. Here, we provide background information and present our hypothesis that IgG4 may not only be a useful tool to measure exposure to infected mosquito bites, but that these bi-specific antibodies may also play an important role in modulation of the immune response against malaria and other vector-borne diseases in endemic settings.
Collapse
Affiliation(s)
- Berlin Londono-Renteria
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine , Columbia, SC , USA
| | - Jenny C Cardenas
- Clinical Laboratory, Hospital Los Patios , Los Patios , Colombia
| | - Andrea Troupin
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine , Columbia, SC , USA
| | - Tonya M Colpitts
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine , Columbia, SC , USA
| |
Collapse
|
20
|
Jackson JA. Immunology in wild nonmodel rodents: an ecological context for studies of health and disease. Parasite Immunol 2015; 37:220-32. [PMID: 25689683 PMCID: PMC7167918 DOI: 10.1111/pim.12180] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 02/04/2015] [Indexed: 12/16/2022]
Abstract
Transcriptomic methods are set to revolutionize the study of the immune system in naturally occurring nonmodel organisms. With this in mind, the present article focuses on ways in which the use of 'nonmodel' rodents (not the familiar laboratory species) can advance studies into the classical, but ever relevant, epidemiologic triad of immune defence, infectious disease and environment. For example, naturally occurring rodents are an interesting system in which to study the environmental stimuli that drive the development and homeostasis of the immune system and, by extension, to identify where these stimuli are altered in anthropogenic environments leading to the formation of immunopathological phenotypes. Measurement of immune expression may help define individual heterogeneity in infectious disease susceptibility and transmission and facilitate our understanding of infection dynamics and risk in the natural environment; furthermore, it may provide a means of surveillance that can filter individuals carrying previously unknown acute infections of potential ecological or zoonotic importance. Finally, the study of immunology in wild animals may reveal interactions within the immune system and between immunity and other organismal traits that are not observable under restricted laboratory conditions. Potentiating much of this is the possibility of combining gene expression profiles with analytical tools derived from ecology and systems biology to reverse engineer interaction networks between immune responses, other organismal traits and the environment (including symbiont exposures), revealing regulatory architecture. Such holistic studies promise to link ecology, epidemiology and immunology in natural systems in a unified approach that can illuminate important problems relevant to human health and animal welfare and production.
Collapse
Affiliation(s)
- J A Jackson
- IBERS, Aberystwyth University, Aberystwyth, Ceredigion, UK
| |
Collapse
|
21
|
Rizzo C, Lombardo F, Ronca R, Mangano V, Sirima SB, Nèbiè I, Fiorentino G, Modiano D, Arcà B. Differential antibody response to the Anopheles gambiae gSG6 and cE5 salivary proteins in individuals naturally exposed to bites of malaria vectors. Parasit Vectors 2014; 7:549. [PMID: 25428638 PMCID: PMC4253619 DOI: 10.1186/s13071-014-0549-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 11/20/2014] [Indexed: 12/05/2022] Open
Abstract
Background Mosquito saliva plays crucial roles in blood feeding but also evokes in hosts an anti-saliva antibody response. The IgG response to the Anopheles gambiae salivary protein gSG6 was previously shown to be a reliable indicator of human exposure to Afrotropical malaria vectors. We analyzed here the humoral response to the salivary anti-thrombin cE5 in a group of individuals from a malaria hyperendemic area of Burkina Faso. Methods ELISA was used to measure the anti-cE5 IgG, IgG1 and IgG4 antibody levels in plasma samples collected in the village of Barkoumbilen (Burkina Faso) among individuals of the Rimaibé ethnic group. Anti-gSG6 IgG levels were also determined for comparison. Anopheles vector density in the study area was evaluated by indoor pyrethrum spray catches. Results The cE5 protein was highly immunogenic and triggered in exposed individuals a relatively long-lasting antibody response, as shown by its unchanged persistence after a few months of absent or very low exposure (dry season). In addition cE5 did not induce immune tolerance, as previously suggested for the gSG6 antigen. Finally, IgG subclass analysis suggested that exposed individuals may mount a Th1-type immune response against the cE5 protein. Conclusions The anti-cE5 IgG response is shown here to be a sensitive indicator of human exposure to anopheline vectors and to represent an additional tool for malaria epidemiological studies. It may be especially useful in conditions of low vector density, to monitor transiently exposed individuals (i.e. travellers/workers/soldiers spending a few months in tropical Africa) and to evaluate the impact of insecticide treated nets on vector control. Moreover, the gSG6 and cE5 salivary proteins were shown to trigger in exposed individuals a strikingly different immune response with (i) gSG6 evoking a short-lived IgG response, characterized by high IgG4 levels and most likely induction of immune tolerance, and (ii) cE5 eliciting a longer-living IgG response, dominated by anti-cE5 IgG1 antibodies and not inducing tolerance mechanisms. We believe that these two antigens may represent useful reagents to further investigate the so far overlooked role of Anopheles saliva and salivary proteins in host early immune response to Plasmodium parasites. Electronic supplementary material The online version of this article (doi:10.1186/s13071-014-0549-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Cinzia Rizzo
- Department of Public Health and Infectious Diseases, Division of Parasitology, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy.
| | - Fabrizio Lombardo
- Department of Public Health and Infectious Diseases, Division of Parasitology, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy.
| | - Raffaele Ronca
- Department of Biology, "Federico II" University, Naples, Italy.
