1
|
Benoit JB, Weaving H, McLellan C, Terblanche JS, Attardo GM, English S. Viviparity and obligate blood feeding: tsetse flies as a unique research system to study climate change. CURRENT OPINION IN INSECT SCIENCE 2025; 69:101369. [PMID: 40122517 DOI: 10.1016/j.cois.2025.101369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Tsetse flies (Glossina species) are unique organisms that combine several remarkable traits: they are obligate blood feeders, serve as critical vectors for African trypanosomes, and reproduce through adenotrophic viviparity - a process in which offspring are nourished with milk-like secretions before being born live. Here, we explore how climate change will impact the physiological processes associated with live birth in tsetse. This includes considerations of how blood feeding, host-pathogen interactions, and host-symbiont dynamics are likely to be impacted by thermal shifts. The highly specialized biology of tsetse flies suggests that this system is likely to have a distinctive response to climate change. Thus, detailed empirical research into these unique features is paramount for predicting tsetse population dynamics under climate change, with caution required when generalizing from other well-studied vectors with contrasting ecology and life histories such as mosquitoes and ticks. At the same time, the reproductive biology of tsetse, as well as microbiome and feeding dynamics, allow for a powerful model to investigate climate change through the lens of pregnancy and associated physiological adaptations in an extensively researched invertebrate.
Collapse
Affiliation(s)
- Joshua B Benoit
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH, United States
| | - Hester Weaving
- School of Biological Sciences, University of Bristol, Bristol, United Kingdom; Department of Pathology, Microbiology & Immunology, University of California Davis, Davis, CA, United States
| | - Callum McLellan
- School of Biological Sciences, University of Bristol, Bristol, United Kingdom
| | - John S Terblanche
- Centre for Invasion Biology, Department of Conservation Ecology & Entomology, Stellenbosch University, Stellenbosch, South Africa
| | - Geoffrey M Attardo
- Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California Davis, Davis, CA, United States
| | - Sinead English
- School of Biological Sciences, University of Bristol, Bristol, United Kingdom.
| |
Collapse
|
2
|
Etzensperger R, Benninger M, Pozzi B, Rehmann R, Naguleswaran A, Schumann G, Van Den Abbeele J, Roditi I. Split-Cre-mediated GFP expression as a permanent marker for flagellar fusion of Trypanosoma brucei in its tsetse fly host. mBio 2025; 16:e0337524. [PMID: 39688410 PMCID: PMC11796343 DOI: 10.1128/mbio.03375-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Trypanosomes have different ways of communicating with each other. While communication via quorum sensing, or by the release and uptake of extracellular vesicles, is widespread in nature, the phenomenon of flagellar fusion has only been observed in Trypanosoma brucei. We showed previously that a small proportion of procyclic culture forms (corresponding to insect midgut forms) can fuse their flagella and exchange cytosolic and membrane proteins. This happens reproducibly in cell culture. It was not known, however, if flagellar fusion also occurs in the tsetse fly host, and at what stage of the life cycle. We have developed a split-Cre-Lox system to permanently label trypanosomes that undergo flagellar fusion. Specifically, we engineered trypanosomes to contain a GFP gene flanked by Lox sites in the reverse orientation to the promoter. In addition, the cells expressed inactive halves of the Cre recombinase, either N-terminal Cre residues 1-244 (N-Cre) or C-terminal Cre residues 245-343 (C-Cre). Upon flagellar fusion, these Cre halves were exchanged between trypanosomes, forming functional full Cre and flipping reverse-GFP into its forward orientation. We showed that cells that acquired the second half Cre through flagellar fusion were permanently modified and that the cells and their progeny constitutively expressed GFP. When tsetse flies were co-infected with N-Cre and C-Cre cells, GFP-positive trypanosomes were observed in the midgut and proventriculus 28-34 days post-infection. These results show that flagellar fusion not only happens in culture but also during the natural life cycle of trypanosomes in their tsetse fly host. IMPORTANCE We have established a procedure to permanently label pairs of trypanosomes that transiently fuse their flagella and exchange proteins. When this occurs, a reporter gene is permanently flipped from the "off" to the "on" position, resulting in the production of green fluorescent protein. Crucially, green trypanosomes can be detected in tsetse flies co-infected with the two cell lines, proving that flagellar fusion occurs in the host. To our knowledge, we are the first to describe a split-Cre-Lox system for lineage tracing and selection in trypanosomes. In addition to its use in trypanosomes, this system could be adapted for other parasites and in other contexts. For example, it could be used to determine whether flagellar fusion occurs in related parasites such as Leishmania and Trypanosoma cruzi or to monitor whether intracellular parasites and their hosts exchange proteins.
Collapse
Affiliation(s)
| | | | - Berta Pozzi
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Ruth Rehmann
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | | | | | - Jan Van Den Abbeele
- Department of Biomedical Sciences, Trypanosoma Unit, Institute of Tropical Medicine, Antwerp, Belgium
| | - Isabel Roditi
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| |
Collapse
|
3
|
Awuoche EO, Smallenberger G, Bruzzese DL, Orfano A, Weiss BL, Aksoy S. Spiroplasma endosymbiont reduction of host lipid synthesis and Stomoxyn-like peptide contribute to trypanosome resistance in the tsetse fly Glossina fuscipes. PLoS Pathog 2025; 21:e1012692. [PMID: 39888974 PMCID: PMC11819587 DOI: 10.1371/journal.ppat.1012692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 02/12/2025] [Accepted: 01/15/2025] [Indexed: 02/02/2025] Open
Abstract
Tsetse flies (Glossina spp.) vector African trypanosomes that cause devastating diseases in humans and domestic animals. Within the Glossina genus, species in the Palpalis subgroup exhibit greater resistance to trypanosome infections compared to those in the Morsitans subgroup. Varying microbiota composition and species-specific genetic traits can significantly influence the efficiency of parasite transmission. Notably, infections with the endosymbiotic bacterium Spiroplasma have been documented in several Palpalis subgroup species, including Glossina fuscipes fuscipes (Gff). While Spiroplasma infections in Gff are known to hinder trypanosome transmission, the underlying mechanisms remain unknown. To investigate Spiroplasma-mediated factors affecting Gff vector competence, we conducted high-throughput RNA sequencing of the gut tissue along with functional assays. Our findings reveal elevated oxidative stress in the gut environment in the presence of Spiroplasma, evidenced by increased expression of nitric oxide synthase, which catalyzes the production of trypanocidal nitric oxide. Additionally, we observed impaired lipid biosynthesis leading to a reduction of this important class of nutrients essential for parasite and host physiologies. In contrast, trypanosome infections in Gff's midgut significantly upregulated various immunity-related genes, including a small peptide, Stomoxyn-like, homologous to Stomoxyn first discovered in the stable fly, Stomoxys calcitrans. We observed that the Stomoxyn-like locus is exclusive to the genomes of Palpalis subgroup tsetse species. GffStomoxyn is constitutively expressed in the cardia (proventriculus) and synthetic GffStomoxyn exhibits potent activity against Escherichia coli and bloodstream form of Trypanosoma brucei parasites, while showing no effect against insect stage procyclic forms or tsetse's commensal endosymbiont Sodalis in vitro. Reducing GffStomoxyn levels significantly increased trypanosome infection prevalence, indicating its potential trypanocidal role in vivo. Collectively, our results suggest that the enhanced resistance to trypanosomes observed in Spiroplasma-infected Gff may be due to the reduced lipid availability necessary for parasite metabolic maintenance. Furthermore, GffStomoxyn could play a crucial role in the initial immune response(s) against mammalian parasites early in the infection process in the gut and prevent gut colonization. We discuss the molecular characteristics of GffStomoxyn, its spatial and temporal expression regulation and its microbicidal activity against Trypanosome parasites. Our findings reinforce the nutritional influences of microbiota on host physiology and host-pathogen dynamics.
