151
|
Zug R, Hammerstein P. Wolbachia and the insect immune system: what reactive oxygen species can tell us about the mechanisms of Wolbachia-host interactions. Front Microbiol 2015; 6:1201. [PMID: 26579107 PMCID: PMC4621438 DOI: 10.3389/fmicb.2015.01201] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 10/15/2015] [Indexed: 01/06/2023] Open
Abstract
Wolbachia are intracellular bacteria that infect a vast range of arthropod species, making them one of the most prevalent endosymbionts in the world. Wolbachia's stunning evolutionary success is mostly due to their reproductive parasitism but also to mutualistic effects such as increased host fecundity or protection against pathogens. However, the mechanisms underlying Wolbachia phenotypes, both parasitic and mutualistic, are only poorly understood. Moreover, it is unclear how the insect immune system is involved in these phenotypes and why it is not more successful in eliminating the bacteria. Here we argue that reactive oxygen species (ROS) are likely to be key in elucidating these issues. ROS are essential players in the insect immune system, and Wolbachia infection can affect ROS levels in the host. Based on recent findings, we elaborate a hypothesis that considers the different effects of Wolbachia on the oxidative environment in novel vs. native hosts. We propose that newly introduced Wolbachia trigger an immune response and cause oxidative stress, whereas in coevolved symbioses, infection is not associated with oxidative stress, but rather with restored redox homeostasis. Redox homeostasis can be restored in different ways, depending on whether Wolbachia or the host is in charge. This hypothesis offers a mechanistic explanation for several of the observed Wolbachia phenotypes.
Collapse
Affiliation(s)
- Roman Zug
- Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | | |
Collapse
|
152
|
Zink SD, Van Slyke GA, Palumbo MJ, Kramer LD, Ciota AT. Exposure to West Nile Virus Increases Bacterial Diversity and Immune Gene Expression in Culex pipiens. Viruses 2015; 7:5619-31. [PMID: 26516902 PMCID: PMC4632394 DOI: 10.3390/v7102886] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 09/23/2015] [Accepted: 10/15/2015] [Indexed: 11/16/2022] Open
Abstract
Complex interactions between microbial residents of mosquitoes and arboviruses are likely to influence many aspects of vectorial capacity and could potentially have profound effects on patterns of arbovirus transmission. Such interactions have not been well studied for West Nile virus (WNV; Flaviviridae, Flavivirus) and Culex spp. mosquitoes. We utilized next-generation sequencing of 16S ribosomal RNA bacterial genes derived from Culex pipiens Linnaeus following WNV exposure and/or infection and compared bacterial populations and broad immune responses to unexposed mosquitoes. Our results demonstrate that WNV infection increases the diversity of bacterial populations and is associated with up-regulation of classical invertebrate immune pathways including RNA interference (RNAi), Toll, and Jak-STAT (Janus kinase-Signal Transducer and Activator of Transcription). In addition, WNV exposure alone, without the establishment of infection, results in similar alterations to microbial and immune signatures, although to a lesser extent. Multiple bacterial genera were found in greater abundance in WNV-exposed and/or infected mosquitoes, yet the most consistent and notable was the genus Serratia.
Collapse
Affiliation(s)
- Steven D Zink
- Griffin Laboratory, Wadsworth Center, New York State Department of Health, Slingerlands, NY 12159, USA.
| | - Greta A Van Slyke
- Griffin Laboratory, Wadsworth Center, New York State Department of Health, Slingerlands, NY 12159, USA.
| | - Michael J Palumbo
- Wadsworth Center Bioinformatics Core, Wadsworth Center, New York State Department of Health, Albany, NY 12222, USA.
| | - Laura D Kramer
- Griffin Laboratory, Wadsworth Center, New York State Department of Health, Slingerlands, NY 12159, USA.
| | - Alexander T Ciota
- Griffin Laboratory, Wadsworth Center, New York State Department of Health, Slingerlands, NY 12159, USA.
| |
Collapse
|
153
|
Vieira LR, Polomé A, Mesquita RD, Salmon D, Braz GRC, Bousbata S. Protein 2DE reference map of the anterior midgut of the blood-sucking bug Rhodnius prolixus. Proteomics 2015; 15:3901-4. [PMID: 26314381 DOI: 10.1002/pmic.201400472] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 07/30/2015] [Accepted: 08/24/2015] [Indexed: 11/09/2022]
Abstract
Rhodnius prolixus is an important vector of Trypanosoma cruzi, the causative agent of Chagas' disease, an illness that affects 20% of Latin America population. The obligatory course of the parasite in the vector digestive tract has made it an important target for investigation in order to control the parasite transmission and thus interrupt its biological cycle in the insect vector. Therefore, an insight into the vector midgut physiology is valuable for insect control as well as to provide potential novel targets for drugs and vaccines development and thus disease treatment. In this study, the first 2DE map of R. prolixus anterior midgut is described. Proteins were separated by 2DE and analyzed by nano-LC MS/MS. The results yielded 489 proteins from 475 spots. These proteins were classified into 28 functional groups and their physiological roles in the insect midgut are discussed. All MS data have been deposited in the ProteomeXchange with identifiers PXD001488 and PXD001489 (http://proteomecentral.proteomexchange.org/dataset/PXD001488, http://proteomecentral.proteomexchange.org/dataset/PXD001489).
Collapse
Affiliation(s)
- Larissa Rezende Vieira
- Proteomic Platform, Laboratory of Molecular Parasitology, Université Libre de Bruxelles, Belgium.,Department of Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Federal University of Rio de Janeiro, Brazil.,Program of Molecular Biology and Biotechnology, Institute of Medical Biochemistry, Federal University of Rio de Janeiro, Brazil
| | - Andy Polomé
- Proteomic Platform, Laboratory of Molecular Parasitology, Université Libre de Bruxelles, Belgium
| | - Rafael Dias Mesquita
- Department of Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Federal University of Rio de Janeiro, Brazil
| | - Didier Salmon
- Program of Molecular Biology and Biotechnology, Institute of Medical Biochemistry, Federal University of Rio de Janeiro, Brazil
| | - Glória Regina Cardoso Braz
- Department of Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Federal University of Rio de Janeiro, Brazil
| | - Sabrina Bousbata
- Proteomic Platform, Laboratory of Molecular Parasitology, Université Libre de Bruxelles, Belgium
| |
Collapse
|
154
|
Hegde S, Rasgon JL, Hughes GL. The microbiome modulates arbovirus transmission in mosquitoes. Curr Opin Virol 2015; 15:97-102. [PMID: 26363996 DOI: 10.1016/j.coviro.2015.08.011] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 08/22/2015] [Accepted: 08/24/2015] [Indexed: 11/18/2022]
Abstract
Mosquito-transmitted arthropod-borne viruses (arboviruses) such as dengue virus, chikungunya virus, and West Nile virus constitute a major public health burden and are increasing in severity and frequency worldwide. The microbiota associated with mosquitoes (comprised of viruses, bacteria, fungi and protozoa) can profoundly influence many host phenotypes including vector competence, which can either be enhanced or suppressed. Thus, the tripartite interactions between the mosquito vector, its microbiota and the pathogens they transmit offer novel possibilities to control arthropod-borne diseases.
Collapse
Affiliation(s)
- Shivanand Hegde
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jason L Rasgon
- Department of Entomology, Pennsylvania State University, University Park, PA 16802, USA; The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA; Center for Infectious Disease Dynamics, Pennsylvania State University, University Park, PA 16802, USA
| | - Grant L Hughes
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
155
|
Heme Signaling Impacts Global Gene Expression, Immunity and Dengue Virus Infectivity in Aedes aegypti. PLoS One 2015; 10:e0135985. [PMID: 26275150 PMCID: PMC4537099 DOI: 10.1371/journal.pone.0135985] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 07/28/2015] [Indexed: 02/01/2023] Open
Abstract
Blood-feeding mosquitoes are exposed to high levels of heme, the product of hemoglobin degradation. Heme is a pro-oxidant that influences a variety of cellular processes. We performed a global analysis of heme-regulated Aedes aegypti (yellow fever mosquito) transcriptional changes to better understand influence on mosquito physiology at the molecular level. We observed an iron- and reactive oxygen species (ROS)-independent signaling induced by heme that comprised genes related to redox metabolism. By modulating the abundance of these transcripts, heme possibly acts as a danger signaling molecule. Furthermore, heme triggered critical changes in the expression of energy metabolism and immune response genes, altering the susceptibility towards bacteria and dengue virus. These findings seem to have implications on the adaptation of mosquitoes to hematophagy and consequently on their ability to transmit diseases. Altogether, these results may also contribute to the understanding of heme cell biology in eukaryotic cells.
Collapse
|
156
|
Wolbachia Do Not Induce Reactive Oxygen Species-Dependent Immune Pathway Activation in Aedes albopictus. Viruses 2015; 7:4624-39. [PMID: 26287231 PMCID: PMC4576197 DOI: 10.3390/v7082836] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 08/03/2015] [Accepted: 08/10/2015] [Indexed: 12/15/2022] Open
Abstract
Aedes albopictus is a major vector of dengue (DENV) and chikungunya (CHIKV) viruses, causing millions of infections annually. It naturally carries, at high frequency, the intracellular inherited bacterial endosymbiont Wolbachia strains wAlbA and wAlbB; transinfection with the higher-density Wolbachia strain wMel from Drosophila melanogaster led to transmission blocking of both arboviruses. The hypothesis that reactive oxygen species (ROS)-induced immune activation plays a role in arbovirus inhibition in this species was examined. In contrast to previous observations in Ae. aegypti, elevation of ROS levels was not observed in either cell lines or mosquito lines carrying the wild-type Wolbachia or higher-density Drosophila Wolbachia strains. There was also no upregulation of genes controlling innate immune pathways or with antioxidant/ROS-producing functions. These data suggest that ROS-mediated immune activation is not an important component of the viral transmission-blocking phenotype in this species.
Collapse
|
157
|
Gonzales KK, Tsujimoto H, Hansen IA. Blood serum and BSA, but neither red blood cells nor hemoglobin can support vitellogenesis and egg production in the dengue vector Aedes aegypti. PeerJ 2015; 3:e938. [PMID: 26020000 PMCID: PMC4435475 DOI: 10.7717/peerj.938] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 04/16/2015] [Indexed: 11/26/2022] Open
Abstract
Aedes aegypti is the major vector of dengue, yellow fever and chikungunya viruses that put millions of people in endemic countries at risk. Mass rearing of this mosquito is crucial for strategies that use modified insects to reduce vector populations and transmission of pathogens, such as sterile insect technique or population replacement. A major problem for vector mosquito mass rearing is the requirement of vertebrate blood for egg production since it poses significant costs as well as potential health hazards. Also, regulations for human and animal use as blood source can pose a significant obstacle. A completely artificial diet that supports egg production in vector mosquitoes can solve this problem. In this study, we compared different blood fractions, serum and red blood cells, as dietary protein sources for mosquito egg production. We also tested artificial diets made from commercially available blood proteins (bovine serum albumin (BSA) and hemoglobin). We found that Ae. aegypti performed vitellogenesis and produced eggs when given whole bovine blood, serum, or an artificial diet containing BSA. Conversely, egg production was impaired after feeding of the red blood cell fraction or an artificial diet containing only hemoglobin. We also found that egg viability of serum-fed mosquitoes were comparable to that of whole blood and an iron supplemented BSA meal produced more viable eggs than a meal containing BSA alone. Our results indicate that serum proteins, not hemoglobin, may replace vertebrate blood in artificial diets for mass mosquito rearing.