| | - Valentina Mangano
- Department of Public Health and Infectious Diseases, Division of Parasitology, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy.
| | | | - Issa Nèbiè
- Centre National de Recherche et de Formation sur le Paludisme, Ouagadougou, Burkina Faso.
| | | | - David Modiano
- Department of Public Health and Infectious Diseases, Division of Parasitology, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy.
| | - Bruno Arcà
- Department of Public Health and Infectious Diseases, Division of Parasitology, Sapienza University, Piazzale Aldo Moro 5, 00185, Rome, Italy.
| |
Collapse
|
22
|
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
|
23
|
Aedes aegypti salivary protein "aegyptin" co-inoculation modulates dengue virus infection in the vertebrate host. Virology 2014; 468-470:133-139. [PMID: 25173089 DOI: 10.1016/j.virol.2014.07.019] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 07/04/2014] [Accepted: 07/11/2014] [Indexed: 01/13/2023]
Abstract
Dengue virus (DENV) is transmitted in the saliva of the mosquito vector Aedes aegypti during blood meal acquisition. This saliva is composed of numerous proteins with the capacity to disrupt hemostasis or modulate the vertebrate immune response. One such protein, termed "aegyptin," is an allergen and inhibitor of clot formation, and has been found in decreased abundance in the saliva of DENV-infected mosquitoes. To examine the influence of aegyptin on DENV infection of the vertebrate, we inoculated IRF-3/7(-/- -/-) mice with DENV serotype 2 strain 1232 with and without co-inoculation of aegyptin. Mice that received aegyptin exhibited decreased DENV titers in inoculation sites and in circulation, as well as increased concentrations of GM-CSF, IFN-γ, IL-5, and IL-6, at 48 h post-inoculation when compared to mice that received inoculation of DENV alone. These and other data suggest that aegyptin impacts DENV perpetuation via elevated induction of the immune response.
Collapse
|
24
|
Role of skin immune cells on the host susceptibility to mosquito-borne viruses. Virology 2014; 464-465:26-32. [PMID: 25043586 DOI: 10.1016/j.virol.2014.06.023] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 05/15/2014] [Accepted: 06/17/2014] [Indexed: 12/13/2022]
Abstract
Due to climate change and the propagation of competent arthropods worldwide, arboviruses have become pathogens of major medical importance. Early transmission to vertebrates is initiated by skin puncture and deposition of virus together with arthropod saliva in the epidermis and dermis. Saliva components have the capacity to modulate skin cell responses by enhancing and/or counteracting initial replication and establishment of systemic viral infection. Here, we review the nature of the cells targeted by arboviruses at the skin level and discuss the type of cellular responses elicited by these pathogens in light of the immunomodulatory properties of arthropod vector-derived salivary factors injected at the inoculation site. Understanding cutaneous arbovirus-host interactions may provide new clues for the design of future therapeutics.
Collapse
|
25
|
Mores CN, Christofferson RC, Davidson SA. The role of the mosquito in a dengue human infection model. J Infect Dis 2014; 209 Suppl 2:S71-8. [PMID: 24872400 DOI: 10.1093/infdis/jiu110] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Recent efforts to combat the growing global threat of dengue disease, including deployment of phase IIb vaccine trials, has continued to be hindered by uncertainty surrounding equitable immune responses of serotypes, relative viral fitness of vaccine vs naturally occurring strains, and the importance of altered immune environments due to natural delivery routes. Human infection models can significantly improve our understanding of the importance of certain phenotypic characteristics of viral strains, and inform strain selection and trial design. With human models, we can further assess the importance of the natural delivery route of DENV and/or the accompanying mosquito salivary milieu. Accordingly, we discuss the use of mosquitoes in such a human infection model with DENV, identify important considerations, and make preliminary recommendations for deployment of such a mosquito improved DENV human infection model (miDHIM).
Collapse
Affiliation(s)
- Christopher N Mores
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Rebecca C Christofferson
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Silas A Davidson
- Department of Entomology, Walter Reed Army Institute of Research, Silver Spring, Maryland
| |
Collapse
|
26
|
Jackson JA, Hall AJ, Friberg IM, Ralli C, Lowe A, Zawadzka M, Turner AK, Stewart A, Birtles RJ, Paterson S, Bradley JE, Begon M. An immunological marker of tolerance to infection in wild rodents. PLoS Biol 2014; 12:e1001901. [PMID: 25004450 PMCID: PMC4086718 DOI: 10.1371/journal.pbio.1001901] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 05/29/2014] [Indexed: 02/02/2023] Open
Abstract
Hosts are likely to respond to parasitic infections by a combination of resistance (expulsion of pathogens) and tolerance (active mitigation of pathology). Of these strategies, the basis of tolerance in animal hosts is relatively poorly understood, with especially little known about how tolerance is manifested in natural populations. We monitored a natural population of field voles using longitudinal and cross-sectional sampling modes and taking measurements on body condition, infection, immune gene expression, and survival. Using analyses stratified by life history stage, we demonstrate a pattern of tolerance to macroparasites in mature compared to immature males. In comparison to immature males, mature males resisted infection less and instead increased investment in body condition in response to accumulating burdens, but at the expense of reduced reproductive effort. We identified expression of the transcription factor Gata3 (a mediator of Th2 immunity) as an immunological biomarker of this tolerance response. Time series data for individual animals suggested that macroparasite infections gave rise to increased expression of Gata3, which gave rise to improved body condition and enhanced survival as hosts aged. These findings provide a clear and unexpected insight into tolerance responses (and their life history sequelae) in a natural vertebrate population. The demonstration that such responses (potentially promoting parasite transmission) can move from resistance to tolerance through the course of an individual's lifetime emphasises the need to incorporate them into our understanding of the dynamics and risk of infection in the natural environment. Moreover, the identification of Gata3 as a marker of tolerance to macroparasites raises important new questions regarding the role of Th2 immunity and the mechanistic nature of the tolerance response itself. A more manipulative, experimental approach is likely to be valuable in elaborating this further.