Collapse
Affiliation(s)
- Erick O. Awuoche
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Gretchen Smallenberger
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Daniel L. Bruzzese
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Alessandra Orfano
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Brian L. Weiss
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Serap Aksoy
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
| |
Collapse
|
4
|
Beaver AK, Keneskhanova Z, Cosentino RO, Weiss BL, Awuoche EO, Smallenberger GM, Buenconsejo GY, Crilly NP, Smith JE, Hakim JMC, Zhang B, Bobb B, Rijo-Ferreira F, Figueiredo LM, Aksoy S, Siegel TN, Mugnier MR. Tissue spaces are reservoirs of antigenic diversity for Trypanosoma brucei. Nature 2024; 636:430-437. [PMID: 39478231 PMCID: PMC11634766 DOI: 10.1038/s41586-024-08151-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 10/03/2024] [Indexed: 11/18/2024]
Abstract
The protozoan parasite Trypanosoma brucei evades clearance by the host immune system through antigenic variation of its dense variant surface glycoprotein (VSG) coat, periodically 'switching' expression of the VSG using a large genomic repertoire of VSG-encoding genes1-6. Recent studies of antigenic variation in vivo have focused near exclusively on parasites in the bloodstream6-8, but research has shown that many, if not most, parasites reside in the interstitial spaces of tissues9-13. We sought to explore the dynamics of antigenic variation in extravascular parasite populations using VSG-seq7, a high-throughput sequencing approach for profiling VSGs expressed in populations of T. brucei. Here we show that tissues, not the blood, are the primary reservoir of antigenic diversity during both needle- and tsetse bite-initiated T. brucei infections, with more than 75% of VSGs found exclusively within extravascular spaces. We found that this increased diversity is correlated with slower parasite clearance in tissue spaces. Together, these data support a model in which the slower immune response in extravascular spaces provides more time to generate the antigenic diversity needed to maintain a chronic infection. Our findings reveal the important role that extravascular spaces can have in pathogen diversification.
Collapse
Affiliation(s)
- Alexander K Beaver
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Zhibek Keneskhanova
- Division of Experimental Parasitology, Faculty of Veterinary Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
- Biomedical Center Munich, Division of Physiological Chemistry, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Raúl O Cosentino
- Division of Experimental Parasitology, Faculty of Veterinary Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
- Biomedical Center Munich, Division of Physiological Chemistry, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Brian L Weiss
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Erick O Awuoche
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Gretchen M Smallenberger
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Gracyn Y Buenconsejo
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Nathan P Crilly
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jaclyn E Smith
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jill M C Hakim
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Bailin Zhang
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Bryce Bobb
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Filipa Rijo-Ferreira
- Division of Infectious Diseases and Vaccinology, Berkeley Public Health Molecular and Cell Biology Department, Berkeley, CA, USA
| | | | - Serap Aksoy
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - T Nicolai Siegel
- Division of Experimental Parasitology, Faculty of Veterinary Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
- Biomedical Center Munich, Division of Physiological Chemistry, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Monica R Mugnier
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
5
|
Awuoche E, Smallenberger G, Bruzzese D, Orfano A, Weiss BL, Aksoy S. Spiroplasma endosymbiont reduction of host lipid synthesis and Stomoxyn-like peptide contribute to trypanosome resistance in the tsetse fly Glossina fuscipes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.24.620045. [PMID: 39484388 PMCID: PMC11527105 DOI: 10.1101/2024.10.24.620045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Tsetse flies (Glossina spp.) vector African trypanosomes that cause devastating diseases in humans and domestic animals. Within the Glossina genus, species in the Palpalis subgroup exhibit greater resistance to trypanosome infections compared to those in the Morsitans subgroup. Varying microbiota composition and species-specific genetic traits can significantly influence the efficiency of parasite transmission. Notably, infections with the endosymbiotic bacterium Spiroplasma have been documented in several Palpalis subgroup species, including Glossina fuscipes fuscipes (Gff). While Spiroplasma infections in Gff are known to hinder trypanosome transmission, the underlying mechanisms remain unknown. To investigate Spiroplasma-mediated factors affecting Gff vector competence, we conducted high-throughput RNA sequencing of the midgut tissue along with functional assays. Our findings reveal elevated oxidative stress in the midgut environment in the presence of Spiroplasma, evidenced by increased expression of nitric oxide synthase, which catalyzes the production of trypanocidal nitric oxide. Additionally, we observed impaired lipid biosynthesis leading to a reduction of this important class of nutrients essential for parasite and host physiologies. In contrast, trypanosome infections in Gff's midgut significantly upregulated various immunity-related genes, including a small peptide, Stomoxyn-like, homologous to Stomoxyns first discovered in the stable fly Stomoxys calcitrans. We observed that the Stomoxyn-like locus is exclusive to the genomes of Palpalis subgroup tsetse species. GffStomoxyn is constitutively expressed in the cardia (proventriculus) and synthetic GffStomoxyn exhibits potent activity against Escherichia coli and bloodstream form of Trypanosoma brucei parasites, while showing no effect against insect stage procyclic forms or tsetse's commensal endosymbiont Sodalis in vitro. Reducing GffStomoxyn levels significantly increased trypanosome infection prevalence, indicating its potential trypanocidal role in vivo. Collectively, our results suggest that the enhanced resistance to trypanosomes observed in Spiroplasma-infected Gff may be due to the reduced lipid availability necessary for parasite metabolic maintenance. Furthermore, GffStomoxyn could play a crucial role in the initial immune response(s) against mammalian parasites early in the infection process in the midgut and prevent gut colonization. We discuss the molecular characteristics of GffStomoxyn, its spatial and temporal expression regulation and its microbicidal activity against Trypanosome parasites. Our findings reinforce the nutritional influences of microbiota on host physiology and host-pathogen dynamics.
Collapse
Affiliation(s)
- Erick Awuoche
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Gretchen Smallenberger
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Daniel Bruzzese
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Alessandra Orfano
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Brian L Weiss
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Serap Aksoy
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| |
Collapse
|
6
|
Peacock L, Kay C, Bailey M, Gibson W. Experimental genetic crosses in tsetse flies of the livestock pathogen Trypanosoma congolense savannah. Parasit Vectors 2024; 17:4. [PMID: 38178172 PMCID: PMC10765672 DOI: 10.1186/s13071-023-06105-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 12/17/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND In tropical Africa animal trypanosomiasis is a disease that has severe impacts on the health and productivity of livestock in tsetse fly-infested regions. Trypanosoma congolense savannah (TCS) is one of the main causative agents and is widely distributed across the sub-Saharan tsetse belt. Population genetics analysis has shown that TCS is genetically heterogeneous and there is evidence for genetic exchange, but to date Trypanosoma brucei is the only tsetse-transmitted trypanosome with experimentally proven capability to undergo sexual reproduction, with meiosis and production of haploid gametes. In T. brucei sex occurs in the fly salivary glands, so by analogy, sex in TCS should occur in the proboscis, where the corresponding portion of the developmental cycle takes place. Here we test this prediction using genetically modified red and green fluorescent clones of TCS. METHODS Three fly-transmissible strains of TCS were transfected with genes for red or green fluorescent protein, linked to a gene for resistance to the antibiotic hygromycin, and experimental crosses were set up by co-transmitting red and green fluorescent lines in different combinations via tsetse flies, Glossina pallidipes. To test whether sex occurred in vitro, co-cultures of attached epimastigotes of one red and one green fluorescent TCS strain were set up and sampled at intervals for 28 days. RESULTS All interclonal crosses of genetically modified trypanosomes produced hybrids containing both red and green fluorescent proteins, but yellow fluorescent hybrids were only present among trypanosomes from the fly proboscis, not from the midgut or proventriculus. It was not possible to identify the precise life cycle stage that undergoes mating, but it is probably attached epimastigotes in the food canal of the proboscis. Yellow hybrids were seen as early as 14 days post-infection. One intraclonal cross in tsetse and in vitro co-cultures of epimastigotes also produced yellow hybrids in small numbers. The hybrid nature of the yellow fluorescent trypanosomes observed was not confirmed by genetic analysis. CONCLUSIONS Despite absence of genetic characterisation of hybrid trypanosomes, the fact that these were produced only in the proboscis and in several independent crosses suggests that they are products of mating rather than cell fusion. The three-way strain compatibility observed is similar to that demonstrated previously for T. brucei, indicating that a simple two mating type system does not apply for either trypanosome species.