Collapse
Affiliation(s)
| | - Hitoshi Tsujimoto
- Department of Biology, New Mexico State University , Las Cruces, NM , USA
| | - Immo A Hansen
- Department of Biology, New Mexico State University , Las Cruces, NM , USA ; Institute for Applied Biosciences, New Mexico State University , Las Cruces, NM , USA ; Molecular Biology Program, New Mexico State University , Las Cruces, NM , USA
| |
Collapse
|
158
|
Zhang L, Lu Z. Expression, purification and characterization of an atypical 2-Cys peroxiredoxin from the silkworm, Bombyx mori. INSECT MOLECULAR BIOLOGY 2015; 24:203-212. [PMID: 25512182 DOI: 10.1111/imb.12149] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Peroxiredoxins (Prxs) play important roles in protecting organisms against damage caused by reactive oxygen species (ROS). In this study, we cloned a cDNA of Bombyx mori peroxiredoxin 5 (BmPrx5), which contained a 565-bp open reading frame for a 188-residue protein. Sequence analysis indicated that BmPrx5 belongs to the atypical 2-Cys peroxiredoxin family. Recombinant BmPrx5 purified from Escherichia coli showed antioxidant activity that removes H2 O2 and protects DNA from oxidative damage. Quantitative real-time PCR showed that the level of BmPrx5 mRNA in haemocytes increased early and decreased by 24 h after injection of H2 O2 whereas, in the fat body, the transcript level decreased at 6 h and increased at 12 h. Pseudomonas aeruginosa and Staphylococcus aureus infection resulted in higher levels of H2 O2 in the haemolymph and of BmPrx5 mRNA in haemocytes at 8 h postinfection. These data suggest that BmPrx5 acts as an antioxidant enzyme to protect the silkworm from oxidative damage induced by bacterial infection. Further study is needed to elucidate the exact role of BmPrx5 in the silkworm immune system.
Collapse
Affiliation(s)
- L Zhang
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, China
| | | |
Collapse
|
159
|
Geiger A, Ponton F, Simo G. Adult blood-feeding tsetse flies, trypanosomes, microbiota and the fluctuating environment in sub-Saharan Africa. ISME JOURNAL 2014; 9:1496-507. [PMID: 25500509 DOI: 10.1038/ismej.2014.236] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2014] [Revised: 11/03/2014] [Accepted: 11/06/2014] [Indexed: 01/01/2023]
Abstract
The tsetse fly vector transmits the protozoan Trypanosoma brucei, responsible for Human African Trypanosomiasis, one of the most neglected tropical diseases. Despite a recent decline in new cases, it is still crucial to develop alternative strategies to combat this disease. Here, we review the literature on the factors that influence trypanosome transmission from the fly vector to its vertebrate host (particularly humans). These factors include climate change effects to pathogen and vector development (in particular climate warming), as well as the distribution of host reservoirs. Finally, we present reports on the relationships between insect vector nutrition, immune function, microbiota and infection, to demonstrate how continuing research on the evolving ecology of these complex systems will help improve control strategies. In the future, such studies will be of increasing importance to understand how vector-borne diseases are spread in a changing world.
Collapse
Affiliation(s)
- Anne Geiger
- UMR 177, IRD-CIRAD, CIRAD TA A-17/G, Campus International de Baillarguet, Montpellier Cedex 5, France
| | - Fleur Ponton
- 1] School of Biological Sciences, The University of Sydney, Sydney, New South Wales, Australia [2] The Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Gustave Simo
- Molecular Parasitology and Entomology Unit, Department of Biochemistry, Faculty of Science, University of Dschang, Dschang, Cameroon
| |
Collapse
|
160
|
Abstract
The bacterium Wolbachia (order Rickettsiales) is probably the world's most successful vertically-transmitted symbiont, distributed among a staggering 40% of terrestrial arthropod species. Wolbachia has great potential in vector control due to its ability to manipulate its hosts' reproduction and to impede the replication and dissemination of arboviruses and other pathogens within haematophagous arthropods. In addition, the unexpected presence of Wolbachia in filarial nematodes of medical and veterinary importance has provided an opportunity to target the adult worms of Wuchereria bancrofti, Onchocerca volvulus, and Dirofilaria immitis with safe drugs such as doxycycline. A striking feature of Wolbachia is its phenotypic plasticity between (and sometimes within) hosts, which may be underpinned by its ability to integrate itself into several key processes within eukaryotic cells: oxidative stress, autophagy, and apoptosis. Importantly, despite significant differences in the genomes of arthropod and filarial Wolbachia strains, these nexuses appear to lie on a continuum in different hosts. Here, we consider how iron metabolism may represent a fundamental aspect of host homeostasis that is impacted by Wolbachia infection, connecting disparate pathways ranging from the provision of haem and ATP to programmed cell death, aging, and the recycling of intracellular resources. Depending on how Wolbachia and host cells interact across networks that depend on iron, the gradient between parasitism and mutualism may shift dynamically in some systems, or alternatively, stabilise on one or the other end of the spectrum.
Collapse
Affiliation(s)
- Alessandra Christina Gill
- Institute of Infection & Global Health, University of Liverpool, Liverpool, Merseyside, United Kingdom
| | - Alistair C. Darby
- Institute of Integrative Biology and the Centre for Genomic Research, University of Liverpool, Liverpool, Merseyside, United Kingdom
| | - Benjamin L. Makepeace
- Institute of Infection & Global Health, University of Liverpool, Liverpool, Merseyside, United Kingdom
- * E-mail:
| |
Collapse
|
161
|
Pernice M, Simpson SJ, Ponton F. Towards an integrated understanding of gut microbiota using insects as model systems. JOURNAL OF INSECT PHYSIOLOGY 2014; 69:12-8. [PMID: 24862156 DOI: 10.1016/j.jinsphys.2014.05.016] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Revised: 05/01/2014] [Accepted: 05/12/2014] [Indexed: 05/26/2023]
Abstract
Metazoans form symbioses with microorganisms that synthesize essential nutritional compounds and increase their efficiency to digest and absorb nutrients. Despite the growing awareness that microbes within the gut play key roles in metabolism, health and development of metazoans, symbiotic relationships within the gut are far from fully understood. Insects, which generally harbor a lower microbial diversity than vertebrates, have recently emerged as potential model systems to study these interactions. In this review, we give a brief overview of the characteristics of the gut microbiota in insects in terms of low diversity but high variability at intra- and interspecific levels and we investigate some of the ecological and methodological factors that might explain such variability. We then emphasize how studies integrating an array of techniques and disciplines have the potential to provide new understanding of the biology of this micro eco-system.
Collapse
Affiliation(s)
- Mathieu Pernice
- School of Biological Sciences, The University of Sydney, NSW 2006, Australia; Plant Functional Biology and Climate Change Cluster, University of Technology Sydney, NSW 2007, Australia
| | - Stephen J Simpson
- School of Biological Sciences, The University of Sydney, NSW 2006, Australia; The Charles Perkins Centre, The University of Sydney, NSW 2006, Australia
| | - Fleur Ponton
- School of Biological Sciences, The University of Sydney, NSW 2006, Australia; The Charles Perkins Centre, The University of Sydney, NSW 2006, Australia.
| |
Collapse
|
162
|
Pike A, Vadlamani A, Sandiford SL, Gacita A, Dimopoulos G. Characterization of the Rel2-regulated transcriptome and proteome of Anopheles stephensi identifies new anti-Plasmodium factors. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2014; 52:82-93. [PMID: 24998399 PMCID: PMC4143444 DOI: 10.1016/j.ibmb.2014.06.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 04/22/2014] [Accepted: 06/16/2014] [Indexed: 06/03/2023]
Abstract
Mosquitoes possess an innate immune system that is capable of limiting infection by a variety of pathogens, including the Plasmodium spp. parasites responsible for human malaria. The Anopheles immune deficiency (IMD) innate immune signaling pathway confers resistance to Plasmodium falciparum. While some previously identified Anopheles anti-Plasmodium effectors are regulated through signaling by Rel2, the transcription factor of the IMD pathway, many components of this defense system remain uncharacterized. To begin to better understand the regulation of immune effector proteins by the IMD pathway, we used oligonucleotide microarrays and iTRAQ to analyze differences in mRNA and protein expression, respectively, between transgenic Anopheles stephensi mosquitoes exhibiting blood meal-inducible overexpression of an active recombinant Rel2 and their wild-type conspecifics. Numerous genes were differentially regulated at both the mRNA and protein levels following induction of Rel2. While multiple immune genes were up-regulated, a majority of the differentially expressed genes have no known immune function in mosquitoes. Selected up-regulated genes from multiple functional categories were tested for both anti-Plasmodium and anti-bacterial action using RNA interference (RNAi). Based on our experimental findings, we conclude that increased expression of the IMD immune pathway-controlled transcription factor Rel2 affects the expression of numerous genes with diverse functions, suggesting a broader physiological impact of immune activation and possible functional versatility of Rel2. Our study has also identified multiple novel genes implicated in anti-Plasmodium defense.
Collapse
Affiliation(s)
- Andrew Pike
- W. Harry Feinstone Department of Molecular Microbiology and Immunology and the Johns Hopkins Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe Street, Baltimore, MD 21205-2179, USA.
| | - Alekhya Vadlamani
- W. Harry Feinstone Department of Molecular Microbiology and Immunology and the Johns Hopkins Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe Street, Baltimore, MD 21205-2179, USA.
| | - Simone L Sandiford
- W. Harry Feinstone Department of Molecular Microbiology and Immunology and the Johns Hopkins Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe Street, Baltimore, MD 21205-2179, USA.
| | - Anthony Gacita
- W. Harry Feinstone Department of Molecular Microbiology and Immunology and the Johns Hopkins Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe Street, Baltimore, MD 21205-2179, USA.
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology and the Johns Hopkins Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, 615 N. Wolfe Street, Baltimore, MD 21205-2179, USA.