Collapse
Affiliation(s)
| | - Amy J. Hall
- School of Life Sciences, The University of Nottingham, Nottingham, United Kingdom
| | - Ida M. Friberg
- School of Life Sciences, The University of Nottingham, Nottingham, United Kingdom
- Institute of Integrative Biology, The University of Liverpool, Liverpool, United Kingdom
| | - Catriona Ralli
- School of Life Sciences, The University of Nottingham, Nottingham, United Kingdom
| | - Ann Lowe
- School of Life Sciences, The University of Nottingham, Nottingham, United Kingdom
| | - Malgorzata Zawadzka
- Institute of Integrative Biology, The University of Liverpool, Liverpool, United Kingdom
| | - Andrew K. Turner
- Institute of Integrative Biology, The University of Liverpool, Liverpool, United Kingdom
| | | | - Richard J. Birtles
- School of Environment and Life Sciences, University of Salford, Salford, United Kingdom
| | - Steve Paterson
- Institute of Integrative Biology, The University of Liverpool, Liverpool, United Kingdom
| | - Janette E. Bradley
- School of Life Sciences, The University of Nottingham, Nottingham, United Kingdom
| | - Mike Begon
- Institute of Integrative Biology, The University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
27
|
Chisenhall DM, Christofferson RC, McCracken MK, Johnson AMF, Londono-Renteria B, Mores CN. Infection with dengue-2 virus alters proteins in naturally expectorated saliva of Aedes aegypti mosquitoes. Parasit Vectors 2014; 7:252. [PMID: 24886023 PMCID: PMC4057903 DOI: 10.1186/1756-3305-7-252] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 05/17/2014] [Indexed: 11/10/2022] Open
Abstract
Background Dengue virus (DENV) is responsible for up to approximately 300 million infections and an increasing number of deaths related to severe manifestations each year in affected countries throughout the tropics. It is critical to understand the drivers of this emergence, including the role of vector-virus interactions. When a DENV-infected Aedes aegypti mosquito bites a vertebrate, the virus is deposited along with a complex mixture of salivary proteins. However, the influence of a DENV infection upon the expectorated salivary proteome of its vector has yet to be determined. Methods Therefore, we conducted a proteomic analysis using 2-D gel electrophoresis coupled with mass spectrometry based protein identification comparing the naturally expectorated saliva of Aedes aegypti infected with DENV-2 relative to that of uninfected Aedes aegypti. Results Several proteins were found to be differentially expressed in the saliva of DENV-2 infected mosquitoes, in particular proteins with anti-hemostatic and pain inhibitory functions were significantly reduced. Hypothetical consequences of these particular protein reductions include increased biting rates and transmission success, and lead to alteration of transmission potential as calculated in our vectorial capacity model. Conclusions We present our characterizations of these changes with regards to viral transmission and mosquito blood-feeding success. Further, we conclude that our proteomic analysis of Aedes aegypti saliva altered by DENV infection provides a unique opportunity to identify pro-viral impacts key to virus transmission.
Collapse
Affiliation(s)
| | | | | | | | | | - Christopher N Mores
- Department of Pathobiological Sciences, Vector-borne Disease Laboratories, Louisiana State University, School of Veterinary Medicine, Baton Rouge, LA, USA.
| |
Collapse
|
28
|
Bizzarro B, Barros MS, Maciel C, Gueroni DI, Lino CN, Campopiano J, Kotsyfakis M, Amarante-Mendes GP, Calvo E, Capurro ML, Sá-Nunes A. Effects of Aedes aegypti salivary components on dendritic cell and lymphocyte biology. Parasit Vectors 2013; 6:329. [PMID: 24238038 PMCID: PMC3843549 DOI: 10.1186/1756-3305-6-329] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Accepted: 11/05/2013] [Indexed: 12/12/2022] Open
Abstract
Background Saliva is a key element of interaction between hematophagous mosquitoes and their vertebrate hosts. In addition to allowing a successful blood meal by neutralizing or delaying hemostatic responses, the salivary cocktail is also able to modulate the effector mechanisms of host immune responses facilitating, in turn, the transmission of several types of microorganisms. Understanding how the mosquito uses its salivary components to circumvent host immunity might help to clarify the mechanisms of transmission of such pathogens and disease establishment. Methods Flow cytometry was used to evaluate if increasing concentrations of A. aegypti salivary gland extract (SGE) affects bone marrow-derived DC differentiation and maturation. Lymphocyte proliferation in the presence of SGE was estimated by a colorimetric assay. Western blot and Annexin V staining assays were used to assess apoptosis in these cells. Naïve and memory cells from mosquito-bite exposed mice or OVA-immunized mice and their respective controls were analyzed by flow cytometry. Results Concentration-response curves were employed to evaluate A. aegypti SGE effects on DC and lymphocyte biology. DCs differentiation from bone marrow precursors, their maturation and function were not directly affected by A. aegypti SGE (concentrations ranging from 2.5 to 40 μg/mL). On the other hand, lymphocytes were very sensitive to the salivary components and died in the presence of A. aegypti SGE, even at concentrations as low as 0.1 μg/mL. In addition, A. aegypti SGE was shown to induce apoptosis in all lymphocyte populations evaluated (CD4+ and CD8+ T cells, and B cells) through a mechanism involving caspase-3 and caspase-8, but not Bim. By using different approaches to generate memory cells, we were able to verify that these cells are resistant to SGE effects. Conclusion Our results show that lymphocytes, and not DCs, are the primary target of A. aegypti salivary components. In the presence of A. aegypti SGE, naïve lymphocyte populations die by apoptosis in a caspase-3- and caspase-8-dependent pathway, while memory cells are selectively more resistant to its effects. The present work contributes to elucidate the activities of A. aegypti salivary molecules on the antigen presenting cell-lymphocyte axis and in the biology of these cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Anderson Sá-Nunes
- Laboratório de Imunologia Experimental, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, 05508-900, SP, Brazil.