Collapse
Affiliation(s)
- Lori Peacock
- School of Biological Sciences, University of Bristol, Bristol, BS8 1TQ, UK
- Bristol Veterinary School, University of Bristol, Langford, Bristol, BS40 7DU, UK
| | - Chris Kay
- School of Biological Sciences, University of Bristol, Bristol, BS8 1TQ, UK
| | - Mick Bailey
- Bristol Veterinary School, University of Bristol, Langford, Bristol, BS40 7DU, UK
| | - Wendy Gibson
- School of Biological Sciences, University of Bristol, Bristol, BS8 1TQ, UK.
| |
Collapse
|
7
|
Peacock L, Kay C, Collett C, Bailey M, Gibson W. Development of the livestock pathogen Trypanosoma (Nannomonas) simiae in the tsetse fly with description of putative sexual stages from the proboscis. Parasit Vectors 2023; 16:231. [PMID: 37434196 DOI: 10.1186/s13071-023-05847-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 06/22/2023] [Indexed: 07/13/2023] Open
Abstract
BACKGROUND Tsetse-transmitted African animal trypanosomiasis is recognised as an important disease of ruminant livestock in sub-Saharan Africa, but also affects domestic pigs, with Trypanosoma simiae notable as a virulent suid pathogen that can rapidly cause death. Trypanosoma simiae is widespread in tsetse-infested regions, but its biology has been little studied compared to T. brucei and T. congolense. METHODS Trypanosoma simiae procyclics were cultured in vitro and transfected using protocols developed for T. brucei. Genetically modified lines, as well as wild-type trypanosomes, were transmitted through tsetse flies, Glossina pallidipes, to study T. simiae development in the tsetse midgut, proventriculus and proboscis. The development of proventricular trypanosomes was also studied in vitro. Image and mensural data were collected and analysed. RESULTS A PFR1::YFP line successfully completed development in tsetse, but a YFP::HOP1 line failed to progress beyond midgut infection. Analysis of image and mensural data confirmed that the vector developmental cycles of T. simiae and T. congolense are closely similar, but we also found putative sexual stages in T. simiae, as judged by morphological similarity to these stages in T. brucei. Putative meiotic dividers were abundant among T. simiae trypanosomes in the proboscis, characterised by a large posterior nucleus and two anterior kinetoplasts. Putative gametes and other meiotic intermediates were also identified by characteristic morphology. In vitro development of proventricular forms of T. simiae followed the pattern previously observed for T. congolense: long proventricular trypanosomes rapidly attached to the substrate and shortened markedly before commencing cell division. CONCLUSIONS To date, T. brucei is the only tsetse-transmitted trypanosome with experimentally proven capability to undergo sexual reproduction, which occurs in the fly salivary glands. By analogy, sexual stages of T. simiae or T. congolense are predicted to occur in the proboscis, where the corresponding portion of the developmental cycle takes place. While no such stages have been observed in T. congolense, for T. simiae putative sexual stages were abundant in the tsetse proboscis. Although our initial attempt to demonstrate expression of a YFP-tagged, meiosis-specific protein was unsuccessful, the future application of transgenic approaches will facilitate the identification of meiotic stages and hybrids in T. simiae.
Collapse
Affiliation(s)
- Lori Peacock
- School of Biological Sciences, University of Bristol, Bristol, BS8 1TQ, UK
- Bristol Veterinary School, University of Bristol, Langford, Bristol, BS40 7DU, UK
| | - Chris Kay
- School of Biological Sciences, University of Bristol, Bristol, BS8 1TQ, UK
| | - Clare Collett
- School of Biological Sciences, University of Bristol, Bristol, BS8 1TQ, UK
- Pathogen Immunology Group, UK Health Security Agency, Porton Down, Salisbury, SP4 0JG, Wiltshire, UK
| | - Mick Bailey
- Bristol Veterinary School, University of Bristol, Langford, Bristol, BS40 7DU, UK
| | - Wendy Gibson
- School of Biological Sciences, University of Bristol, Bristol, BS8 1TQ, UK.
| |
Collapse
|
8
|
Mirieri CK, Abd-Alla AM, Ros VI, van Oers MM. Evaluating the Effect of Irradiation on the Densities of Two RNA Viruses in Glossina morsitans morsitans. INSECTS 2023; 14:397. [PMID: 37103212 PMCID: PMC10140815 DOI: 10.3390/insects14040397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/16/2023] [Accepted: 04/18/2023] [Indexed: 06/19/2023]
Abstract
Tsetse flies are cyclic vectors of Trypanosoma parasites, which cause debilitating diseases in humans and animals. To decrease the disease burden, the number of flies is reduced using the sterile insect technique (SIT), where male flies are sterilized through irradiation and released into the field. This procedure requires the mass rearing of high-quality male flies able to compete with wild male flies for mating with wild females. Recently, two RNA viruses, an iflavirus and a negevirus, were discovered in mass-reared Glossina morsitans morsitans and named GmmIV and GmmNegeV, respectively. The aim of this study was to evaluate whether the densities of these viruses in tsetse flies are affected by the irradiation treatment. Therefore, we exposed tsetse pupae to various doses (0-150 Gy) of ionizing radiation, either in air (normoxia) or without air (hypoxia), for which oxygen was displaced by nitrogen. Pupae and/or emerging flies were collected immediately afterwards, and at three days post irradiation, virus densities were quantified through RT-qPCR. Generally, the results show that irradiation exposure had no significant impact on the densities of GmmIV and GmmNegeV, suggesting that the viruses are relatively radiation-resistant, even at higher doses. However, sampling over a longer period after irradiation would be needed to verify that densities of these insect viruses are not changed by the sterilisation treatment.
Collapse
Affiliation(s)
- Caroline K. Mirieri
- Laboratory of Virology, Wageningen University and Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
- Insect Pest Control Laboratory, Joint FAO/IAEA Programme of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency, Vienna International Centre, P.O. Box 100, 1400 Vienna, Austria;
| | - Adly M.M. Abd-Alla
- Insect Pest Control Laboratory, Joint FAO/IAEA Programme of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency, Vienna International Centre, P.O. Box 100, 1400 Vienna, Austria;
| | - Vera I.D. Ros
- Laboratory of Virology, Wageningen University and Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| | - Monique M. van Oers
- Laboratory of Virology, Wageningen University and Research, Droevendaalsesteeg 1, 6708 PB Wageningen, The Netherlands
| |
Collapse
|
9
|
Heme-deficient metabolism and impaired cellular differentiation as an evolutionary trade-off for human infectivity in Trypanosoma brucei gambiense. Nat Commun 2022; 13:7075. [PMID: 36400774 PMCID: PMC9674590 DOI: 10.1038/s41467-022-34501-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/27/2022] [Indexed: 11/19/2022] Open
Abstract
Resistance to African trypanosomes in humans relies in part on the high affinity targeting of a trypanosome lytic factor 1 (TLF1) to a trypanosome haptoglobin-hemoglobin receptor (HpHbR). While TLF1 avoidance by the inactivation of HpHbR contributes to Trypanosoma brucei gambiense human infectivity, the evolutionary trade-off of this adaptation is unknown, as the physiological function of the receptor remains to be elucidated. Here we show that uptake of hemoglobin via HpHbR constitutes the sole heme import pathway in the trypanosome bloodstream stage. T. b. gambiense strains carrying the inactivating mutation in HpHbR, as well as genetically engineered T. b. brucei HpHbR knock-out lines show only trace levels of intracellular heme and lack hemoprotein-based enzymatic activities, thereby providing an uncommon example of aerobic parasitic proliferation in the absence of heme. We further show that HpHbR facilitates the developmental progression from proliferating long slender forms to cell cycle-arrested stumpy forms in T. b. brucei. Accordingly, T. b. gambiense was found to be poorly competent for slender-to-stumpy differentiation unless a functional HpHbR receptor derived from T. b. brucei was genetically restored. Altogether, we identify heme-deficient metabolism and disrupted cellular differentiation as two distinct HpHbR-dependent evolutionary trade-offs for T. b. gambiense human infectivity.