| |
Collapse
|
163
|
Hughes GL, Dodson BL, Johnson RM, Murdock CC, Tsujimoto H, Suzuki Y, Patt AA, Cui L, Nossa CW, Barry RM, Sakamoto JM, Hornett EA, Rasgon JL. Native microbiome impedes vertical transmission of Wolbachia in Anopheles mosquitoes. Proc Natl Acad Sci U S A 2014; 111:12498-503. [PMID: 25114252 PMCID: PMC4151774 DOI: 10.1073/pnas.1408888111] [Citation(s) in RCA: 172] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Over evolutionary time, Wolbachia has been repeatedly transferred between host species contributing to the widespread distribution of the symbiont in arthropods. For novel infections to be maintained, Wolbachia must infect the female germ line after being acquired by horizontal transfer. Although mechanistic examples of horizontal transfer exist, there is a poor understanding of factors that lead to successful vertical maintenance of the acquired infection. Using Anopheles mosquitoes (which are naturally uninfected by Wolbachia) we demonstrate that the native mosquito microbiota is a major barrier to vertical transmission of a horizontally acquired Wolbachia infection. After injection into adult Anopheles gambiae, some strains of Wolbachia invade the germ line, but are poorly transmitted to the next generation. In Anopheles stephensi, Wolbachia infection elicited massive blood meal-induced mortality, preventing development of progeny. Manipulation of the mosquito microbiota by antibiotic treatment resulted in perfect maternal transmission at significantly elevated titers of the wAlbB Wolbachia strain in A. gambiae, and alleviated blood meal-induced mortality in A. stephensi enabling production of Wolbachia-infected offspring. Microbiome analysis using high-throughput sequencing identified that the bacterium Asaia was significantly reduced by antibiotic treatment in both mosquito species. Supplementation of an antibiotic-resistant mutant of Asaia to antibiotic-treated mosquitoes completely inhibited Wolbachia transmission and partly contributed to blood meal-induced mortality. These data suggest that the components of the native mosquito microbiota can impede Wolbachia transmission in Anopheles. Incompatibility between the microbiota and Wolbachia may in part explain why some hosts are uninfected by this endosymbiont in nature.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Carlos W Nossa
- Department of Ecology and Evolutionary Biology, Rice University, Houston, TX 77005; and
| | | | | | - Emily A Hornett
- Biology, Center for Infectious Disease Dynamics, and the Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802; The Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, United Kingdom
| | | |
Collapse
|
164
|
Oliver SV, Brooke BD. The effect of multiple blood-feeding on the longevity and insecticide resistant phenotype in the major malaria vector Anopheles arabiensis (Diptera: Culicidae). Parasit Vectors 2014; 7:390. [PMID: 25150975 PMCID: PMC4161849 DOI: 10.1186/1756-3305-7-390] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 08/13/2014] [Indexed: 11/10/2022] Open
Abstract
Background Anopheles arabiensis is a major malaria vector in Africa. Adult females are likely to imbibe multiple blood meals during their lifetime. This results in regular exposure to potential toxins and blood-meal induced oxidative stress. Defence responses to these stressors may affect other factors of epidemiological significance, such as insecticide resistance and longevity. The aims of this study were to examine the effect of multiple blood-feeding on insecticide tolerance/resistance with increasing age, to assess the underlying biochemical mechanisms for the responses recorded, and to assess the effect of multiple blood-feeding on the life histories of adult females drawn from insecticide resistant and susceptible laboratory reared An. arabiensis. Methods Laboratory reared An. arabiensis females from an insecticide resistant and an insecticide susceptible colony were offered either a single blood meal or multiple blood meals at 3-day intervals. Their tolerance or resistance to insecticide was then monitored by WHO bioassay four hours post blood-feeding. The biochemical basis of the phenotypic response was assessed by examining the effect of blood on detoxification enzyme activity and the effect of blood-meals on detoxification enzyme activity in ageing mosquitoes. Results Control cohorts that were not offered any blood meals showed steadily decreasing levels of insecticide tolerance/resistance with age, whereas a single blood meal significantly increased tolerance/resistance primarily at the age of three days. The expression of resistance/tolerance in those cohorts fed multiple blood meals generally showed the least variation with age. These results were consistent following exposure to DDT and pyrethroids but not to malathion. Multiple blood-meals also maintained the DDT and permethrin resistant phenotype, even after treatment females had stopped taking blood-meals. Biochemical analysis suggests that this phenotypic effect in resistant females may be mediated by the maintenance of increased glutathione s-transferase activity as a consequence of multiple blood-feeding. Multiple blood-feeding increased the longevity of insecticide resistant females regardless of their mating status, but only increased the longevity of unmated susceptible females. Conclusion These data suggest that multiple blood-feeding confers a competitive advantage to insecticide resistant females by increased longevity and maintenance of the expression of resistance with age.
Collapse
Affiliation(s)
- Shüné V Oliver
- Centre for Opportunistic, Tropical and Hospital Infections, National Institute for Communicable Diseases/NHLS, Sandringham, Johannesburg, South Africa.
| | | |
Collapse
|
165
|
Gene discovery and differential expression analysis of humoral immune response elements in female Culicoides sonorensis (Diptera: Ceratopogonidae). Parasit Vectors 2014; 7:388. [PMID: 25145345 PMCID: PMC4158122 DOI: 10.1186/1756-3305-7-388] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 08/01/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Female Culicoides sonorensis midges (Diptera: Ceratopogonidae) are vectors of pathogens that impact livestock and wildlife in the United States. Little is known about their biology on a molecular-genetic level, including components of their immune system. Because the insect immune response is involved with important processes such as gut microbial homeostasis and vector competence, our aims were to identify components of the midge innate immune system and examine their expression profiles in response to diet across time. METHODS In our previous work, we de novo sequenced and analyzed the transcriptional landscape of female midges under several feeding states including teneral (unfed) and early and late time points after blood and sucrose. Here, those transcriptomes were further analyzed to identify insect innate immune orthologs, particularly humoral immune response elements. Additionally, we examined immune gene expression profiles in response to diet over time, on both a transcriptome-wide, whole-midge level and more specifically via qRTPCR analysis of antimicrobial peptide (AMP) expression in the alimentary canal. RESULTS We identified functional units comprising the immune deficiency (Imd), Toll and JAK/STAT pathways, including humoral factors, transmembrane receptors, signaling components, transcription factors/regulators and effectors such as AMPs. Feeding altered the expression of receptors, regulators, AMPs, prophenoloxidase and thioester-containing proteins, where blood had a greater effect than sucrose on the expression profiles of most innate immune components. qRTPCR of AMP genes showed that all five were significantly upregulated in the alimentary canal after blood feeding, possibly in response to proliferating populations of gut bacteria. CONCLUSIONS Identification and functional insight of humoral/innate immune components in female C. sonorensis updates our knowledge of the molecular biology of this important vector. Because diet alone influenced the expression of immune pathway components, including their effectors, subsequent study of the role of innate immunity in biological processes such as gut homeostasis and life history are being pursued. Furthermore, since the humoral response is a key contributor in gut immunity, manipulating immune gene expression will help in uncovering genetic components of vector competence, including midgut barriers to infection. The results of such studies will serve as a platform for designing novel transmission-blocking strategies.
Collapse
|
166
|
Alves-Bezerra M, Cosentino-Gomes D, Vieira LP, Rocco-Machado N, Gondim KC, Meyer-Fernandes JR. Identification of uncoupling protein 4 from the blood-sucking insect Rhodnius prolixus and its possible role on protection against oxidative stress. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2014; 50:24-33. [PMID: 24746771 DOI: 10.1016/j.ibmb.2014.03.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 03/14/2014] [Accepted: 03/24/2014] [Indexed: 06/03/2023]
Abstract
Uncoupling proteins (UCPs) play a critical role in the control of the mitochondrial membrane potential (ΔΨm) due to their ability to dissipate the proton gradient, which results in the uncoupling of mitochondrial respiration from ATP production. Most reactive oxygen species generation in mitochondria occurs in complex III, due to an increase of semiquinone (Q(-)) half-life. When active, UCPs can account as a potential antioxidant system by decreasing ΔΨm and increasing mitochondrial respiration, thus reducing Q(-) life time. The hematophagous insect Rhodnius prolixus, a vector of Chagas disease, is exposed to a huge increase in oxidative stress after a blood meal because of the hydrolysis of hemoglobin and the release of the cytotoxic heme molecule. Although some protective mechanisms were already described for this insect and other hematophagous arthropods, the putative role of UCP proteins as antioxidants in this context has not been explored. In this report, two genes encoding UCP proteins (RpUcp4 and RpUcp5) were identified in the R. prolixus genome. RpUcp4 is the predominant transcript in most analyzed organs, and both mRNA and protein expression are upregulated (13- and 3-fold increase, respectively) in enterocytes the first day after the blood feeding. The increase in UCP4 expression is coincident with the decrease in hydrogen peroxide (H2O2) generation by midgut cells. Furthermore, in mitochondria isolated from enterocytes, the modulation of UCP activity by palmitic acid and GDP resulted in altered ΔΨm, as well as modulation of H2O2 generation rates. These results indicate that R. prolixus UCP4 may function in an antioxidation mechanism to protect the midgut cells against oxidative damage caused by blood digestion.
Collapse
Affiliation(s)
- Michele Alves-Bezerra
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Daniela Cosentino-Gomes
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lisvane P Vieira
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Nathália Rocco-Machado
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Katia C Gondim
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Brazil
| | - José R Meyer-Fernandes
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Brazil.
| |
Collapse
|
167
|
De Deken X, Corvilain B, Dumont JE, Miot F. Roles of DUOX-mediated hydrogen peroxide in metabolism, host defense, and signaling. Antioxid Redox Signal 2014; 20:2776-93. [PMID: 24161126 DOI: 10.1089/ars.2013.5602] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE Among the NADPH oxidases, the dual oxidases, DUOX1 and DUOX2, constitute a distinct subfamily initially called thyroid oxidases, based on their high level of expression in thyroid tissue. Genetic alterations causing inherited hypothyroidism clearly demonstrate their physiological implication in thyroid hormonogenesis. However, a growing list of biological functions triggered by DUOX-dependent reactive oxygen species (ROS) in highly differentiated mucosae have recently emerged. RECENT ADVANCES A role of DUOX enzymes as ROS providers for lactoperoxidase-mediated killing of invading pathogens has been well established and a role in bacteria chemorepulsion has been proposed. Control of DUOX expression and activity by inflammatory molecules and immune receptor activation consolidates their contributions to innate immune defense of mucosal surfaces. Recent studies conducted in ancestral organisms have identified effectors of DUOX redox signaling involved in wound healing including epithelium regeneration and leukocyte recruitment. Moreover, local generation of hydrogen peroxide (H2O2) by DUOX has also been suggested to constitute a positive feedback loop to promote receptor signaling activation. CRITICAL ISSUES A correct balance between H2O2 generation and detoxification mechanisms must be properly maintained to avoid oxidative damages. Overexpression of DUOX genes has been associated with an increasing number of chronic inflammatory diseases. Furthermore, H2O2-mediated DNA damage supports a mutagenic function promoting tumor development. FUTURE DIRECTIONS Despite the high sequence similarity shared between DUOX1 and DUOX2, the two isoforms present distinct regulations, tissue expression and catalytic functions. The phenotypic characterization of novel DUOX/DUOXA invalidated animal models will be very useful for defining their medical importance in pathological conditions.
Collapse
Affiliation(s)
- Xavier De Deken
- Faculté de Médecine, Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB) , Brussels, Belgium
| | | | | | | |
Collapse
|
168
|
Abstract
Many arboviral diseases are uncontrolled, and the viruses that cause them are globally emerging or reemerging pathogens that produce significant disease throughout the world. The increased spread and prevalence of disease are occurring during a period of substantial scientific growth in the vector-borne disease research community. This growth has been supported by advances in genomics and proteomics, and by the ability to genetically alter disease vectors. For the first time, researchers are elucidating the molecular details of vector host-seeking behavior, the susceptibility of disease vectors to arboviruses, the immunological control of infection in disease vectors, and the determinants that facilitate transmission of arboviruses from a vector to a host. These discoveries are facilitating the development of novel strategies to combat arboviral disease, including the release of transgenic mosquitoes harboring dominant lethal genes, the introduction of arbovirus-blocking microbes into mosquito populations, and the development of acquisition- and transmission-blocking therapeutics. Understanding the role of the vector in arbovirus transmission has provided critical practical and theoretical tools to control arboviral disease.