| |
Collapse
|
29
|
Puiprom O, Morales Vargas RE, Potiwat R, Chaichana P, Ikuta K, Ramasoota P, Okabayashi T. Characterization of chikungunya virus infection of a human keratinocyte cell line: role of mosquito salivary gland protein in suppressing the host immune response. INFECTION GENETICS AND EVOLUTION 2013; 17:210-5. [PMID: 23583544 DOI: 10.1016/j.meegid.2013.04.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 03/14/2013] [Accepted: 04/03/2013] [Indexed: 10/26/2022]
Abstract
The chikungunya virus (CHIKV) is a mosquito-borne virus that has recently re-emerged in several countries. On infection, the first vertebrate cells to come into contact with CHIKV are skin cells; mosquitoes inoculate the virus together with salivary gland protein into host skin while probing and feeding on blood. However, there is little known about the susceptibility of human skin cells to CHIKV infection. To clarify this, we investigated the kinetics of CHIKV in the human keratinocyte cell line, HaCaT. CHIKV actively replicated in HaCaT cells, with virus titers in the supernatant increasing to 2.8 × 10(4) plaque-forming units (PFU) ml(-1) 24h post infection. CHIKV infection suppressed production of interleukin-8 (IL-8) in HaCaT cells. The function of IL-8 is to recruit immune cells to virus-infected sites, a process known as chemotaxis. Furthermore, we assessed the role of mosquito salivary gland protein in CHIKV infections by comparing the levels of CHIKV gene expression and chemokine production in HaCaT cells with and without salivary gland extract (SGE). SGE enhanced both the expression of the CHIKV gene and the suppression effect of CHIKV on IL-8 production. Our data suggest that the HaCaT cell line represents an effective tool for investigating the mechanism of CHIKV transmission and spread in skin cells. At the mosquito bite site, CHIKV works together with SGE to ensure the virus replicates in skin cells and escapes the host immune system by suppression of IL-8 production.
Collapse
Affiliation(s)
- Orapim Puiprom
- Mahidol-Osaka Center for Infectious Diseases, Faculty of Tropical Medicine, Mahidol University, Ratchathewi, Bangkok, Thailand
| | | | | | | | | | | | | |
Collapse
|
30
|
Fassbinder-Orth CA, Barak VA, Brown CR. Immune responses of a native and an invasive bird to Buggy Creek Virus (Togaviridae: Alphavirus) and its arthropod vector, the swallow bug (Oeciacus vicarius). PLoS One 2013; 8:e58045. [PMID: 23460922 PMCID: PMC3584039 DOI: 10.1371/journal.pone.0058045] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 01/29/2013] [Indexed: 02/06/2023] Open
Abstract
Invasive species often display different patterns of parasite burden and virulence compared to their native counterparts. These differences may be the result of variability in host-parasite co-evolutionary relationships, the occurrence of novel host-parasite encounters, or possibly innate differences in physiological responses to infection between invasive and native hosts. Here we examine the adaptive, humoral immune responses of a resistant, native bird and a susceptible, invasive bird to an arbovirus (Buggy Creek virus; Togaviridae: Alphavirus) and its ectoparasitic arthropod vector (the swallow bug; Oeciacus vicarius). Swallow bugs parasitize the native, colonially nesting cliff swallow (Petrochelidon pyrrhonota) and the introduced house sparrow (Passer domesticus) that occupies nests in cliff swallow colonies. We measured levels of BCRV-specific and swallow bug-specific IgY levels before nesting (prior to swallow bug exposure) and after nesting (after swallow bug exposure) in house sparrows and cliff swallows in western Nebraska. Levels of BCRV-specific IgY increased significantly following nesting in the house sparrow but not in the cliff swallow. Additionally, house sparrows displayed consistently higher levels of swallow bug-specific antibodies both before and after nesting compared to cliff swallows. The higher levels of BCRV and swallow bug specific antibodies detected in house sparrows may be reflective of significant differences in both antiviral and anti-ectoparasite immune responses that exist between these two avian species. To our knowledge, this is the first study to compare the macro- and microparasite-specific immune responses of an invasive and a native avian host exposed to the same parasites.