Collapse
|
10
|
Ali V, Behera S, Nawaz A, Equbal A, Pandey K. Unique thiol metabolism in trypanosomatids: Redox homeostasis and drug resistance. ADVANCES IN PARASITOLOGY 2022; 117:75-155. [PMID: 35878950 DOI: 10.1016/bs.apar.2022.04.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Trypanosomatids are mainly responsible for heterogeneous parasitic diseases: Leishmaniasis, Sleeping sickness, and Chagas disease and control of these diseases implicates serious challenges due to the emergence of drug resistance. Redox-active biomolecules are the endogenous substances in organisms, which play important role in the regulation of redox homeostasis. The redox-active substances like glutathione, trypanothione, cysteine, cysteine persulfides, etc., and other inorganic intermediates (hydrogen peroxide, nitric oxide) are very useful as defence mechanism. In the present review, the suitability of trypanothione and other essential thiol molecules of trypanosomatids as drug targets are described in Leishmania and Trypanosoma. We have explored the role of tryparedoxin, tryparedoxin peroxidase, ascorbate peroxidase, superoxide dismutase, and glutaredoxins in the anti-oxidant mechanism and drug resistance. Up-regulation of some proteins in trypanothione metabolism helps the parasites in survival against drug pressure (sodium stibogluconate, Amphotericin B, etc.) and oxidative stress. These molecules accept electrons from the reduced trypanothione and donate their electrons to other proteins, and these proteins reduce toxic molecules, neutralize reactive oxygen, or nitrogen species; and help parasites to cope with oxidative stress. Thus, a better understanding of the role of these molecules in drug resistance and redox homeostasis will help to target metabolic pathway proteins to combat Leishmaniasis and trypanosomiases.
Collapse
Affiliation(s)
- Vahab Ali
- Laboratory of Molecular Biochemistry and Cell Biology, Department of Biochemistry, ICMR-Rajendra Memorial Research Institute of Medical Sciences (RMRIMS), Patna, Bihar, India.
| | - Sachidananda Behera
- Laboratory of Molecular Biochemistry and Cell Biology, Department of Biochemistry, ICMR-Rajendra Memorial Research Institute of Medical Sciences (RMRIMS), Patna, Bihar, India
| | - Afreen Nawaz
- Laboratory of Molecular Biochemistry and Cell Biology, Department of Biochemistry, ICMR-Rajendra Memorial Research Institute of Medical Sciences (RMRIMS), Patna, Bihar, India
| | - Asif Equbal
- Laboratory of Molecular Biochemistry and Cell Biology, Department of Biochemistry, ICMR-Rajendra Memorial Research Institute of Medical Sciences (RMRIMS), Patna, Bihar, India; Department of Botany, Araria College, Purnea University, Purnia, Bihar, India
| | - Krishna Pandey
- Department of Clinical Medicine, ICMR-Rajendra Memorial Research Institute of Medical Sciences (RMRIMS), Patna, Bihar, India
| |
Collapse
|
11
|
Patnaik BB, Lee YS, Han YS, Jo YH. Editorial: Innate Immune System Guiding Physiological Plasticity in Invertebrates. Front Physiol 2022; 13:947707. [PMID: 35774282 PMCID: PMC9237642 DOI: 10.3389/fphys.2022.947707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 11/15/2022] Open
Affiliation(s)
- Bharat Bhusan Patnaik
- Department of Biosciences and Biotechnology, Fakir Mohan University, Balasore, India
- Department of Biology, College of Natural Sciences, Soonchunhyang University, Asan, South Korea
- *Correspondence: Bharat Bhusan Patnaik,
| | - Yong Seok Lee
- Department of Biology, College of Natural Sciences, Soonchunhyang University, Asan, South Korea
| | - Yeon Soo Han
- Department of Applied Biology, Institute of Environmentally-Friendly Agriculture, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, South Korea
| | - Yong Hun Jo
- Department of Applied Biology, Institute of Environmentally-Friendly Agriculture, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, South Korea
| |
Collapse
|
12
|
Tihon E, Rubio-Peña K, Dujeancourt-Henry A, Crouzols A, Rotureau B, Glover L. VEX1 Influences mVSG Expression During the Transition to Mammalian Infectivity in Trypanosoma brucei. Front Cell Dev Biol 2022; 10:851475. [PMID: 35450294 PMCID: PMC9017762 DOI: 10.3389/fcell.2022.851475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 02/28/2022] [Indexed: 11/28/2022] Open
Abstract
The Trypanosoma (T) brucei life cycle alternates between the tsetse fly vector and the mammalian host. In the insect, T. brucei undergoes several developmental stages until it reaches the salivary gland and differentiates into the metacyclic form, which is capable of infecting the next mammalian host. Mammalian infectivity is dependent on expression of the metacyclic variant surface glycoprotein genes as the cells develop into mature metacyclics. The VEX complex is essential for monoallelic variant surface glycoprotein expression in T. brucei bloodstream form, however, initiation of expression of the surface proteins genes during metacyclic differentiation is poorly understood. To better understand the transition to mature metacyclics and the control of metacyclic variant surface glycoprotein expression we examined the role of VEX1 in this process. We show that modulating VEX1 expression leads to a dysregulation of variant surface glycoprotein expression during metacyclogenesis, and that following both in vivo and in vitro metacyclic differentiation VEX1 relocalises from multiple nuclear foci in procyclic cells to one to two distinct nuclear foci in metacyclic cells - a pattern like the one seen in mammalian infective bloodstream forms. Our data suggest a role for VEX1 in the metacyclic differentiation process and their capacity to become infectious to the mammalian host.
Collapse
Affiliation(s)
- Eliane Tihon
- Trypanosome Molecular Biology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Karinna Rubio-Peña
- Trypanosome Molecular Biology, Institut Pasteur, Université Paris Cité, Paris, France
| | | | - Aline Crouzols
- Trypanosome Transmission Group, Trypanosome Cell Biology Unit, INSERM U1201 and, Institut Pasteur, Paris, France
| | - Brice Rotureau
- Trypanosome Transmission Group, Trypanosome Cell Biology Unit, INSERM U1201 and, Institut Pasteur, Paris, France
- Parasitology Lab, Institut Pasteur of Guinea, Conakry, Guinea
| | - Lucy Glover
- Trypanosome Molecular Biology, Institut Pasteur, Université Paris Cité, Paris, France
- *Correspondence: Lucy Glover,
| |
Collapse
|
13
|
De Vooght L, De Ridder K, Hussain S, Stijlemans B, De Baetselier P, Caljon G, Van Den Abbeele J. Targeting the tsetse-trypanosome interplay using genetically engineered Sodalis glossinidius. PLoS Pathog 2022; 18:e1010376. [PMID: 35271685 PMCID: PMC8939806 DOI: 10.1371/journal.ppat.1010376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 03/22/2022] [Accepted: 02/15/2022] [Indexed: 11/18/2022] Open
Abstract
Sodalis glossinidius, a secondary bacterial symbiont of the tsetse fly, is currently considered as a potential delivery system for anti-trypanosomal components interfering with African trypanosome transmission (i.e. paratransgenesis). Nanobodies (Nbs) have been proposed as potential candidates to target the parasite during development in the tsetse fly. In this study, we have generated an immune Nb-library and developed a panning strategy to select Nbs against the Trypanosoma brucei brucei procyclic developmental stage present in the tsetse fly midgut. Selected Nbs were expressed, purified, assessed for binding and tested for their impact on the survival and growth of in vitro cultured procyclic T. b. brucei parasites. Next, we engineered S. glossinidius to express the selected Nbs and validated their ability to block T. brucei development in the tsetse fly midgut. Genetically engineered S. glossinidius expressing Nb_88 significantly compromised parasite development in the tsetse fly midgut both at the level of infection rate and parasite load. Interestingly, expression of Nb_19 by S. glossinidius resulted in a significantly enhanced midgut establishment. These data are the first to show in situ delivery by S. glossinidius of effector molecules that can target the trypanosome-tsetse fly crosstalk, interfering with parasite development in the fly. These proof-of-principle data represent a major step forward in the development of a control strategy based on paratransgenic tsetse flies. Finally, S. glossinidius-based Nb delivery can also be applied as a powerful laboratory tool to unravel the molecular determinants of the parasite-vector association.