Collapse
Affiliation(s)
- Michael J Conway
- Foundational Sciences, Central Michigan University College of Medicine, Mt. Pleasant, Michigan 48859
| | - Tonya M Colpitts
- Department of Tropical Medicine, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana 70112
| | - Erol Fikrig
- Department of Internal Medicine, Infectious Diseases Section, Yale University School of Medicine, New Haven, Connecticut 06520; .,Howard Hughes Medical Institute, Chevy Chase, Maryland 20815
| |
Collapse
|
169
|
Dutra FF, Bozza MT. Heme on innate immunity and inflammation. Front Pharmacol 2014; 5:115. [PMID: 24904418 PMCID: PMC4035012 DOI: 10.3389/fphar.2014.00115] [Citation(s) in RCA: 235] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Accepted: 04/29/2014] [Indexed: 12/30/2022] Open
Abstract
Heme is an essential molecule expressed ubiquitously all through our tissues. Heme plays major functions in cellular physiology and metabolism as the prosthetic group of diverse proteins. Once released from cells and from hemeproteins free heme causes oxidative damage and inflammation, thus acting as a prototypic damage-associated molecular pattern. In this context, free heme is a critical component of the pathological process of sterile and infectious hemolytic conditions including malaria, hemolytic anemias, ischemia-reperfusion, and hemorrhage. The plasma scavenger proteins hemopexin and albumin reduce heme toxicity and are responsible for transporting free heme to intracellular compartments where it is catabolized by heme-oxygenase enzymes. Upon hemolysis or severe cellular damage the serum capacity to scavenge heme may saturate and increase free heme to sufficient amounts to cause tissue damage in various organs. The mechanism by which heme causes reactive oxygen generation, activation of cells of the innate immune system and cell death are not fully understood. Although heme can directly promote lipid peroxidation by its iron atom, heme can also induce reactive oxygen species generation and production of inflammatory mediators through the activation of selective signaling pathways. Heme activates innate immune cells such as macrophages and neutrophils through activation of innate immune receptors. The importance of these events has been demonstrated in infectious and non-infectious diseases models. In this review, we will discuss the mechanisms behind heme-induced cytotoxicity and inflammation and the consequences of these events on different tissues and diseases.
Collapse
Affiliation(s)
- Fabianno F. Dutra
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Marcelo T. Bozza
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Instituto de Microbiologia, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| |
Collapse
|
170
|
Sharma P, Sharma S, Maurya RK, De TD, Thomas T, Lata S, Singh N, Pandey KC, Valecha N, Dixit R. Salivary glands harbor more diverse microbial communities than gut in Anopheles culicifacies. Parasit Vectors 2014; 7:235. [PMID: 24886293 PMCID: PMC4062515 DOI: 10.1186/1756-3305-7-235] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 05/14/2014] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND In recent years, it has been well documented that gut flora not only influence mosquito physiology, but also significantly alter vector competency. Although, salivary gland and gut constitute key partners of the digestive system, it is still believed that salivary glands may harbor less flora than gut (Parasit Vectors 6: 146, 2013). METHODS Using a metagenomic approach, we have identified for the first time the diverse microbial community associated with these two physiologically different tissues of the digestive system in the mosquito Anopheles culicifacies. RESULTS A total of 17 different phyla could be assigned to the whole metagenomic dataset, predominated by the phylum Proteobacteria, Firmicutes, Bacteriodetes, Tenericutes and Actinomycetes. Common bacteria included the members of Enhydrobacter, Agromonas, Serratia, Ralsonia, Lactobacillus, Pseudomonas, Streptococcus, Rubrobacter, Anaerococcus, Methylobacterium, Turicibacter, Elizabethkingia etc. in both the tissues representing 'core microbiota' of the mosquito digestive system. Salivary associated unique bacterial community included the members of Chloriflexi, Chlorobi, Cyanobacteria, Nitrospira, TM7, Armatimonadetes, Planctomycetes, Fibrobacteres etc. CONCLUSION We find that the salivary gland microbial community structure is more diverse than gut of the mosquito, probably due to differential feeding associated engagements such as food acquisition, ingestion and digestion processes.
Collapse
Affiliation(s)
- Punita Sharma
- Host-Parasite Interaction Biology Group, National Institute of Malaria Research, Sector-8, Dwarka, Delhi 110077, India
- Department of Bio & Nano Technology, Guru Jambheswar University of Science and Technology, Hisar, Haryana 125001, India
| | - Swati Sharma
- Host-Parasite Interaction Biology Group, National Institute of Malaria Research, Sector-8, Dwarka, Delhi 110077, India
| | | | - Tanwee Das De
- Host-Parasite Interaction Biology Group, National Institute of Malaria Research, Sector-8, Dwarka, Delhi 110077, India
| | - Tina Thomas
- Host-Parasite Interaction Biology Group, National Institute of Malaria Research, Sector-8, Dwarka, Delhi 110077, India
| | - Suman Lata
- Host-Parasite Interaction Biology Group, National Institute of Malaria Research, Sector-8, Dwarka, Delhi 110077, India
| | - Namita Singh
- Department of Bio & Nano Technology, Guru Jambheswar University of Science and Technology, Hisar, Haryana 125001, India
| | - Kailash Chand Pandey
- Host-Parasite Interaction Biology Group, National Institute of Malaria Research, Sector-8, Dwarka, Delhi 110077, India
| | - Neena Valecha
- Host-Parasite Interaction Biology Group, National Institute of Malaria Research, Sector-8, Dwarka, Delhi 110077, India
| | - Rajnikant Dixit
- Host-Parasite Interaction Biology Group, National Institute of Malaria Research, Sector-8, Dwarka, Delhi 110077, India
| |
Collapse
|
171
|
Coon KL, Vogel KJ, Brown MR, Strand MR. Mosquitoes rely on their gut microbiota for development. Mol Ecol 2014; 23:2727-39. [PMID: 24766707 DOI: 10.1111/mec.12771] [Citation(s) in RCA: 354] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Revised: 04/20/2014] [Accepted: 04/23/2014] [Indexed: 02/01/2023]
Abstract
Field studies indicate adult mosquitoes (Culicidae) host low diversity communities of bacteria that vary greatly among individuals and species. In contrast, it remains unclear how adult mosquitoes acquire their microbiome, what influences community structure, and whether the microbiome is important for survival. Here, we used pyrosequencing of 16S rRNA to characterize the bacterial communities of three mosquito species reared under identical conditions. Two of these species, Aedes aegypti and Anopheles gambiae, are anautogenous and must blood-feed to produce eggs, while one, Georgecraigius atropalpus, is autogenous and produces eggs without blood feeding. Each mosquito species contained a low diversity community comprised primarily of aerobic bacteria acquired from the aquatic habitat in which larvae developed. Our results suggested that the communities in Ae. aegypti and An. gambiae larvae share more similarities with one another than with G. atropalpus. Studies with Ae. aegypti also strongly suggested that adults transstadially acquired several members of the larval bacterial community, but only four genera of bacteria present in blood fed females were detected on eggs. Functional assays showed that axenic larvae of each species failed to develop beyond the first instar. Experiments with Ae. aegypti indicated several members of the microbial community and Escherichia coli successfully colonized axenic larvae and rescued development. Overall, our results provide new insights about the acquisition and structure of bacterial communities in mosquitoes. They also indicate that three mosquito species spanning the breadth of the Culicidae depend on their gut microbiome for development.
Collapse
Affiliation(s)
- Kerri L Coon
- Department of Entomology, The University of Georgia, 120 Cedar Street, 420 Biological Sciences, Athens, GA, 30602, USA
| | | | | | | |
Collapse
|
172
|
Moné Y, Monnin D, Kremer N. The oxidative environment: a mediator of interspecies communication that drives symbiosis evolution. Proc Biol Sci 2014; 281:20133112. [PMID: 24807248 DOI: 10.1098/rspb.2013.3112] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Symbiotic interactions are ubiquitous in nature and play a major role in driving the evolution of life. Interactions between partners are often mediated by shared signalling pathways, which strongly influence both partners' biology and the evolution of the association in various environments. As an example of 'common language', the regulation of the oxidative environment plays an important role in driving the evolution of symbiotic associations. Such processes have been occurring for billions of years, including the increase in Earth's atmospheric oxygen and the subsequent evolution of mitochondria. The effect of reactive oxygen species and reactive nitrogen species (RONS) has been characterized functionally, but the molecular dialogue between partners has not been integrated within a broader evolutionary context yet. Given the pleiotropic role of RONS in cell-cell communication, development and immunity, but also their associated physiological costs, we discuss here how their regulation can influence the establishment, the maintenance and the breakdown of various symbiotic associations. By synthesizing recent developments in redox biology, we aim to provide an interdisciplinary understanding of the influence of such mediators of interspecies communication on the evolution and stability of symbioses, which in turn can shape ecosystems and play a role in health and disease.
Collapse
Affiliation(s)
- Yves Moné
- Université Lyon 1, CNRS, UMR 5558, Laboratoire de Biométrie et Biologie Evolutive, Université de Lyon, , Villeurbanne, France, INSA-Lyon, INRA, UMR 203, Biologie Fonctionnelle Insectes et Interactions, Université de Lyon, , Villeurbanne, France, Medical Microbiology and Immunology, University of Wisconsin-Madison, , Madison, WI, USA
| | | | | |
Collapse
|
173
|
The molecular basis of bacterial-insect symbiosis. J Mol Biol 2014; 426:3830-7. [PMID: 24735869 DOI: 10.1016/j.jmb.2014.04.005] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/02/2014] [Accepted: 04/08/2014] [Indexed: 12/12/2022]
Abstract
Insects provide experimentally tractable and cost-effective model systems to investigate the molecular basis of animal-bacterial interactions. Recent research is revealing the central role of the insect innate immune system, especially anti-microbial peptides and reactive oxygen species, in regulating the abundance and composition of the microbiota in various insects, including Drosophila and the mosquitoes Aedes and Anopheles. Interactions between the immune system and microbiota are, however, bidirectional with evidence that members of the resident microbiota can promote immune function, conferring resistance to pathogens and parasites by both activation of immune effectors and production of toxins. Antagonistic and mutualistic interactions among bacteria have also been implicated as determinants of the microbiota composition, including exclusion of pathogens, but the molecular mechanisms are largely unknown. Some bacteria are crucial for insect nutrition, through provisioning of specific nutrients (e.g., B vitamins, essential amino acids) and modulation of the insect nutritional sensing and signaling pathways (e.g., insulin signaling) that regulate nutrient allocation, especially to lipid and other energy reserves. A key challenge for future research is to identify the molecular interaction between specific bacterial effectors and animal receptors, as well as to determine how these interactions translate into microbiota-dependent signaling, metabolism, and immune function in the host.
Collapse
|
174
|
Bahia AC, Dong Y, Blumberg BJ, Mlambo G, Tripathi A, BenMarzouk-Hidalgo OJ, Chandra R, Dimopoulos G. Exploring Anopheles gut bacteria for Plasmodium blocking activity. Environ Microbiol 2014; 16:2980-94. [PMID: 24428613 DOI: 10.1111/1462-2920.12381] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 12/31/2013] [Indexed: 12/30/2022]
Abstract
Malaria parasite transmission requires the successful development of Plasmodium gametocytes into flagellated microgametes upon mosquito blood ingestion, and the subsequent fertilization of microgametes and macrogametes for the development of motile zygotes, called ookinetes, which invade and transverse the Anopheles vector mosquito midgut at around 18-36 h after blood ingestion. Within the mosquito midgut, the malaria parasite has to withstand the mosquito's innate immune response and the detrimental effect of its commensal bacterial flora. We have assessed the midgut colonization capacity of five gut bacterial isolates from field-derived, and two from laboratory colony, mosquitoes and their effect on Plasmodium development in vivo and in vitro, along with their impact on mosquito survival. Some bacterial isolates activated the mosquito's immune system, affected the mosquito's lifespan, and were capable of blocking Plasmodium development. We have also shown that the ability of these bacteria to inhibit the parasites is likely to involve different mechanisms and factors. A Serratia marcescens isolate was particularly efficient in colonizing the mosquitoes' gut, compromising mosquito survival and inhibiting both Plasmodium sexual- and asexual-stage through secreted factors, thereby rendering it a potential candidate for the development of a malaria transmission intervention strategy.