Collapse
|
31
|
Leitner WW, Costero-Saint Denis A, Wali T. Immunological consequences of arthropod vector-derived salivary factors. Eur J Immunol 2012; 41:3396-400. [PMID: 22125007 DOI: 10.1002/eji.201190075] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Diseases, such as malaria, dengue, leishmaniasis and tick-borne encephalitis, affect a substantial percentage of the world's population and continue to result in significant morbidity and mortality. One common aspect of these diseases is that the pathogens that cause them are transmitted by the bite of an infected arthropod (e.g. mosquito, sand fly, tick). The pathogens are delivered into the skin of the mammalian host along with arthropod saliva, which contains a wide variety of bioactive molecules. These saliva components are capable of altering hemostasis and immune responses and may contribute to the ability of the pathogen to establish an infection. The biological and immunological events that occur during pathogen transmission are poorly understood but may hold the key to novel approaches to prevent transmission and/or infection. In May 2011, the National Institute of Allergy and Infectious Diseases (NIAID) of the US National Institutes of Health (NIH) in the Department of Health and Human Services hosted a workshop entitled Immunological Consequences of Vector-Derived Factors which brought together experts in skin immunology, parasitology and vector biology to outline the gaps in our understanding of the process of pathogen transmission, to explore new approaches to control pathogen transmission, and to initiate and foster multidisciplinary collaborations among these investigators.
Collapse
Affiliation(s)
- Wolfgang W Leitner
- Division of Allergy, Immunology and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | |
Collapse
|
32
|
Liu S, Kelvin DJ, Leon AJ, Jin L, Farooqui A. Induction of Fas mediated caspase-8 independent apoptosis in immune cells by Armigeres subalbatus saliva. PLoS One 2012; 7:e41145. [PMID: 22815944 PMCID: PMC3398892 DOI: 10.1371/journal.pone.0041145] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 06/18/2012] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND It is widely recognized that the introduction of saliva of bloodsucking arthropods at the site of pathogen transmission might play a central role in vector-borne infections. However, how the interaction between salivary components and the host immune system takes place and which physiological processes this leads to has yet to be investigated. Armigeres subalbatus is one of the prominent types of mosquitoes involved in the transmission of parasitic and viral diseases in humans and animals. METHODOLOGY/PRINCIPAL FINDINGS Using murine peritoneal macrophages and lymphocytes, and human peripheral mononuclear cells (PBMCs), this study shows that saliva of the female Ar. subalbatus induces apoptosis via interaction with the Fas receptor within a few hours but without activating caspase-8. The process further activates downstream p38 MAPK signaling, a cascade that leads to the induction of apoptosis in capase-3 dependent manner. We further illustrate that Ar. subalbatus saliva suppresses proinflammatory cytokines without changing IL-10 levels, which might happen as a result of apoptosis. CONCLUSIONS Our study shows for the first time that saliva-induced apoptosis is the leading phenomenon exerted by Ar.subalbatus that impede immune cells leading to the suppression of their effecter mechanism.
Collapse
Affiliation(s)
- Shanshan Liu
- Division of Immunology, International Institute of Infection and Immunity, Shantou University Medical College, Shantou, People’s Republic of China
- Department of Pathogen Biology, Shantou University Medical College, Shantou, People’s Republic of China
| | - David J. Kelvin
- Division of Immunology, International Institute of Infection and Immunity, Shantou University Medical College, Shantou, People’s Republic of China
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Alberto J. Leon
- Division of Immunology, International Institute of Infection and Immunity, Shantou University Medical College, Shantou, People’s Republic of China
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Liqun Jin
- Department of Pathogen Biology, Shantou University Medical College, Shantou, People’s Republic of China
- * E-mail: (LJ); (AF)
| | - Amber Farooqui
- Division of Immunology, International Institute of Infection and Immunity, Shantou University Medical College, Shantou, People’s Republic of China
- Division of Experimental Therapeutics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- * E-mail: (LJ); (AF)
| |
Collapse
|
33
|
Abstract
Huge emphasis has been placed on the role of the adaptive immune system in dengue pathogenesis. Yet there is increasing evidence for the importance of the innate immune system in regulating dengue infection and possibly influencing the disease. This review focuses on the interplay between the innate immune system and dengue and highlights the role of soluble immunological mediators. Type I and type II interferons of the innate immune system demonstrate non-overlapping roles in dengue infection. Furthermore, while some IFN responses to dengue are protective, others may exert disease-related effects on the host. But aside from interferons, a number of cytokines have also been implicated in dengue pathogenesis. Our expanding knowledge of cytokines indicates that these soluble mediators act upon a complicated network of events to provoke the disease. This cytokine storm is generally attributed to massive T cell activation as an outcome of secondary infection. However, there is reason to believe that innate immune response-derived cytokines also have contributory effects, especially in the context of severe cases of primary dengue infection. Another less popular but interesting perspective on dengue pathogenesis is the effect of mosquito feeding on host immune responses and viral infection. Various studies have shown that soluble factors from vector saliva have the capacity to alter immune reactions and thereby influence pathogen transmission and establishment. Hence, modulation of the innate immune system at various levels of infection is a critical component of dengue disease. In the absence of an approved drug or vaccine for dengue, soluble mediators of the innate immune system could be a strategic foothold for developing anti-viral therapeutics and improving clinical management.