Collapse
Affiliation(s)
- Linda De Vooght
- Department of Biomedical Sciences, Trypanosoma Unit, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Wilrijk, Belgium
- * E-mail: (LDV); (JVDA)
| | - Karin De Ridder
- Department of Biomedical Sciences, Trypanosoma Unit, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | - Shahid Hussain
- Unit of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Benoît Stijlemans
- Unit of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Inflammation Research Center, Gent, Belgium
| | - Patrick De Baetselier
- Unit of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Wilrijk, Belgium
| | - Jan Van Den Abbeele
- Department of Biomedical Sciences, Trypanosoma Unit, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
- * E-mail: (LDV); (JVDA)
| |
Collapse
|
14
|
Kwon H, Smith R. Anopheles gambiae Actively Metabolizes Uric Acid Following Plasmodium Infection to Limit Malaria Parasite Survival. Front Physiol 2022; 12:821869. [PMID: 35140633 PMCID: PMC8818946 DOI: 10.3389/fphys.2021.821869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 12/31/2021] [Indexed: 02/04/2023] Open
Abstract
Characterizing the physiological changes that accompany malaria parasite infection of the mosquito host is crucial to our understanding of vectorial capacity in Anopheles mosquitoes, yet has not fully been explored. In this study, we examine the role of uric acid metabolism in the mosquito, Anopheles gambiae, following malaria parasite infection. We demonstrate that levels of uric acid are significantly decreased in the excreta and the mosquito at 24 and 48 h post-Plasmodium infection when compared to controls fed on naïve mouse blood. When we examine the expression of well-known enzymes responsible for uric acid metabolism, we see a significant increase in both urate oxidase (UO) and allatoicase (ALLC) expression following Plasmodium infection. Targeting the essential first step in uric acid metabolism by silencing UO resulted in elevated levels of uric acid, enhancing malaria parasite survival. With implications from other insect systems that bacteria can modulate UO expression, we examined the possibility that the mosquito microbiota and its expansion following blood-feeding may contribute to increased UO levels. However, there was no difference in uric acid metabolism between septic and aseptic mosquitoes, indicating that the mosquito microbiome is not associated with the manipulation of UO expression. Together, our study provides new evidence that Plasmodium infection causes the mosquito host to actively metabolize uric acid by increasing UO expression to limit Plasmodium oocyst survival, suggesting that nitrogen metabolism is an essential pathway in defining mosquito vector competence.
Collapse
|
15
|
Brown LD, Maness R, Hall C, Gibson JD. Reactive oxygen species-mediated immunity against bacterial infection in the gut of cat fleas (Ctenocephalides felis). INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2021; 136:103620. [PMID: 34216781 DOI: 10.1016/j.ibmb.2021.103620] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/21/2021] [Accepted: 06/29/2021] [Indexed: 06/13/2023]
Abstract
Fleas (Order Siphonaptera) transmit numerous bacterial pathogens that cause severe human diseases (e.g., cat scratch disease, flea-borne spotted fever, murine typhus, plague). Because initial entry of these infectious agents occurs while blood feeding, the immune response in the flea gut is considered to be the first line of defense against invading microbes. However, relatively few studies have identified the flea immune molecules that effectively resist or limit infection in the gut. In other hematophagous insects, an immediate immune response to imbibed pathogens is the generation of reactive oxygen species (ROS). In this study, we utilized cat fleas (Ctenocephalides felis) to investigate whether oral infection with a well-known insect bacterial pathogen (Serratia marcescens) induces ROS synthesis in the flea gut, and whether production of ROS provides a defense mechanism against microbial colonization. Specifically, we treated fleas with an antioxidant to limit the number of free radicals in the digestive tract prior to infection, and then measured the following: S. marcescens infection loads, hydrogen peroxide (ROS) levels, and mRNA abundance of ROS signaling pathway genes. Overall, our data shows that ROS levels increase in response to infection in the flea gut, and that this increase helps to strengthen the flea immune response through the microbicidal activity of ROS.
Collapse
Affiliation(s)
- Lisa D Brown
- Department of Biology, Georgia Southern University, 4324 Old Register Rd., Statesboro, GA, 30460, USA.
| | - Ryne Maness
- Department of Biology, Georgia Southern University, 4324 Old Register Rd., Statesboro, GA, 30460, USA
| | - Clark Hall
- Department of Biology, Georgia Southern University, 4324 Old Register Rd., Statesboro, GA, 30460, USA
| | - Joshua D Gibson
- Department of Biology, Georgia Southern University, 4324 Old Register Rd., Statesboro, GA, 30460, USA
| |
Collapse
|
16
|
Schuster S, Lisack J, Subota I, Zimmermann H, Reuter C, Mueller T, Morriswood B, Engstler M. Unexpected plasticity in the life cycle of Trypanosoma brucei. eLife 2021; 10:66028. [PMID: 34355698 PMCID: PMC8448533 DOI: 10.7554/elife.66028] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 08/05/2021] [Indexed: 12/22/2022] Open
Abstract
African trypanosomes cause sleeping sickness in humans and nagana in cattle. These unicellular parasites are transmitted by the bloodsucking tsetse fly. In the mammalian host’s circulation, proliferating slender stage cells differentiate into cell cycle-arrested stumpy stage cells when they reach high population densities. This stage transition is thought to fulfil two main functions: first, it auto-regulates the parasite load in the host; second, the stumpy stage is regarded as the only stage capable of successful vector transmission. Here, we show that proliferating slender stage trypanosomes express the mRNA and protein of a known stumpy stage marker, complete the complex life cycle in the fly as successfully as the stumpy stage, and require only a single parasite for productive infection. These findings suggest a reassessment of the traditional view of the trypanosome life cycle. They may also provide a solution to a long-lasting paradox, namely the successful transmission of parasites in chronic infections, despite low parasitemia.
Collapse
Affiliation(s)
- Sarah Schuster
- Department of Cell and Developmental Biology, University of Würzburg, Würzburg, Germany
| | - Jaime Lisack
- Department of Cell and Developmental Biology, University of Würzburg, Würzburg, Germany
| | - Ines Subota
- Department of Cell and Developmental Biology, University of Würzburg, Würzburg, Germany
| | - Henriette Zimmermann
- Department of Cell and Developmental Biology, University of Würzburg, Würzburg, Germany
| | - Christian Reuter
- Department of Cell and Developmental Biology, University of Würzburg, Würzburg, Germany
| | | | | | - Markus Engstler
- Department of Cell and Developmental Biology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
17
|
Dofuor AK, Ademolue TS, Amisigo CM, Kyeremeh K, Gwira TM. Chemical Derivatization and Characterization of Novel Antitrypanosomals for African Trypanosomiasis. Molecules 2021; 26:molecules26154488. [PMID: 34361641 PMCID: PMC8347361 DOI: 10.3390/molecules26154488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 11/16/2022] Open
Abstract
The search for novel antitrypanosomals and the investigation into their mode of action remain crucial due to the toxicity and resistance of commercially available antitrypanosomal drugs. In this study, two novel antitrypanosomals, tortodofuordioxamide (compound 2) and tortodofuorpyramide (compound 3), were chemically derived from the natural N-alkylamide tortozanthoxylamide (compound 1) through structural modification. The chemical structures of these compounds were confirmed through spectrometric and spectroscopic analysis, and their in vitro efficacy and possible mechanisms of action were, subsequently, investigated in Trypanosoma brucei (T. brucei), one of the causative species of African trypanosomiasis (AT). The novel compounds 2 and 3 displayed significant antitrypanosomal potencies in terms of half-maximal effective concentrations (EC50) and selectivity indices (SI) (compound 1, EC50 = 7.3 μM, SI = 29.5; compound 2, EC50 = 3.2 μM, SI = 91.3; compound 3, EC50 = 4.5 μM, SI = 69.9). Microscopic analysis indicated that at the EC50 values, the compounds resulted in the coiling and clumping of parasite subpopulations without significantly affecting the normal ratio of nuclei to kinetoplasts. In contrast to the animal antitrypanosomal drug diminazene, compounds 1, 2 and 3 exhibited antioxidant absorbance properties comparable to the standard antioxidant Trolox (Trolox, 0.11 A; diminazene, 0.50 A; compound 1, 0.10 A; compound 2, 0.09 A; compound 3, 0.11 A). The analysis of growth kinetics suggested that the compounds exhibited a relatively gradual but consistent growth inhibition of T. brucei at different concentrations. The results suggest that further pharmacological optimization of compounds 2 and 3 may facilitate their development into novel AT chemotherapy.