Collapse
Affiliation(s)
- Ana C Bahia
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | |
Collapse
|
175
|
Adamson SW, Browning RE, Budachetri K, Ribeiro JMC, Karim S. Knockdown of selenocysteine-specific elongation factor in Amblyomma maculatum alters the pathogen burden of Rickettsia parkeri with epigenetic control by the Sin3 histone deacetylase corepressor complex. PLoS One 2013; 8:e82012. [PMID: 24282621 PMCID: PMC3840058 DOI: 10.1371/journal.pone.0082012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 10/27/2013] [Indexed: 01/21/2023] Open
Abstract
Selenocysteine is the 21st naturally-occurring amino acid. Selenoproteins have diverse functions and many remain uncharacterized, but they are typically associated with antioxidant activity. The incorporation of selenocysteine into the nascent polypeptide chain recodes the TGA stop codon and this process depends upon a number of essential factors including the selenocysteine elongation factor (SEF). The transcriptional expression of SEF did not change significantly in tick midguts throughout the blood meal, but decreased in salivary glands to 20% at the end of the fast feeding phase. Since selenoprotein translation requires this specialized elongation factor, we targeted this gene for knockdown by RNAi to gain a global view of the role selenoproteins play in tick physiology. We found no significant differences in tick engorgement and embryogenesis but detected no antioxidant capacity in tick saliva. The transcriptional profile of selenoproteins in R. parkeri-infected Amblyomma maculatum revealed declined activity of selenoprotein M and catalase and increased activity of selenoprotein O, selenoprotein S, and selenoprotein T. Furthermore, the pathogen burden was significantly altered in SEF-knockdowns. We then determined the global impact of SEF-knockdown by RNA-seq, and mapped huge shifts in secretory gene expression that could be the result of downregulation of the Sin3 histone deacetylase corepressor complex.
Collapse
Affiliation(s)
- Steven W. Adamson
- Department of Biological Sciences, the University of Southern Mississippi, Hattiesburg, Mississippi, United States of America
| | - Rebecca E. Browning
- Department of Biological Sciences, the University of Southern Mississippi, Hattiesburg, Mississippi, United States of America
| | - Khemraj Budachetri
- Department of Biological Sciences, the University of Southern Mississippi, Hattiesburg, Mississippi, United States of America
| | - José M. C. Ribeiro
- Vector Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Shahid Karim
- Department of Biological Sciences, the University of Southern Mississippi, Hattiesburg, Mississippi, United States of America
- * E-mail:
| |
Collapse
|
176
|
Pakpour N, Camp L, Smithers HM, Wang B, Tu Z, Nadler SA, Luckhart S. Protein kinase C-dependent signaling controls the midgut epithelial barrier to malaria parasite infection in anopheline mosquitoes. PLoS One 2013; 8:e76535. [PMID: 24146884 PMCID: PMC3795702 DOI: 10.1371/journal.pone.0076535] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 09/01/2013] [Indexed: 11/19/2022] Open
Abstract
Anopheline mosquitoes are the primary vectors of parasites in the genus Plasmodium, the causative agents of malaria. Malaria parasites undergo a series of complex transformations upon ingestion by the mosquito host. During this process, the physical barrier of the midgut epithelium, along with innate immune defenses, functionally restrict parasite development. Although these defenses have been studied for some time, the regulatory factors that control them are poorly understood. The protein kinase C (PKC) gene family consists of serine/threonine kinases that serve as central signaling molecules and regulators of a broad spectrum of cellular processes including epithelial barrier function and immunity. Indeed, PKCs are highly conserved, ranging from 7 isoforms in Drosophila to 16 isoforms in mammals, yet none have been identified in mosquitoes. Despite conservation of the PKC gene family and their potential as targets for transmission-blocking strategies for malaria, no direct connections between PKCs, the mosquito immune response or epithelial barrier integrity are known. Here, we identify and characterize six PKC gene family members--PKCδ, PKCε, PKCζ, PKD, PKN, and an indeterminate conventional PKC--in Anopheles gambiae and Anopheles stephensi. Sequence and phylogenetic analyses of the anopheline PKCs support most subfamily assignments. All six PKCs are expressed in the midgut epithelia of A. gambiae and A. stephensi post-blood feeding, indicating availability for signaling in a tissue that is critical for malaria parasite development. Although inhibition of PKC enzymatic activity decreased NF-κB-regulated anti-microbial peptide expression in mosquito cells in vitro, PKC inhibition had no effect on expression of a panel of immune genes in the midgut epithelium in vivo. PKC inhibition did, however, significantly increase midgut barrier integrity and decrease development of P. falciparum oocysts in A. stephensi, suggesting that PKC-dependent signaling is a negative regulator of epithelial barrier function and a potential new target for transmission-blocking strategies.
Collapse
Affiliation(s)
- Nazzy Pakpour
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, California, United States of America
| | - Lauren Camp
- Department of Entomology and Nematology, University of California Davis, Davis, California, United States of America
| | - Hannah M. Smithers
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, California, United States of America
| | - Bo Wang
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, California, United States of America
| | - Zhijian Tu
- Department of Biochemistry, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, United States of America
| | - Steven A. Nadler
- Department of Entomology and Nematology, University of California Davis, Davis, California, United States of America
| | - Shirley Luckhart
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, California, United States of America
| |
Collapse
|
177
|
Cheng D, Lane RS, Moore BD, Zhong J. Host blood meal-dependent growth ensures transovarial transmission and transstadial passage of Rickettsia sp. phylotype G021 in the western black-legged tick (Ixodes pacificus). Ticks Tick Borne Dis 2013; 4:421-6. [PMID: 23876278 DOI: 10.1016/j.ttbdis.2013.04.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 04/19/2013] [Accepted: 04/19/2013] [Indexed: 02/04/2023]
Abstract
In this study, we explored the growth dynamics of Rickettsia sp. phylotype G021 during transovarial transmission and transstadial passage by Ixodes pacificus using real-time quantitative PCR. Four parental engorged I. pacificus females were allowed to complete their developmental stages until the F2-generation eggs yielded unfed larvae. All eggs, larvae, nymphs, and adults tested through 2 generations were found to be infected with phylotype G021. Hence, we conclude that the efficiency of transovarial transmission and transstadial passage of this phylotype in I. pacificus was 100%. Acquisition of a blood meal by all 3 parasitic stages (larva, nymph, adult) significantly increased the rickettsial burden as fed larvae, nymphs, and adults had respective 19-, 12-, and 313-fold increases of rickettsiae compared with unfed ticks representing each developmental stage. I. pacificus eggs contained high rickettsial burdens at the time of oviposition. While I. pacificus egg cells underwent rapid proliferation during early embryonic development, the rickettsiae remained relatively quiescent, which resulted in depressed numbers of phylotype G021 per tick cell. However, the rickettsial burden remained constant over a period of 56 days, as the rate of I. pacificus cell division slowed during later embryonic development.
Collapse
Affiliation(s)
- Du Cheng
- Department of Biological Sciences, Humboldt State University, Arcata, CA 95521, USA
| | | | | | | |
Collapse
|
178
|
Hajdušek O, Síma R, Ayllón N, Jalovecká M, Perner J, de la Fuente J, Kopáček P. Interaction of the tick immune system with transmitted pathogens. Front Cell Infect Microbiol 2013; 3:26. [PMID: 23875177 PMCID: PMC3712896 DOI: 10.3389/fcimb.2013.00026] [Citation(s) in RCA: 181] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 06/13/2013] [Indexed: 12/04/2022] Open
Abstract
Ticks are hematophagous arachnids transmitting a wide variety of pathogens including viruses, bacteria, and protozoans to their vertebrate hosts. The tick vector competence has to be intimately linked to the ability of transmitted pathogens to evade tick defense mechanisms encountered on their route through the tick body comprising midgut, hemolymph, salivary glands or ovaries. Tick innate immunity is, like in other invertebrates, based on an orchestrated action of humoral and cellular immune responses. The direct antimicrobial defense in ticks is accomplished by a variety of small molecules such as defensins, lysozymes or by tick-specific antimicrobial compounds such as microplusin/hebraein or 5.3-kDa family proteins. Phagocytosis of the invading microbes by tick hemocytes is likely mediated by the primordial complement-like system composed of thioester-containing proteins, fibrinogen-related lectins and convertase-like factors. Moreover, an important role in survival of the ingested microbes seems to be played by host proteins and redox balance maintenance in the tick midgut. Here, we summarize recent knowledge about the major components of tick immune system and focus on their interaction with the relevant tick-transmitted pathogens, represented by spirochetes (Borrelia), rickettsiae (Anaplasma), and protozoans (Babesia). Availability of the tick genomic database and feasibility of functional genomics based on RNA interference greatly contribute to the understanding of molecular and cellular interplay at the tick-pathogen interface and may provide new targets for blocking the transmission of tick pathogens.
Collapse
Affiliation(s)
- Ondřej Hajdušek
- Biological Centre ASCR, Institute of Parasitology České Budějovice, Czech Republic
| | | | | | | | | | | | | |
Collapse
|
179
|
Garcia-Garcia E, Galindo-Villegas J, Mulero V. Mucosal immunity in the gut: the non-vertebrate perspective. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2013; 40:278-288. [PMID: 23537860 DOI: 10.1016/j.dci.2013.03.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 03/07/2013] [Accepted: 03/14/2013] [Indexed: 06/02/2023]
Abstract
Much is now known about the vertebrate mechanisms involved in mucosal immunity, and the requirement of commensal microbiota at mucosal surfaces for the proper functioning of the immune system. In comparison, very little is known about the mechanisms of immunity at the barrier epithelia of non-vertebrate organisms. The purpose of this review is to summarize key experimental evidence illustrating how non-vertebrate immune mechanisms at barrier epithelia compare to those of higher vertebrates, using the gut as a model organ. Not only effector mechanisms of gut immunity are similar between vertebrates and non-vertebrates, but it also seems that the proper functioning of non-vertebrate gut defense mechanisms requires the presence of a resident microbiota. As more information becomes available, it will be possible to obtain a more accurate picture of how mucosal immunity has evolved, and how it adapts to the organisms' life styles.
Collapse
Affiliation(s)
- Erick Garcia-Garcia
- Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, Campus Universitario de Espinardo, 30100 Murcia, Spain.
| | | | | |
Collapse
|
180
|
Minard G, Mavingui P, Moro CV. Diversity and function of bacterial microbiota in the mosquito holobiont. Parasit Vectors 2013; 6:146. [PMID: 23688194 PMCID: PMC3667145 DOI: 10.1186/1756-3305-6-146] [Citation(s) in RCA: 266] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 05/08/2013] [Indexed: 11/15/2022] Open
Abstract
Mosquitoes (Diptera: Culicidae) have been shown to host diverse bacterial communities that vary depending on the sex of the mosquito, the developmental stage, and ecological factors. Some studies have suggested a potential role of microbiota in the nutritional, developmental and reproductive biology of mosquitoes. Here, we present a review of the diversity and functions of mosquito-associated bacteria across multiple variation factors, emphasizing recent findings. Mosquito microbiota is considered in the context of possible extended phenotypes conferred on the insect hosts that allow niche diversification and rapid adaptive evolution in other insects. These kinds of observations have prompted the recent development of new mosquito control methods based on the use of symbiotically-modified mosquitoes to interfere with pathogen transmission or reduce the host life span and reproduction. New opportunities for exploiting bacterial function for vector control are highlighted.