Collapse
Affiliation(s)
- Lyre Anni Espada-Murao
- Department of Virology, Institute of Tropical Medicine, GCOE Programme, Nagasaki University, Sakamoto machi 1-12-4, Nagasaki 852-8523, Japan
| | | |
Collapse
|
34
|
Bethony JM, Cole RN, Guo X, Kamhawi S, Lightowlers MW, Loukas A, Petri W, Reed S, Valenzuela JG, Hotez PJ. Vaccines to combat the neglected tropical diseases. Immunol Rev 2011; 239:237-70. [PMID: 21198676 DOI: 10.1111/j.1600-065x.2010.00976.x] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The neglected tropical diseases (NTDs) represent a group of parasitic and related infectious diseases such as amebiasis, Chagas disease, cysticercosis, echinococcosis, hookworm, leishmaniasis, and schistosomiasis. Together, these conditions are considered the most common infections in low- and middle-income countries, where they produce a level of global disability and human suffering equivalent to better known conditions such as human immunodeficiency virus/acquired immunodeficiency syndrome and malaria. Despite their global public health importance, progress on developing vaccines for NTD pathogens has lagged because of some key technical hurdles and the fact that these infections occur almost exclusively in the world's poorest people living below the World Bank poverty line. In the absence of financial incentives for new products, the multinational pharmaceutical companies have not embarked on substantive research and development programs for the neglected tropical disease vaccines. Here, we review the current status of scientific and technical progress in the development of new neglected tropical disease vaccines, highlighting the successes that have been achieved (cysticercosis and echinococcosis) and identifying the challenges and opportunities for development of new vaccines for NTDs. Also highlighted are the contributions being made by non-profit product development partnerships that are working to overcome some of the economic challenges in vaccine manufacture, clinical testing, and global access.
Collapse
Affiliation(s)
- Jeffrey M Bethony
- Microbiology, Immunology, and Tropical Medicine, George Washington University Medical Center, Washington, DC 20037, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Affiliation(s)
- Judith E Allen
- Institutes of Evolution, Immunology and Infection Research, University of Edinburgh, Edinburgh, United Kingdom.
| | | |
Collapse
|
36
|
Schneider BS, Mathieu C, Peronet R, Mécheri S. Anopheles stephensiSaliva Enhances Progression of Cerebral Malaria in a Murine Model. Vector Borne Zoonotic Dis 2011; 11:423-32. [DOI: 10.1089/vbz.2010.0120] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Bradley S. Schneider
- Biology of Host-Parasite Interactions Unit, Department of Parasitology and Mycology, Institut Pasteur, Paris Cedex, France
| | - Cedric Mathieu
- Biology of Host-Parasite Interactions Unit, Department of Parasitology and Mycology, Institut Pasteur, Paris Cedex, France
| | - Roger Peronet
- Biology of Host-Parasite Interactions Unit, Department of Parasitology and Mycology, Institut Pasteur, Paris Cedex, France
| | - Salaheddine Mécheri
- Biology of Host-Parasite Interactions Unit, Department of Parasitology and Mycology, Institut Pasteur, Paris Cedex, France
| |
Collapse
|
37
|
Ribeiro JM, Mans BJ, Arcà B. An insight into the sialome of blood-feeding Nematocera. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2010; 40:767-84. [PMID: 20728537 PMCID: PMC2950210 DOI: 10.1016/j.ibmb.2010.08.002] [Citation(s) in RCA: 134] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 08/04/2010] [Accepted: 08/09/2010] [Indexed: 05/10/2023]
Abstract
Within the Diptera and outside the suborder Brachycera, the blood-feeding habit occurred at least twice, producing the present day sand flies, and the Culicomorpha, including the mosquitoes (Culicidae), black flies (Simulidae), biting midges (Ceratopogonidae) and frog feeding flies (Corethrellidae). Alternatives to this scenario are also discussed. Successful blood-feeding requires adaptations to antagonize the vertebrate's mechanisms of blood clotting, platelet aggregation, vasoconstriction, pain and itching, which are triggered by tissue destruction and immune reactions to insect products. Saliva of these insects provides a complex pharmacological armamentarium to block these vertebrate reactions. With the advent of transcriptomics, the sialomes (from the Greek word sialo = saliva) of at least two species of each of these families have been studied (except for the frog feeders), allowing an insight into the diverse pathways leading to today's salivary composition within the Culicomorpha, having the sand flies as an outgroup. This review catalogs 1288 salivary proteins in 10 generic classes comprising over 150 different protein families, most of which we have no functional knowledge. These proteins and many sequence comparisons are displayed in a hyperlinked spreadsheet that hopefully will stimulate and facilitate the task of functional characterization of these proteins, and their possible use as novel pharmacological agents and epidemiological markers of insect vector exposure.