Collapse
Affiliation(s)
- Aboagye Kwarteng Dofuor
- West African Center for Cell Biology of Infectious Pathogens, University of Ghana, Legon, Accra P.O. Box LG 54, Ghana; (A.K.D.); (T.S.A.); (C.M.A.)
- Department of Biological, Physical and Mathematical Sciences, University of Environment and Sustainable Development, PMB, Somanya, Ghana
| | - Temitayo Samson Ademolue
- West African Center for Cell Biology of Infectious Pathogens, University of Ghana, Legon, Accra P.O. Box LG 54, Ghana; (A.K.D.); (T.S.A.); (C.M.A.)
- Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra P.O. Box LG 54, Ghana
| | - Cynthia Mmalebna Amisigo
- West African Center for Cell Biology of Infectious Pathogens, University of Ghana, Legon, Accra P.O. Box LG 54, Ghana; (A.K.D.); (T.S.A.); (C.M.A.)
- Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra P.O. Box LG 54, Ghana
| | - Kwaku Kyeremeh
- Department of Chemistry, University of Ghana, Legon, Accra P.O. Box LG 56, Ghana;
| | - Theresa Manful Gwira
- West African Center for Cell Biology of Infectious Pathogens, University of Ghana, Legon, Accra P.O. Box LG 54, Ghana; (A.K.D.); (T.S.A.); (C.M.A.)
- Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Accra P.O. Box LG 54, Ghana
- Correspondence:
| |
Collapse
|
18
|
Yang L, Weiss BL, Williams AE, Aksoy E, de Silva Orfano A, Son JH, Wu Y, Vigneron A, Karakus M, Aksoy S. Paratransgenic manipulation of a tsetse microRNA alters the physiological homeostasis of the fly's midgut environment. PLoS Pathog 2021; 17:e1009475. [PMID: 34107000 PMCID: PMC8216540 DOI: 10.1371/journal.ppat.1009475] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/21/2021] [Accepted: 05/13/2021] [Indexed: 12/27/2022] Open
Abstract
Tsetse flies are vectors of parasitic African trypanosomes, the etiological agents of human and animal African trypanosomoses. Current disease control methods include fly-repelling pesticides, fly trapping, and chemotherapeutic treatment of infected people and animals. Inhibiting tsetse's ability to transmit trypanosomes by strengthening the fly's natural barriers can serve as an alternative approach to reduce disease. The peritrophic matrix (PM) is a chitinous and proteinaceous barrier that lines the insect midgut and serves as a protective barrier that inhibits infection with pathogens. African trypanosomes must cross tsetse's PM in order to establish an infection in the fly, and PM structural integrity negatively correlates with trypanosome infection outcomes. Bloodstream form trypanosomes shed variant surface glycoproteins (VSG) into tsetse's gut lumen early during the infection establishment, and free VSG molecules are internalized by the fly's PM-producing cardia. This process results in a reduction in the expression of a tsetse microRNA (miR275) and a sequential molecular cascade that compromises PM integrity. miRNAs are small non-coding RNAs that are critical in regulating many physiological processes. In the present study, we investigated the role(s) of tsetse miR275 by developing a paratransgenic expression system that employs tsetse's facultative bacterial endosymbiont, Sodalis glossinidius, to express tandem antagomir-275 repeats (or miR275 sponges). This system induces a constitutive, 40% reduction in miR275 transcript abundance in the fly's midgut and results in obstructed blood digestion (gut weights increased by 52%), a significant increase (p-value < 0.0001) in fly survival following infection with an entomopathogenic bacteria, and a 78% increase in trypanosome infection prevalence. RNA sequencing of cardia and midgut tissues from paratransgenic tsetse confirmed that miR275 regulates processes related to the expression of PM-associated proteins and digestive enzymes as well as genes that encode abundant secretory proteins. Our study demonstrates that paratransgenesis can be employed to study microRNA regulated pathways in arthropods that house symbiotic bacteria.
Collapse
Affiliation(s)
- Liu Yang
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Brian L. Weiss
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Adeline E. Williams
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
- Department of Microbiology, Immunology, Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Emre Aksoy
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Alessandra de Silva Orfano
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Jae Hak Son
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Yineng Wu
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Aurelien Vigneron
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
- Department of Evolutionary Ecology, Institute for Organismic and Molecular Evolution, Johannes Gutenberg University, Mainz, Germany
| | - Mehmet Karakus
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
- Department of Medical Microbiology, Faculty of Medicine, University of Health Sciences, Istanbul, Turkey
| | - Serap Aksoy
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America
| |
Collapse
|
19
|
Medina Munoz M, Brenner C, Richmond D, Spencer N, Rio RVM. The holobiont transcriptome of teneral tsetse fly species of varying vector competence. BMC Genomics 2021; 22:400. [PMID: 34058984 PMCID: PMC8166097 DOI: 10.1186/s12864-021-07729-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 05/21/2021] [Indexed: 12/13/2022] Open
Abstract
Background Tsetse flies are the obligate vectors of African trypanosomes, which cause Human and Animal African Trypanosomiasis. Teneral flies (newly eclosed adults) are especially susceptible to parasite establishment and development, yet our understanding of why remains fragmentary. The tsetse gut microbiome is dominated by two Gammaproteobacteria, an essential and ancient mutualist Wigglesworthia glossinidia and a commensal Sodalis glossinidius. Here, we characterize and compare the metatranscriptome of teneral Glossina morsitans to that of G. brevipalpis and describe unique immunological, physiological, and metabolic landscapes that may impact vector competence differences between these two species. Results An active expression profile was observed for Wigglesworthia immediately following host adult metamorphosis. Specifically, ‘translation, ribosomal structure and biogenesis’ followed by ‘coenzyme transport and metabolism’ were the most enriched clusters of orthologous genes (COGs), highlighting the importance of nutrient transport and metabolism even following host species diversification. Despite the significantly smaller Wigglesworthia genome more differentially expressed genes (DEGs) were identified between interspecific isolates (n = 326, ~ 55% of protein coding genes) than between the corresponding Sodalis isolates (n = 235, ~ 5% of protein coding genes) likely reflecting distinctions in host co-evolution and adaptation. DEGs between Sodalis isolates included genes involved in chitin degradation that may contribute towards trypanosome susceptibility by compromising the immunological protection provided by the peritrophic matrix. Lastly, G. brevipalpis tenerals demonstrate a more immunologically robust background with significant upregulation of IMD and melanization pathways. Conclusions These transcriptomic differences may collectively contribute to vector competence differences between tsetse species and offers translational relevance towards the design of novel vector control strategies. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-07729-5.
Collapse
Affiliation(s)
- Miguel Medina Munoz
- Department of Biology, Eberly College of Arts and Sciences, West Virginia University, Morgantown, WV, 26505, USA
| | - Caitlyn Brenner
- Department of Biology, Washington and Jefferson College, Washington, PA, 15301, USA
| | - Dylan Richmond
- Department of Biology, Eberly College of Arts and Sciences, West Virginia University, Morgantown, WV, 26505, USA
| | - Noah Spencer
- Department of Biology, Eberly College of Arts and Sciences, West Virginia University, Morgantown, WV, 26505, USA
| | - Rita V M Rio
- Department of Biology, Eberly College of Arts and Sciences, West Virginia University, Morgantown, WV, 26505, USA.
| |
Collapse
|
20
|
Calvo-Álvarez E, Bonnefoy S, Salles A, Benson FE, McKean PG, Bastin P, Rotureau B. Redistribution of FLAgellar Member 8 during the trypanosome life cycle: Consequences for cell fate prediction. Cell Microbiol 2021; 23:e13347. [PMID: 33896083 PMCID: PMC8459223 DOI: 10.1111/cmi.13347] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 04/17/2021] [Accepted: 04/22/2021] [Indexed: 11/28/2022]
Abstract
The single flagellum of African trypanosomes is essential in multiple aspects of the parasites' development. The FLAgellar Member 8 protein (FLAM8), localised to the tip of the flagellum in cultured insect forms of Trypanosoma brucei, was identified as a marker of the locking event that controls flagellum length. Here, we investigated whether FLAM8 could also reflect the flagellum maturation state in other parasite cycle stages. We observed that FLAM8 distribution extended along the entire flagellar cytoskeleton in mammalian‐infective forms. Then, a rapid FLAM8 concentration to the distal tip occurs during differentiation into early insect forms, illustrating the remodelling of an existing flagellum. In the tsetse cardia, FLAM8 further localises to the entire length of the new flagellum during an asymmetric division. Strikingly, in parasites dividing in the tsetse midgut and in the salivary glands, the amount and distribution of FLAM8 in the new flagellum were seen to predict the daughter cell fate. We propose and discuss how FLAM8 could be considered a meta‐marker of the flagellum stage and maturation state in trypanosomes.