Collapse
Affiliation(s)
- Guillaume Minard
- UMR CNRS 5557, USC INRA 1364, VetAgro Sup, Ecologie Microbienne, FR41 BioEnvironment and Health, Université de Lyon 1, Villeurbanne F-69622, France
| | | | | |
Collapse
|
181
|
The role of reactive oxygen species in Anopheles aquasalis response to Plasmodium vivax infection. PLoS One 2013; 8:e57014. [PMID: 23441231 PMCID: PMC3575503 DOI: 10.1371/journal.pone.0057014] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 01/17/2013] [Indexed: 12/22/2022] Open
Abstract
Malaria affects millions of people worldwide and hundreds of thousands of people each year in Brazil. The mosquito Anopheles aquasalis is an important vector of Plasmodium vivax, the main human malaria parasite in the Americas. Reactive oxygen species (ROS) have been shown to have a role in insect innate immune responses as a potent pathogen-killing agent. We investigated the mechanisms of free radicals modulation after A. aquasalis infection with P. vivax. ROS metabolism was evaluated in the vector by studying expression and activity of three key detoxification enzymes, one catalase and two superoxide dismutases (SOD3A and SOD3B). Also, the involvement of free radicals in the mosquito immunity was measured by silencing the catalase gene followed by infection of A. aquasalis with P. vivax. Catalase, SOD3A and SOD3B expression in whole A. aquasalis were at the same levels of controls at 24 h and upregulated 36 h after ingestion of blood containing P. vivax. However, in the insect isolated midgut, the mRNA for these enzymes was not regulated by P. vivax infection, while catalase activity was reduced 24 h after the infectious meal. RNAi-mediated silencing of catalase reduced enzyme activity in the midgut, resulted in increased P. vivax infection and prevalence, and decreased bacterial load in the mosquito midgut. Our findings suggest that the interactions between A. aquasalis and P. vivax do not follow the model of ROS-induced parasite killing. It appears that P. vivax manipulates the mosquito detoxification system in order to allow its own development. This can be an indirect effect of fewer competitive bacteria present in the mosquito midgut caused by the increase of ROS after catalase silencing. These findings provide novel information on unique aspects of the main malaria parasite in the Americas interaction with one of its natural vectors.
Collapse
|
182
|
Turturice BA, Lamm MA, Tasch JJ, Zalewski A, Kooistra R, Schroeter EH, Sharma S, Kawazu SI, Kanzok SM. Expression of cytosolic peroxiredoxins in Plasmodium berghei ookinetes is regulated by environmental factors in the mosquito bloodmeal. PLoS Pathog 2013; 9:e1003136. [PMID: 23382676 PMCID: PMC3561267 DOI: 10.1371/journal.ppat.1003136] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2012] [Accepted: 11/29/2012] [Indexed: 11/19/2022] Open
Abstract
The Plasmodium ookinete develops over several hours in the bloodmeal of its mosquito vector where it is exposed to exogenous stresses, including cytotoxic reactive oxygen species (ROS). How the parasite adapts to these challenging conditions is not well understood. We have systematically investigated the expression of three cytosolic antioxidant proteins, thioredoxin-1 (Trx-1), peroxiredoxin-1 (TPx-1), and 1-Cys peroxiredoxin (1-Cys Prx), in developing ookinetes of the rodent parasite Plasmodium berghei under various growth conditions. Transcriptional profiling showed that tpx-1 and 1-cys prx but not trx-1 are more strongly upregulated in ookinetes developing in the mosquito bloodmeal when compared to ookinetes growing under culture conditions. Confocal immunofluorescence imaging revealed comparable expression patterns on the corresponding proteins. 1-Cys Prx in particular exhibited strong expression in mosquito-derived ookinetes but was not detectable in cultured ookinetes. Furthermore, ookinetes growing in culture upregulated tpx-1 and 1-cys prx when challenged with exogenous ROS in a dose-dependent fashion. This suggests that environmental factors in the mosquito bloodmeal induce upregulation of cytosolic antioxidant proteins in Plasmodium ookinetes. We found that in a parasite line lacking TPx-1 (TPx-1KO), expression of 1-Cys Prx occurred significantly earlier in mosquito-derived TPx-1KO ookinetes when compared to wild type (WT) ookinetes. The protein was also readily detectable in cultured TPx-1KO ookinetes, indicating that 1-Cys Prx at least in part compensates for the loss of TPx-1 in vivo. We hypothesize that this dynamic expression of the cytosolic peroxiredoxins reflects the capacity of the developing Plasmodium ookinete to rapidly adapt to the changing conditions in the mosquito bloodmeal. This would significantly increase its chances of survival, maturation and subsequent escape. Our results also emphasize that environmental conditions must be taken into account when investigating Plasmodium-mosquito interactions. The malaria parasite Plasmodium is transmitted by Anopheles mosquitoes. Within the midgut of the insect, it is exposed to multiple environmental stresses, including cytotoxic reactive oxygen species (ROS). To avoid destruction, the parasite develops into a motile ookinete capable of leaving the midgut. Yet, ookinete development lasts over several hours and requires the parasite to adapt to an increasingly challenging environment. Here we show that ookinetes of the rodent parasite Plasmodium berghei during development in the mosquito midgut increase the expression of the protective antioxidant proteins peroxiredoxin-1 (TPx-1) and 1-Cys peroxiredoxin (1-Cys Prx). This upregulation was also inducible in cultured ookinetes by challenging them with ROS. This suggests that ookinetes actively modulate the expression of their antioxidant proteins in response to the changing conditions in the mosquito. We also found that ookinetes lacking TPx-1 (TPx-1KO) upregulated 1-Cys Prx expression significantly earlier than wild type ookinetes. This indicates that the TPx-1KO parasites compensate for the loss of TPx-1 by altering the expression pattern of the functionally related 1-Cys Prx. The observed dynamic regulation of the cytosolic antioxidant proteins may help the Plasmodium ookinete to adapt to rapidly changing environmental conditions and thus to increase the probability of survival, maturation and escape from the mosquito midgut.
Collapse
Affiliation(s)
- Benjamin A. Turturice
- Department of Biology, Loyola University Chicago, Chicago, Illinois, United States of America
| | - Michael A. Lamm
- Department of Biology, Loyola University Chicago, Chicago, Illinois, United States of America
| | - James J. Tasch
- Department of Biology, Loyola University Chicago, Chicago, Illinois, United States of America
| | - Angelika Zalewski
- Department of Biology, Loyola University Chicago, Chicago, Illinois, United States of America
| | - Rachel Kooistra
- Department of Biology, Loyola University Chicago, Chicago, Illinois, United States of America
| | - Eric H. Schroeter
- Department of Biology, Loyola University Chicago, Chicago, Illinois, United States of America
| | - Sapna Sharma
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Shin-Ichiro Kawazu
- Obihiro University of Agriculture and Veterinarian Medicine, National Research Center for Protozoan Diseases, Obihiro, Hokkaido, Japan
| | - Stefan M. Kanzok
- Department of Biology, Loyola University Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
183
|
Pakpour N, Akman-Anderson L, Vodovotz Y, Luckhart S. The effects of ingested mammalian blood factors on vector arthropod immunity and physiology. Microbes Infect 2013; 15:243-54. [PMID: 23370408 DOI: 10.1016/j.micinf.2013.01.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 01/14/2013] [Accepted: 01/15/2013] [Indexed: 12/11/2022]
Abstract
The blood feeding behavior of disease-transmitting arthropods creates a unique intersection between vertebrate and invertebrate physiology. Here, we review host blood-derived factors that persist through blood digestion to affect the lifespan, reproduction, and immune responses of some of the most common arthropod vectors of human disease.
Collapse
Affiliation(s)
- Nazzy Pakpour
- Department of Medical Microbiology and Immunology, University of California, Davis, CA 95616, United States.
| | | | | | | |
Collapse
|
184
|
Bacterial feeding, Leishmania infection and distinct infection routes induce differential defensin expression in Lutzomyia longipalpis. Parasit Vectors 2013; 6:12. [PMID: 23311993 PMCID: PMC3573903 DOI: 10.1186/1756-3305-6-12] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 12/14/2012] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Phlebotomine insects harbor bacterial, viral and parasitic pathogens that can cause diseases of public health importance. Lutzomyia longipalpis is the main vector of visceral leishmaniasis in the New World. Insects can mount a powerful innate immune response to pathogens. Defensin peptides take part in this response and are known to be active against Gram-positive and Gram-negative bacteria, and some parasites. We studied the expression of a defensin gene from Lutzomyia longipalpis to understand its role in sand fly immune response. METHODS We identified, sequenced and evaluated the expression of a L. longipalpis defensin gene by semi-quantitative RT-PCR. The gene sequence was compared to other vectors defensins and expression was determined along developmental stages and after exposure of adult female L. longipalpis to bacteria and Leishmania. RESULTS Phylogenetic analysis showed that the L. longipalpis defensin is closely related to a defensin from the Old World sand fly Phlebotomus duboscqi. Expression was high in late L4 larvae and pupae in comparison to early larval stages and newly emerged flies. Defensin expression was modulated by oral infection with bacteria. The Gram-positive Micrococcus luteus induced early high defensin expression, whilst the Gram-negative entomopathogenic Serratia marcescens induced a later response. Bacterial injection also induced defensin expression in adult insects. Female sand flies infected orally with Leishmania mexicana showed no significant difference in defensin expression compared to blood fed insects apart from a lower defensin expression 5 days post Leishmania infection. When Leishmania was introduced into the hemolymph by injection there was no induction of defensin expression until 72 h later. CONCLUSIONS Our results suggest that L. longipalpis modulates defensin expression upon bacterial and Leishmania infection, with patterns of expression that are distinct among bacterial species and routes of infection.
Collapse
|
185
|
Andrews ES, Crain PR, Fu Y, Howe DK, Dobson SL. Reactive oxygen species production and Brugia pahangi survivorship in Aedes polynesiensis with artificial Wolbachia infection types. PLoS Pathog 2012; 8:e1003075. [PMID: 23236284 PMCID: PMC3516568 DOI: 10.1371/journal.ppat.1003075] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 10/22/2012] [Indexed: 12/31/2022] Open
Abstract
Heterologous transinfection with the endosymbiotic bacterium Wolbachia has been shown previously to induce pathogen interference phenotypes in mosquito hosts. Here we examine an artificially infected strain of Aedes polynesiensis, the primary vector of Wuchereria bancrofti, which is the causative agent of Lymphatic filariasis (LF) throughout much of the South Pacific. Embryonic microinjection was used to transfer the wAlbB infection from Aedes albopictus into an aposymbiotic strain of Ae. polynesiensis. The resulting strain (designated "MTB") experiences a stable artificial infection with high maternal inheritance. Reciprocal crosses of MTB with naturally infected wild-type Ae. polynesiensis demonstrate strong bidirectional incompatibility. Levels of reactive oxygen species (ROS) in the MTB strain differ significantly relative to that of the wild-type, indicating an impaired ability to regulate oxidative stress. Following a challenge with Brugia pahangi, the number of filarial worms achieving the infective stage is significantly reduced in MTB as compared to the naturally infected and aposymbiotic strains. Survivorship of MTB differed significantly from that of the wild-type, with an interactive effect between survivorship and blood feeding. The results demonstrate a direct correlation between decreased ROS levels and decreased survival of adult female Aedes polynesiensis. The results are discussed in relation to the interaction of Wolbachia with ROS production and antioxidant expression, iron homeostasis and the insect immune system. We discuss the potential applied use of the MTB strain for impacting Ae. polynesiensis populations and strategies for reducing LF incidence in the South Pacific.