Collapse
Affiliation(s)
- José M.C. Ribeiro
- Section of Vector Biology, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, 12735 Twinbrook Parkway Room 2E32D, Rockville MD 20852, USA
- To whom correspondence should be addressed.
| | - Ben J. Mans
- Parasites, Vectors and Vector-Borne Diseases, Onderstepoort Veterinary Institute, Pretoria, South Africa and the Department of Veterinary Tropical Diseases, University of Pretoria, Pretoria, South Africa
| | - Bruno Arcà
- Department of Structural and Functional Biology, University Federico II, Naples, Italy
| |
Collapse
|
38
|
Harrington D, Robinson K, Guy J, Sparagano O. Characterization of the immunological response to Dermanyssus gallinae infestation in domestic fowl. Transbound Emerg Dis 2010; 57:107-10. [PMID: 20537120 DOI: 10.1111/j.1865-1682.2010.01109.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Dermanyssus gallinae is a haematophagous ectoparasite of birds, which adversely affects both production and welfare of commercial poultry. Poultry in commercial production systems chronically exposed to D. gallinae do not appear to develop immunity to the mite. The objective of the current study was to determine the initial immune response of domestic fowl following exposure to D. gallinae. Two groups of birds (11 birds/group) had mite chambers secured to their backs. Controls received no mites, while infested birds received 200 unfed female D. gallinae on day 0 which were then removed on day 1 or 2. Spleen samples were collected on days -1, 1, 2 and 5. The expression of Th1 (IFNgamma, CXCLi2, IL6 and IL18), Th2 (IL4, IL10 and IL13) cytokines/chemokines normalized against a reference gene, GAPDH, were determined by semi-quantitative RT-PCR. Although there were no significant differences between treatments, numerical trends were observed. Th2 cytokine expression was not detected in any birds on any day. IL6, CXCLi2, IFNgamma and IL18 expression was increased on day 1 in the infested group, while on day 2 CXCLi2 and IFNgamma were lower and IL6 and IL18 levels were similar between treatments. The IL18 expression was similar between treatments on day 5, while IL6 and IFNgamma levels were increased and CXCLi2 expression was decreased in the infested group. Data suggest that D. gallinae feeding stimulates Th1 and pro-inflammatory cytokines/chemokines initially (day 1) followed by their subsequent down regulation. This study is the first report of the characterization of the immunological response of the domestic fowl to controlled numbers of D. gallinae.
Collapse
Affiliation(s)
- D Harrington
- School of Agriculture, Food and Rural Development, Newcastle University, UK
| | | | | | | |
Collapse
|
39
|
Thangamani S, Higgs S, Ziegler S, Vanlandingham D, Tesh R, Wikel S. Host immune response to mosquito-transmitted chikungunya virus differs from that elicited by needle inoculated virus. PLoS One 2010; 5:e12137. [PMID: 20711354 PMCID: PMC2920837 DOI: 10.1371/journal.pone.0012137] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Accepted: 07/20/2010] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Mosquito-borne diseases are a worldwide public health threat. Mosquitoes transmit viruses or parasites during feeding, along with salivary proteins that modulate host responses to facilitate both blood feeding and pathogen transmission. Understanding these earliest events in mosquito transmission of arboviruses by mosquitoes is essential for development and assessment of rational vaccine and treatment strategies. In this report, we compared host immune responses to chikungunya virus (CHIKV) transmission by (1) mosquito bite, or (2) by needle inoculation. METHODS AND FINDINGS Differential cytokine expression was measured using quantitative real-time RT-PCR, at sites of uninfected mosquito bites, CHIKV-infected mosquito bites, and needle-inoculated CHIKV. Both uninfected and CHIKV infected mosquitoes polarized host cytokine response to a TH2 profile. Compared to uninfected mosquito bites, expression of IL-4 induced by CHIKV-infected mosquitoes were 150 fold and 527.1 fold higher at 3 hours post feeding (hpf) and 6 hpf, respectively. A significant suppression of TH1 cytokines and TLR-3 was also observed. These significant differences may result from variation in the composition of uninfected and CHIKV-infected mosquito saliva. Needle injected CHIKV induced a robust interferon-gamma, no detectable IL-4, and a significant up-regulation of TLR-3. CONCLUSIONS This report describes the first analysis of cutaneous cytokines in mice bitten by CHIKV-infected mosquitoes. Our data demonstrate contrasting immune activation in the response to CHIKV infection by mosquito bite or needle inoculation. The significant role of mosquito saliva in these earliest events of CHIKV transmission and infection are highlighted.
Collapse
Affiliation(s)
- Saravanan Thangamani
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America.
| | | | | | | | | | | |
Collapse
|
40
|
Tsujimoto H, Gray EW, Champagne DE. Black fly salivary gland extract inhibits proliferation and induces apoptosis in murine splenocytes. Parasite Immunol 2010; 32:275-84. [PMID: 20398228 DOI: 10.1111/j.1365-3024.2009.01186.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Black flies are known to be vectors of pathogens including Onchocerca volvulus, which causes human onchocerciasis, and Vesicular Stomatitis Virus. Their salivary secretion has been shown to contain a complex cocktail of anti-haemostatic factors and immunomodulatory activities, which may contribute to efficient transmission of the pathogens. Black fly salivary gland extract (SGE) inhibits mitogen-stimulated mouse splenocyte proliferation, including proliferation of both CD4(+) and CD8(+) T cells. The factor responsible for the inhibition was determined to be a protein (or protein complex) of a size larger than 50 kDa. Moreover, exposure to SGE results in activation of caspase 3 and characteristic morphological changes in CD4(+) and CD8(+) T cells, suggesting that induction of apoptosis could, at least in part, be responsible for this inhibition.