Collapse
Affiliation(s)
- Estefanía Calvo-Álvarez
- Trypanosome Cell Biology Unit, Institut Pasteur and INSERM U1201, Paris, France.,Trypanosome Transmission Group, Institut Pasteur, Paris, France
| | - Serge Bonnefoy
- Trypanosome Cell Biology Unit, Institut Pasteur and INSERM U1201, Paris, France
| | - Audrey Salles
- Unit of Technology and Service Photonic BioImaging (UTechS PBI), C2RT, Institut Pasteur, Paris, France
| | - Fiona E Benson
- Division of Biomedical and Life Sciences, Lancaster University, Lancaster, UK
| | - Paul G McKean
- Division of Biomedical and Life Sciences, Lancaster University, Lancaster, UK
| | - Philippe Bastin
- Trypanosome Cell Biology Unit, Institut Pasteur and INSERM U1201, Paris, France
| | - Brice Rotureau
- Trypanosome Cell Biology Unit, Institut Pasteur and INSERM U1201, Paris, France.,Trypanosome Transmission Group, Institut Pasteur, Paris, France
| |
Collapse
|
21
|
Bertiaux E, Mallet A, Rotureau B, Bastin P. Intraflagellar transport during assembly of flagella of different length in Trypanosoma brucei isolated from tsetse flies. J Cell Sci 2020; 133:jcs248989. [PMID: 32843573 DOI: 10.1242/jcs.248989] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 08/10/2020] [Indexed: 11/20/2022] Open
Abstract
Multicellular organisms assemble cilia and flagella of precise lengths differing from one cell to another, yet little is known about the mechanisms governing these differences. Similarly, protists assemble flagella of different lengths according to the stage of their life cycle. Trypanosoma brucei assembles flagella of 3 to 30 µm during its development in the tsetse fly. This provides an opportunity to examine how cells naturally modulate organelle length. Flagella are constructed by addition of new blocks at their distal end via intraflagellar transport (IFT). Immunofluorescence assays, 3D electron microscopy and live-cell imaging revealed that IFT was present in all T. brucei life cycle stages. IFT proteins are concentrated at the base, and IFT trains are located along doublets 3-4 and 7-8 and travel bidirectionally in the flagellum. Quantitative analysis demonstrated that the total amount of flagellar IFT proteins correlates with the length of the flagellum. Surprisingly, the shortest flagellum exhibited a supplementary large amount of dynamic IFT material at its distal end. The contribution of IFT and other factors to the regulation of flagellum length is discussed.
Collapse
Affiliation(s)
- Eloïse Bertiaux
- Trypanosome Cell Biology Unit, INSERM U1201, Institut Pasteur, 25 Rue du Docteur Roux, 75015 Paris, France
- Sorbonne Université école doctorale complexité du vivant, ED 515, 7, quai Saint-Bernard, case 32, 75252 Paris Cedex 05, France
| | - Adeline Mallet
- Trypanosome Cell Biology Unit, INSERM U1201, Institut Pasteur, 25 Rue du Docteur Roux, 75015 Paris, France
- Sorbonne Université école doctorale complexité du vivant, ED 515, 7, quai Saint-Bernard, case 32, 75252 Paris Cedex 05, France
- Ultrastructural Bio Imaging Unit, C2RT, Institut Pasteur, 25 Rue du Docteur Roux, 75015 Paris, France
| | - Brice Rotureau
- Trypanosome Cell Biology Unit, INSERM U1201, Institut Pasteur, 25 Rue du Docteur Roux, 75015 Paris, France
| | - Philippe Bastin
- Trypanosome Cell Biology Unit, INSERM U1201, Institut Pasteur, 25 Rue du Docteur Roux, 75015 Paris, France
| |
Collapse
|
22
|
Peacock L, Gibson W. Tsetse Fly Transmission Studies of African Trypanosomes. Methods Mol Biol 2020; 2116:49-67. [PMID: 32221913 DOI: 10.1007/978-1-0716-0294-2_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2024]
Abstract
African trypanosomes are naturally transmitted by bloodsucking tsetse flies in sub-Saharan Africa and these transmission cycles can be reproduced in the laboratory if clean tsetse flies and suitable trypanosomes are available for experiments. Tsetse transmission gives access to more trypanosome developmental stages than are available from in vitro culture, albeit in very small numbers; for example, the sexual stages of Trypanosoma brucei have been isolated from infected tsetse salivary glands, but have not yet been reported from culture. Tsetse transmission also allows for the natural transition between different developmental stages to be studied.Both wild-type and genetically modified trypanosomes have been successfully fly transmitted, and it is possible to manipulate the trypanosome environment inside the fly to some extent, for example, the induction of expression of genes controlled by the Tet repressor by feeding flies with tetracycline.
Collapse
Affiliation(s)
- Lori Peacock
- Bristol Veterinary School and School of Biological Sciences, University of Bristol, Bristol, UK
| | - Wendy Gibson
- Bristol Veterinary School and School of Biological Sciences, University of Bristol, Bristol, UK.
| |
Collapse
|
23
|
Horáková E, Faktorová D, Kraeva N, Kaur B, Van Den Abbeele J, Yurchenko V, Lukeš J. Catalase compromises the development of the insect and mammalian stages of Trypanosoma brucei. FEBS J 2019; 287:964-977. [PMID: 31593329 DOI: 10.1111/febs.15083] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 06/21/2019] [Accepted: 10/04/2019] [Indexed: 12/14/2022]
Abstract
Catalase is a widespread heme-containing enzyme, which converts hydrogen peroxide (H2 O2 ) to water and molecular oxygen, thereby protecting cells from the toxic effects of H2 O2 . Trypanosoma brucei is an aerobic protist, which conspicuously lacks this potent enzyme, present in virtually all organisms exposed to oxidative stress. To uncover the reasons for its absence in T. brucei, we overexpressed different catalases in procyclic and bloodstream stages of the parasite. The heterologous enzymes originated from the related insect-confined trypanosomatid Crithidia fasciculata and the human. While the trypanosomatid enzyme (cCAT) operates at low temperatures, its human homolog (hCAT) is adapted to the warm-blooded environment. Despite the presence of peroxisomal targeting signal in hCAT, both human and C. fasciculata catalases localized to the cytosol of T. brucei. Even though cCAT was efficiently expressed in both life cycle stages, the enzyme was active in the procyclic stage, increasing cell's resistance to the H2 O2 stress, yet its activity was suppressed in the cultured bloodstream stage. Surprisingly, following the expression of hCAT, the ability to establish the T. brucei infection in the tsetse fly midgut was compromised. In the mouse model, hCAT attenuated parasitemia and, consequently, increased the host's survival. Hence, we suggest that the activity of catalase in T. brucei is beneficial in vitro, yet it becomes detrimental for parasite's proliferation in both invertebrate and vertebrate hosts, leading to an inability to carry this, otherwise omnipresent, enzyme.