Collapse
Affiliation(s)
- Elizabeth S. Andrews
- Department of Entomology, College of Agriculture, University of Kentucky, Lexington, Kentucky, United States of America
| | - Philip R. Crain
- Department of Entomology, College of Agriculture, University of Kentucky, Lexington, Kentucky, United States of America
| | - Yuqing Fu
- Department of Entomology, College of Agriculture, University of Kentucky, Lexington, Kentucky, United States of America
- Tropical Research and Education Center, University of Florida, Homestead, Florida, United States of America
| | - Daniel K. Howe
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, Kentucky, United States of America
| | - Stephen L. Dobson
- Department of Entomology, College of Agriculture, University of Kentucky, Lexington, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
186
|
Analysis of candidate colitis genes in the Gdac1 locus of mice deficient in glutathione peroxidase-1 and -2. PLoS One 2012; 7:e44262. [PMID: 22970191 PMCID: PMC3435402 DOI: 10.1371/journal.pone.0044262] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2012] [Accepted: 07/31/2012] [Indexed: 12/21/2022] Open
Abstract
Background Mice that are deficient for glutathione peroxidases 1 and 2 (GPX) show large variations in the penetrance and severity of colitis in C57BL/6J and 129S1/SvImJ backgrounds. We mapped a locus contributing to this difference to distal chromosome 2 (∼119–133 mbp) and named it glutathione peroxidase-deficiency-associated colitis 1 (Gdac1). The aim of this study was to identify the best gene candidates within the Gdac1 locus contributing to the murine colitis phenotype. Method/Principal Findings We refined the boundaries of Gdac1 to 118–125 mbp (95% confidence interval) by increasing sample size and marker density across the interval. The narrowed region contains 128 well-annotated protein coding genes but it excludes Fermt1, a human inflammatory bowel disease candidate that was within the original boundaries of Gdac1. The locus we identified may be the Cdcs3 locus mapped by others studying IL10-knockout mice. Using in silico analysis of the 128 genes, based on published colon expression data, the relevance of pathways to colitis, gene mutations, presence of non-synonymous-single-nucleotide polymorphisms (nsSNPs) and whether the nsSNPs are predicted to have an impact on protein function or expression, we excluded 42 genes. Based on a similar analysis, twenty-five genes from the remaining 86 genes were analyzed for expression-quantitative-trait loci, and another 15 genes were excluded. Conclusion/Significance Among the remaining 10 genes, we identified Pla2g4f and Duox2 as the most likely colitis gene candidates, because GPX metabolizes PLA2G4F and DUOX2 products. Pla2g4f is a phospholipase A2 that has three potentially significant nsSNP variants and showed expression differences across mouse strains. PLA2G4F produces arachidonic acid, which is a substrate for lipoxygenases and, in turn, for GPXs. DUOX2 produces H2O2 and may control microbial populations. DUOX-1 and -2 control microbial populations in mammalian lung and in the gut of several insects and zebrafish. Dysbiosis is a phenotype that differentiates 129S1/SvImJ from C57BL/6J and may be due to strain differences in DUOX2 activity.
Collapse
|
187
|
Gonçalves RLS, Oliveira JHM, Oliveira GA, Andersen JF, Oliveira MF, Oliveira PL, Barillas-Mury C. Mitochondrial reactive oxygen species modulate mosquito susceptibility to Plasmodium infection. PLoS One 2012; 7:e41083. [PMID: 22815925 PMCID: PMC3399787 DOI: 10.1371/journal.pone.0041083] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Accepted: 06/18/2012] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Mitochondria perform multiple roles in cell biology, acting as the site of aerobic energy-transducing pathways and as an important source of reactive oxygen species (ROS) that modulate redox metabolism. METHODOLOGY/PRINCIPAL FINDINGS We demonstrate that a novel member of the mitochondrial transporter protein family, Anopheles gambiae mitochondrial carrier 1 (AgMC1), is required to maintain mitochondrial membrane potential in mosquito midgut cells and modulates epithelial responses to Plasmodium infection. AgMC1 silencing reduces mitochondrial membrane potential, resulting in increased proton-leak and uncoupling of oxidative phosphorylation. These metabolic changes reduce midgut ROS generation and increase A. gambiae susceptibility to Plasmodium infection. CONCLUSION We provide direct experimental evidence indicating that ROS derived from mitochondria can modulate mosquito epithelial responses to Plasmodium infection.
Collapse
Affiliation(s)
- Renata L. S. Gonçalves
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
- Laboratório de Bioquímica de Resposta ao Estresse, Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório de Inflamação e Metabolismo, Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jose Henrique M. Oliveira
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Giselle A. Oliveira
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - John F. Andersen
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Marcus F. Oliveira
- Laboratório de Bioquímica de Resposta ao Estresse, Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório de Inflamação e Metabolismo, Instituto Nacional de Ciência e Tecnologia de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro L. Oliveira
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, Brazil
| | - Carolina Barillas-Mury
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
- * E-mail:
| |
Collapse
|
188
|
Coggins SA, Estévez-Lao TY, Hillyer JF. Increased survivorship following bacterial infection by the mosquito Aedes aegypti as compared to Anopheles gambiae correlates with increased transcriptional induction of antimicrobial peptides. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2012; 37:390-401. [PMID: 22326457 DOI: 10.1016/j.dci.2012.01.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Revised: 01/24/2012] [Accepted: 01/25/2012] [Indexed: 05/31/2023]
Abstract
Mosquitoes defend themselves from pathogens by mounting cellular and humoral innate immune responses. Bioinformatic analyses have revealed considerable divergence in immune gene repertoires between mosquito species, but interspecies empirical comparisons of immune responses are lacking. Here, we present a comparative analysis of the antimicrobial responses of two distantly related disease vectors: Aedes aegypti and Anopheles gambiae. Survival studies showed that Ae. aegypti are more proficient in surviving a bacterial infection than An. gambiae, and this correlates with Ae. aegypti's superior ability to kill bacteria in their hemocoels. Hemocytes from both species swiftly phagocytose bacteria, but phagocytosis does not explain Ae. aegypti's increased robustness: An. gambiae contain more circulating hemocytes and display a higher phagocytic index, but the phagocytic capacity of individual hemocytes is greater in Ae. aegypti. Then, profiling of 19 immunity genes revealed that transcriptional induction following infection is significantly elevated in Ae. aegypti when compared to An. gambiae, with the largest change seen in the transcription of cecropin and defensin. These data show that Ae. aegypti is better equipped to survive a bacterial infection than An. gambiae, and this correlates with Ae. aegypti's increased transcriptional induction of antimicrobial peptides and other humoral immune factors in response to infection.
Collapse
Affiliation(s)
- Sarah A Coggins
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235-1634, USA
| | | | | |
Collapse
|
189
|
Lima VLA, Dias F, Nunes RD, Pereira LO, Santos TSR, Chiarini LB, Ramos TD, Silva-Mendes BJ, Perales J, Valente RH, Oliveira PL. The antioxidant role of xanthurenic acid in the Aedes aegypti midgut during digestion of a blood meal. PLoS One 2012; 7:e38349. [PMID: 22701629 PMCID: PMC3372515 DOI: 10.1371/journal.pone.0038349] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Accepted: 05/03/2012] [Indexed: 11/19/2022] Open
Abstract
In the midgut of the mosquito Aedes aegypti, a vector of dengue and yellow fever, an intense release of heme and iron takes place during the digestion of a blood meal. Here, we demonstrated via chromatography, light absorption and mass spectrometry that xanthurenic acid (XA), a product of the oxidative metabolism of tryptophan, is produced in the digestive apparatus after the ingestion of a blood meal and reaches milimolar levels after 24 h, the period of maximal digestive activity. XA formation does not occur in the White Eye (WE) strain, which lacks kynurenine hydroxylase and accumulates kynurenic acid. The formation of XA can be diminished by feeding the insect with 3,4-dimethoxy-N-[4-(3-nitrophenyl)thiazol-2-yl] benzenesulfonamide (Ro-61-8048), an inhibitor of XA biosynthesis. Moreover, XA inhibits the phospholipid oxidation induced by heme or iron. A major fraction of this antioxidant activity is due to the capacity of XA to bind both heme and iron, which occurs at a slightly alkaline pH (7.5-8.0), a condition found in the insect midgut. The midgut epithelial cells of the WE mosquito has a marked increase in occurrence of cell death, which is reversed to levels similar to the wild type mosquitoes by feeding the insects with blood supplemented with XA, confirming the protective role of this molecule. Collectively, these results suggest a new role for XA as a heme and iron chelator that provides protection as an antioxidant and may help these animals adapt to a blood feeding habit.
Collapse
Affiliation(s)
- Vitor L. A. Lima
- Instituto de Química e Biotecnologia, Universidade Federal de Alagoas, Maceió, Alagoas, Brazil
| | - Felipe Dias
- Instituto de Bioquímica Médica, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rodrigo D. Nunes
- Instituto de Bioquímica Médica, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luiza O. Pereira
- Laboratório Interdisciplinar de Pesquisas Médicas - Instituto Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tiago S. R. Santos
- Instituto de Bioquímica Médica, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana B. Chiarini
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tadeu D. Ramos
- Instituto de Bioquímica Médica, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bernardo J. Silva-Mendes
- Instituto de Bioquímica Médica, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jonas Perales
- Laboratório de Toxinologia, Instituto Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Richard H. Valente
- Laboratório de Toxinologia, Instituto Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro L. Oliveira
- Instituto de Bioquímica Médica, Programa de Biologia Molecular e Biotecnologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
190
|
Diaz-Albiter H, Sant'Anna MRV, Genta FA, Dillon RJ. Reactive oxygen species-mediated immunity against Leishmania mexicana and Serratia marcescens in the sand phlebotomine fly Lutzomyia longipalpis. J Biol Chem 2012; 287:23995-4003. [PMID: 22645126 DOI: 10.1074/jbc.m112.376095] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Phlebotomine sand flies are the vectors of medically important Leishmania. The Leishmania protozoa reside in the sand fly gut, but the nature of the immune response to the presence of Leishmania is unknown. Reactive oxygen species (ROS) are a major component of insect innate immune pathways regulating gut-microbe homeostasis. Here we show that the concentration of ROS increased in sand fly midguts after they fed on the insect pathogen Serratia marcescens but not after feeding on the Leishmania that uses the sand fly as a vector. Moreover, the Leishmania is sensitive to ROS either by oral administration of ROS to the infected fly or by silencing a gene that expresses a sand fly ROS-scavenging enzyme. Finally, the treatment of sand flies with an exogenous ROS scavenger (uric acid) altered the gut microbial homeostasis, led to an increased commensal gut microbiota, and reduced insect survival after oral infection with S. marcescens. Our study demonstrates a differential response of the sand fly ROS system to gut microbiota, an insect pathogen, and the Leishmania that utilize the sand fly as a vehicle for transmission between mammalian hosts.
Collapse
Affiliation(s)
- Hector Diaz-Albiter
- Vector Group, Liverpool School of Tropical Medicine, Liverpool L3 5QA, United Kingdom
| | | | | | | |
Collapse
|
191
|
Weiss BL, Maltz M, Aksoy S. Obligate symbionts activate immune system development in the tsetse fly. THE JOURNAL OF IMMUNOLOGY 2012; 188:3395-403. [PMID: 22368278 DOI: 10.4049/jimmunol.1103691] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Many insects rely on the presence of symbiotic bacteria for proper immune system function. However, the molecular mechanisms that underlie this phenomenon are poorly understood. Adult tsetse flies (Glossina spp.) house three symbiotic bacteria that are vertically transmitted from mother to offspring during this insect's unique viviparous mode of reproduction. Larval tsetse that undergo intrauterine development in the absence of their obligate mutualist, Wigglesworthia, exhibit a compromised immune system during adulthood. In this study, we characterize the immune phenotype of tsetse that develop in the absence of all of their endogenous symbiotic microbes. Aposymbiotic tsetse (Glossina morsitans morsitans [Gmm(Apo)]) present a severely compromised immune system that is characterized by the absence of phagocytic hemocytes and atypical expression of immunity-related genes. Correspondingly, these flies quickly succumb to infection with normally nonpathogenic Escherichia coli. The susceptible phenotype exhibited by Gmm(Apo) adults can be reversed when they receive hemocytes transplanted from wild-type donor flies prior to infection. Furthermore, the process of immune system development can be restored in intrauterine Gmm(Apo) larvae when their mothers are fed a diet supplemented with Wigglesworthia cell extracts. Our finding that molecular components of Wigglesworthia exhibit immunostimulatory activity within tsetse is representative of a novel evolutionary adaptation that steadfastly links an obligate symbiont with its host.