Collapse
Affiliation(s)
- H Tsujimoto
- Department of Entomology, University of Georgia, Athens, GA 30602, USA
| | | | | |
Collapse
|
41
|
Harrington DWJ, Robinson K, Sparagano OAE. Immune responses of the domestic fowl to Dermanyssus gallinae under laboratory conditions. Parasitol Res 2010; 106:1425-34. [PMID: 20333400 DOI: 10.1007/s00436-010-1821-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2010] [Accepted: 03/02/2010] [Indexed: 11/24/2022]
Abstract
There appear to be few reports in the literature regarding the host-poultry red mite (Dermanyssus gallinae) immunological relationship, despite the negative impact D. gallinae can have on both bird welfare and egg production, as well as its potential to act as a reservoir of zoonotic infections. The current study investigated the effect of either continuous infestation (CI) for 22 days or repeated infestation (RI) for four 24-h periods 7 days apart with D. gallinae on humoral immunity (IgM and IgY) and Th1/Th2 cytokine mRNA expression in peripheral blood mononuclear cells (PBMC) compared to non-infested controls. Serum IgY levels and IgM concentration were significantly higher in CI than RI and control birds although Th1 and Th2 mRNA expression in PBMC did not differ significantly between groups. D. gallinae appeared to modify reproductive behaviour and progeny survival following successive feeding events. In the RI group, the proportion of eggs/mite was significantly higher (P < 0.05) after first infestation than later infestations while larval/nymphal mortality was significantly higher (P < 0.05) after the first two infestations than later infestations. These data suggest that D. gallinae might adopt a feeding strategy of minimal host interference while D. gallinae could determine host immune status via nymphal/larval survival rates. Further research is required to better understand the host immunomodulation or avoidance strategy of D. gallinae as well as whether the mite is able to determine host immunocompetence perhaps using progeny survival.
Collapse
Affiliation(s)
- David W J Harrington
- School of Agriculture, Food and Rural Development, Newcastle University, Newcastle upon Tyne, NE1 7RU England, UK
| | | | | |
Collapse
|
42
|
Abstract
Chikungunya disease is a severely debilitating, mosquito-borne, viral illness that has reached epidemic proportions in Africa, Asia, and the islands of the Indian Ocean. A mutation enhancing the ability of the chikungunya virus (CHIKV) to infect and be transmitted by Aedes albopictus has increased the geographical range at risk for infection due to the continuing global spread of this mosquito. Research into disease pathogenesis, vaccine development, and therapeutic design has been hindered by the lack of appropriate animal models of this disease. The meticulous study reported in this issue of the JCI by Labadie et al. is one of the first reports describing CHIKV infection of adult immunocompetent nonhuman primates. Using traditional and modern molecular and immunological approaches, the authors demonstrate that macaques infected with CHIKV are a good model of human CHIKV infection and also show that persistent arthralgia in humans may be caused by persistent CHIKV infection of macrophages.
Collapse
Affiliation(s)
- Stephen Higgs
- Department of Pathology, University of Texas Medical Branch, Galveston 77555-0609, USA.
| | | |
Collapse
|
43
|
Boppana VD, Thangamani S, Alarcon-Chaidez FJ, Adler AJ, Wikel SK. Blood feeding by the Rocky Mountain spotted fever vector, Dermacentor andersoni, induces interleukin-4 expression by cognate antigen responding CD4+ T cells. Parasit Vectors 2009; 2:47. [PMID: 19814808 PMCID: PMC2764639 DOI: 10.1186/1756-3305-2-47] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2009] [Accepted: 10/08/2009] [Indexed: 11/10/2022] Open
Abstract
Background Tick modulation of host defenses facilitates both blood feeding and pathogen transmission. Several tick species deviate host T cell responses toward a Th2 cytokine profile. The majority of studies of modulation of T cell cytokine expression by ticks were performed with lymphocytes from infested mice stimulated in vitro with polyclonal T cell activators. Those reports did not examine tick modulation of antigen specific responses. We report use of a transgenic T cell receptor (TCR) adoptive transfer model reactive with influenza hemagglutinin peptide (110-120) to examine CD4+ T cell intracellular cytokine responses during infestation with the metastriate tick, Dermacentor andersoni, or exposure to salivary gland extracts. Results Infestation with pathogen-free D. andersoni nymphs or administration of an intradermal injection of female or male tick salivary gland extract induced significant increases of IL-4 transcripts in skin and draining lymph nodes of BALB/c mice as measured by quantitative real-time RT-PCR. Furthermore, IL-10 transcripts were significantly increased in skin while IL-2 and IFN-γ transcripts were not significantly changed by tick feeding or intradermal injection of salivary gland proteins, suggesting a superimposed Th2 response. Infestation induced TCR transgenic CD4+ T cells to divide more frequently as measured by CFSE dilution, but more notably these CD4+ T cells also gained the capacity to express IL-4. Intracellular levels of IL-4 were significantly increased. A second infestation administered 14 days after a primary exposure to ticks resulted in partially reduced CFSE dilution with no change in IL-4 expression when compared to one exposure to ticks. Intradermal inoculation of salivary gland extracts from both male and female ticks also induced IL-4 expression. Conclusion This is the first report of the influence of a metastriate tick on the cytokine profile of antigen specific CD4+ T cells. Blood feeding by D. andersoni pathogen-free nymphs or intradermal injection of salivary gland extracts programs influenza hemagglutinin influenza peptide specific TCR transgenic CD4+ T cells to express IL-4.
Collapse
Affiliation(s)
- Venkata D Boppana
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases and Center for Tropical Diseases, School of Medicine, University of Texas Medical Branch, Galveston, Texas, 77555, USA.
| | | | | | | | | |
Collapse
|