Collapse
Affiliation(s)
- Brian L Weiss
- Division of Epidemiology of Microbial Diseases, Department of Epidemiology and Public Health, Yale University School of Medicine, New Haven, CT 06520, USA.
| | | | | |
Collapse
|
192
|
Bonizzoni M, Dunn WA, Campbell CL, Olson KE, Marinotti O, James AA. Strain Variation in the Transcriptome of the Dengue Fever Vector, Aedes aegypti. G3 (BETHESDA, MD.) 2012; 2:103-14. [PMID: 22384387 PMCID: PMC3276191 DOI: 10.1534/g3.111.001107] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 11/15/2011] [Indexed: 11/18/2022]
Abstract
Studies of transcriptome dynamics provide a basis for understanding functional elements of the genome and the complexity of gene regulation. The dengue vector mosquito, Aedes aegypti, exhibits great adaptability to diverse ecological conditions, is phenotypically polymorphic, and shows variation in vectorial capacity to arboviruses. Previous genome sequencing showed richness in repetitive DNA and transposable elements that can contribute to genome plasticity. Population genetic studies revealed a varying degree of worldwide genetic polymorphism. However, the extent of functional genetic polymorphism across strains is unknown. The transcriptomes of three Ae. aegypti strains, Chetumal (CTM), Rexville D-Puerto Rico (Rex-D) and Liverpool (LVP), were compared. CTM is more susceptible than Rex- D to infection by dengue virus serotype 2. A total of 4188 transcripts exhibit either no or small variation (<2-fold) among sugar-fed samples of the three strains and between sugar- and blood-fed samples within each strain, corresponding most likely to genes encoding products necessary for vital functions. Transcripts enriched in blood-fed mosquitoes encode proteins associated with catalytic activities, molecular transport, metabolism of lipids, carbohydrates and amino acids, and functions related to blood digestion and the progression of the gonotropic cycle. Significant qualitative and quantitative differences were found in individual transcripts among strains including differential representation of paralogous gene products. The majority of immunity-associated transcripts decreased in accumulation after a bloodmeal and the results are discussed in relation to the different susceptibility of CTM and Rex-D mosquitoes to DENV2 infection.
Collapse
Affiliation(s)
| | - W. Augustine Dunn
- Department of Molecular Biology and Biochemistry, and
- Institute for Genomics and Bioinformatics, University of California, Irvine, California 92697
| | | | - Ken E. Olson
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado 80523
| | | | - Anthony A. James
- Department of Molecular Biology and Biochemistry, and
- Department of Microbiology and Molecular Genetics, University of California, California, Irvine 92697
| |
Collapse
|
193
|
Abstract
Bartonella spp. are facultative intracellular pathogens that employ a unique stealth infection strategy comprising immune evasion and modulation, intimate interaction with nucleated cells, and intraerythrocytic persistence. Infections with Bartonella are ubiquitous among mammals, and many species can infect humans either as their natural host or incidentally as zoonotic pathogens. Upon inoculation into a naive host, the bartonellae first colonize a primary niche that is widely accepted to involve the manipulation of nucleated host cells, e.g., in the microvasculature. Consistently, in vitro research showed that Bartonella harbors an ample arsenal of virulence factors to modulate the response of such cells, gain entrance, and establish an intracellular niche. Subsequently, the bacteria are seeded into the bloodstream where they invade erythrocytes and give rise to a typically asymptomatic intraerythrocytic bacteremia. While this course of infection is characteristic for natural hosts, zoonotic infections or the infection of immunocompromised patients may alter the path of Bartonella and result in considerable morbidity. In this review we compile current knowledge on the molecular processes underlying both the infection strategy and pathogenesis of Bartonella and discuss their connection to the clinical presentation of human patients, which ranges from minor complaints to life-threatening disease.
Collapse
Affiliation(s)
- Alexander Harms
- Focal Area Infection Biology, Biozentrum, University of Basel, Switzerland
| | | |
Collapse
|
194
|
Castillo J, Brown MR, Strand MR. Blood feeding and insulin-like peptide 3 stimulate proliferation of hemocytes in the mosquito Aedes aegypti. PLoS Pathog 2011; 7:e1002274. [PMID: 21998579 PMCID: PMC3188524 DOI: 10.1371/journal.ppat.1002274] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Accepted: 08/01/2011] [Indexed: 12/14/2022] Open
Abstract
All vector mosquito species must feed on the blood of a vertebrate host to produce eggs. Multiple cycles of blood feeding also promote frequent contacts with hosts, which enhance the risk of exposure to infectious agents and disease transmission. Blood feeding triggers the release of insulin-like peptides (ILPs) from the brain of the mosquito Aedes aegypti, which regulate blood meal digestion and egg formation. In turn, hemocytes serve as the most important constitutive defense in mosquitoes against pathogens that enter the hemocoel. Prior studies indicated that blood feeding stimulates hemocytes to increase in abundance, but how this increase in abundance is regulated is unknown. Here, we determined that phagocytic granulocytes and oenocytoids express the A. aegypti insulin receptor (AaMIR). We then showed that: 1) decapitation of mosquitoes after blood feeding inhibited hemocyte proliferation, 2) a single dose of insulin-like peptide 3 (ILP3) sufficient to stimulate egg production rescued proliferation, and 3) knockdown of the AaMIR inhibited ILP3 rescue activity. Infection studies indicated that increased hemocyte abundance enhanced clearance of the bacterium Escherichia coli at lower levels of infection. Surprisingly, however, non-blood fed females better survived intermediate and high levels of E. coli infection than blood fed females. Taken together, our results reveal a previously unrecognized role for the insulin signaling pathway in regulating hemocyte proliferation. Our results also indicate that blood feeding enhances resistance to E. coli at lower levels of infection but reduces tolerance at higher levels of infection. Mosquitoes are vectors of several important diseases of humans and other mammals including Dengue fever, malaria and filariasis. These diseases adversely affect worldwide health by killing or debilitating millions of individuals. The key feature of mosquito biology that makes them such important disease vectors is that adult females must feed on the blood of their vertebrate host(s) to produce eggs. In turn, repeated bouts of blood feeding and egg development elevate the risk of mosquitoes feeding on an infected host and transmitting a given pathogen from one individual to another. A key regulator of egg development following blood feeding is the release of insulin-like peptides from the mosquito brain. We have found that insulin-like peptides enhance production of immune cells (hemocytes) that serve as the first line of defense against infection. Conversely, the molecular pathways that regulate egg development and hemocyte proliferation reduce the ability of mosquitoes to tolerate a persistent systemic infection. Taken together, our results indicate that trade-offs exist between reproduction and immune defense in mosquitoes, which is a subject of fundamental interest to evolutionary biologists and of applied importance in understanding disease transmission.
Collapse
Affiliation(s)
- Julio Castillo
- Department of Entomology, University of Georgia, Athens, Georgia, United States of America
| | - Mark R. Brown
- Department of Entomology, University of Georgia, Athens, Georgia, United States of America
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, United States of America
| | - Michael R. Strand
- Department of Entomology, University of Georgia, Athens, Georgia, United States of America
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, United States of America
- * E-mail:
| |
Collapse
|
195
|
Dynamic gut microbiome across life history of the malaria mosquito Anopheles gambiae in Kenya. PLoS One 2011; 6:e24767. [PMID: 21957459 PMCID: PMC3177825 DOI: 10.1371/journal.pone.0024767] [Citation(s) in RCA: 386] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Accepted: 08/17/2011] [Indexed: 11/19/2022] Open
Abstract
The mosquito gut represents an ecosystem that accommodates a complex, intimately associated microbiome. It is increasingly clear that the gut microbiome influences a wide variety of host traits, such as fitness and immunity. Understanding the microbial community structure and its dynamics across mosquito life is a prerequisite for comprehending the symbiotic relationship between the mosquito and its gut microbial residents. Here we characterized gut bacterial communities across larvae, pupae and adults of Anopheles gambiae reared in semi-natural habitats in Kenya by pyrosequencing bacterial 16S rRNA fragments. Immatures and adults showed distinctive gut community structures. Photosynthetic Cyanobacteria were predominant in the larval and pupal guts while Proteobacteria and Bacteroidetes dominated the adult guts, with core taxa of Enterobacteriaceae and Flavobacteriaceae. At the adult stage, diet regime (sugar meal and blood meal) significantly affects the microbial structure. Intriguingly, blood meals drastically reduced the community diversity and favored enteric bacteria. Comparative genomic analysis revealed that the enriched enteric bacteria possess large genetic redox capacity of coping with oxidative and nitrosative stresses that are associated with the catabolism of blood meal, suggesting a beneficial role in maintaining gut redox homeostasis. Interestingly, gut community structure was similar in the adult stage between the field and laboratory mosquitoes, indicating that mosquito gut is a selective eco-environment for its microbiome. This comprehensive gut metatgenomic profile suggests a concerted symbiotic genetic association between gut inhabitants and host.
Collapse
|
196
|
Hughes GL, Koga R, Xue P, Fukatsu T, Rasgon JL. Wolbachia infections are virulent and inhibit the human malaria parasite Plasmodium falciparum in Anopheles gambiae. PLoS Pathog 2011; 7:e1002043. [PMID: 21625582 PMCID: PMC3098226 DOI: 10.1371/journal.ppat.1002043] [Citation(s) in RCA: 257] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Accepted: 03/14/2011] [Indexed: 11/18/2022] Open
Abstract
Endosymbiotic Wolbachia bacteria are potent modulators of pathogen infection and transmission in multiple naturally and artificially infected insect species, including important vectors of human pathogens. Anopheles mosquitoes are naturally uninfected with Wolbachia, and stable artificial infections have not yet succeeded in this genus. Recent techniques have enabled establishment of somatic Wolbachia infections in Anopheles. Here, we characterize somatic infections of two diverse Wolbachia strains (wMelPop and wAlbB) in Anopheles gambiae, the major vector of human malaria. After infection, wMelPop disseminates widely in the mosquito, infecting the fat body, head, sensory organs and other tissues but is notably absent from the midgut and ovaries. Wolbachia initially induces the mosquito immune system, coincident with initial clearing of the infection, but then suppresses expression of immune genes, coincident with Wolbachia replication in the mosquito. Both wMelPop and wAlbB significantly inhibit Plasmodium falciparum oocyst levels in the mosquito midgut. Although not virulent in non-bloodfed mosquitoes, wMelPop exhibits a novel phenotype and is extremely virulent for approximately 12-24 hours post-bloodmeal, after which surviving mosquitoes exhibit similar mortality trajectories to control mosquitoes. The data suggest that if stable transinfections act in a similar manner to somatic infections, Wolbachia could potentially be used as part of a strategy to control the Anopheles mosquitoes that transmit malaria.
Collapse
Affiliation(s)
- Grant L. Hughes
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
- The Johns Hopkins Malaria Research Institute, Baltimore, Maryland, United States of America
| | - Ryuichi Koga
- National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Ping Xue
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
- The Johns Hopkins Malaria Research Institute, Baltimore, Maryland, United States of America
| | - Takema Fukatsu
- National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Jason L. Rasgon
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
- The Johns Hopkins Malaria Research Institute, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
197
|
|