Collapse
Affiliation(s)
- Eva Horáková
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, České Budějovice (Budweis), Czech Republic
| | - Drahomíra Faktorová
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, České Budějovice (Budweis), Czech Republic.,Faculty of Science, University of South Bohemia, České Budějovice (Budweis), Czech Republic
| | - Natalia Kraeva
- Life Science Research Centre, Faculty of Science, University of Ostrava, Czech Republic
| | - Binnypreet Kaur
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, České Budějovice (Budweis), Czech Republic.,Faculty of Science, University of South Bohemia, České Budějovice (Budweis), Czech Republic
| | - Jan Van Den Abbeele
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Vyacheslav Yurchenko
- Life Science Research Centre, Faculty of Science, University of Ostrava, Czech Republic.,Martsinovsky Institute of Medical Parasitology, Tropical and Vector Borne Diseases, Sechenov University, Moscow, Russia
| | - Julius Lukeš
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, České Budějovice (Budweis), Czech Republic.,Faculty of Science, University of South Bohemia, České Budějovice (Budweis), Czech Republic
| |
Collapse
|
24
|
Aksoy S. Tsetse peritrophic matrix influences for trypanosome transmission. JOURNAL OF INSECT PHYSIOLOGY 2019; 118:103919. [PMID: 31425686 PMCID: PMC6853167 DOI: 10.1016/j.jinsphys.2019.103919] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 08/09/2019] [Accepted: 08/09/2019] [Indexed: 06/10/2023]
Abstract
Tsetse flies are important vectors of parasitic African trypanosomes, agents of human and animal trypanosomiasis. Easily administrable and effective tools for disease control in the mammalian host are still lacking but reduction of the tsetse vector populations can reduce disease. An alternative approach is to reduce the transmission of trypanosomes in the tsetse vector. The gut peritrophic matrix (PM) has emerged as an important regulator of parasite transmission success in tsetse. Tsetse has a Type II PM that is constitutively produced by cells in the cardia organ. Tsetse PM lines the entire gut and functions as an immunological barrier to prevent the gut epithelia from responding to commensal environmental microbes present in the gut lumen. Tsetse PM also functions as a physical barrier to trypanosome infections that enter into the gut lumen in an infective blood meal. For persistence in the gut, African trypanosomes have developed an adaptive manipulative process to transiently reduce PM efficacy. The process is mediated by mammalian trypanosome surface coat proteins, Variant Surface Glycoproteins (VSGs) which are shed in the gut lumen and taken up by cardia cells. The mechanism of PM reduction involves a tsetse microRNA (miR-275) which acts thru the Wnt signaling pathway. The PM efficacy is once again reduced later in the infection process to enable the gut established parasites to reenter into the gut lumen to colonize the salivary glands, an essential process for transmission. The ability to modulate PM integrity can lead to innovative approaches to reduce disease transmission.
Collapse
Affiliation(s)
- Serap Aksoy
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, 60 College St, LEPH 624, New Haven, CT 06520, United States.
| |
Collapse
|
25
|
Attardo GM, Abd-Alla AMM, Acosta-Serrano A, Allen JE, Bateta R, Benoit JB, Bourtzis K, Caers J, Caljon G, Christensen MB, Farrow DW, Friedrich M, Hua-Van A, Jennings EC, Larkin DM, Lawson D, Lehane MJ, Lenis VP, Lowy-Gallego E, Macharia RW, Malacrida AR, Marco HG, Masiga D, Maslen GL, Matetovici I, Meisel RP, Meki I, Michalkova V, Miller WJ, Minx P, Mireji PO, Ometto L, Parker AG, Rio R, Rose C, Rosendale AJ, Rota-Stabelli O, Savini G, Schoofs L, Scolari F, Swain MT, Takáč P, Tomlinson C, Tsiamis G, Van Den Abbeele J, Vigneron A, Wang J, Warren WC, Waterhouse RM, Weirauch MT, Weiss BL, Wilson RK, Zhao X, Aksoy S. Comparative genomic analysis of six Glossina genomes, vectors of African trypanosomes. Genome Biol 2019; 20:187. [PMID: 31477173 PMCID: PMC6721284 DOI: 10.1186/s13059-019-1768-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 07/22/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Tsetse flies (Glossina sp.) are the vectors of human and animal trypanosomiasis throughout sub-Saharan Africa. Tsetse flies are distinguished from other Diptera by unique adaptations, including lactation and the birthing of live young (obligate viviparity), a vertebrate blood-specific diet by both sexes, and obligate bacterial symbiosis. This work describes the comparative analysis of six Glossina genomes representing three sub-genera: Morsitans (G. morsitans morsitans, G. pallidipes, G. austeni), Palpalis (G. palpalis, G. fuscipes), and Fusca (G. brevipalpis) which represent different habitats, host preferences, and vectorial capacity. RESULTS Genomic analyses validate established evolutionary relationships and sub-genera. Syntenic analysis of Glossina relative to Drosophila melanogaster shows reduced structural conservation across the sex-linked X chromosome. Sex-linked scaffolds show increased rates of female-specific gene expression and lower evolutionary rates relative to autosome associated genes. Tsetse-specific genes are enriched in protease, odorant-binding, and helicase activities. Lactation-associated genes are conserved across all Glossina species while male seminal proteins are rapidly evolving. Olfactory and gustatory genes are reduced across the genus relative to other insects. Vision-associated Rhodopsin genes show conservation of motion detection/tracking functions and variance in the Rhodopsin detecting colors in the blue wavelength ranges. CONCLUSIONS Expanded genomic discoveries reveal the genetics underlying Glossina biology and provide a rich body of knowledge for basic science and disease control. They also provide insight into the evolutionary biology underlying novel adaptations and are relevant to applied aspects of vector control such as trap design and discovery of novel pest and disease control strategies.
Collapse
Affiliation(s)
- Geoffrey M Attardo
- Department of Entomology and Nematology, University of California, Davis, Davis, CA, USA.
| | - Adly M M Abd-Alla
- Insect Pest Control Laboratory, Joint FAO/IAEA Division of Nuclear Techniques in Food & Agriculture, Vienna, Vienna, Austria
| | - Alvaro Acosta-Serrano
- Department of Vector Biology, Liverpool School of Tropical Medicine, Merseyside, Liverpool, UK
| | - James E Allen
- VectorBase, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, Cambridgeshire, UK
| | - Rosemary Bateta
- Department of Biochemistry, Biotechnology Research Institute - Kenya Agricultural and Livestock Research Organization, Kikuyu, Kenya
| | - Joshua B Benoit
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH, USA
| | - Kostas Bourtzis
- Insect Pest Control Laboratory, Joint FAO/IAEA Division of Nuclear Techniques in Food & Agriculture, Vienna, Vienna, Austria
| | - Jelle Caers
- Department of Biology - Functional Genomics and Proteomics Group, KU Leuven, Leuven, Belgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene, University of Antwerp, Antwerp, Belgium
| | - Mikkel B Christensen
- VectorBase, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, Cambridgeshire, UK
| | - David W Farrow
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH, USA
| | - Markus Friedrich
- Department of Biological Sciences, Wayne State University, Detroit, MI, USA
| | - Aurélie Hua-Van
- Laboratoire Evolution, Genomes, Comportement, Ecologie, CNRS, IRD, Univ. Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Emily C Jennings
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH, USA
| | - Denis M Larkin
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Daniel Lawson
- Department of Life Sciences, Imperial College London, London, UK
| | - Michael J Lehane
- Department of Vector Biology, Liverpool School of Tropical Medicine, Merseyside, Liverpool, UK
| | - Vasileios P Lenis
- Schools of Medicine and Dentistry, University of Plymouth, Plymouth, UK
| | - Ernesto Lowy-Gallego
- VectorBase, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, Cambridgeshire, UK
| | - Rosaline W Macharia
- Molecular Biology and Bioinformatics Unit, International Center for Insect Physiology and Ecology, Nairobi, Kenya.,Centre for Biotechnology and Bioinformatics, University of Nairobi, Nairobi, Kenya
| | - Anna R Malacrida
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Heather G Marco
- Department of Biological Sciences, University of Cape Town, Rondebosch, South Africa
| | - Daniel Masiga
- Molecular Biology and Bioinformatics Unit, International Center for Insect Physiology and Ecology, Nairobi, Kenya
| | - Gareth L Maslen
- VectorBase, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, Cambridgeshire, UK
| | - Irina Matetovici
- Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Richard P Meisel
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Irene Meki
- Insect Pest Control Laboratory, Joint FAO/IAEA Division of Nuclear Techniques in Food & Agriculture, Vienna, Vienna, Austria
| | - Veronika Michalkova
- Department of Biological Sciences, Florida International University, Miami, Florida, USA.,Institute of Zoology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Wolfgang J Miller
- Department of Cell and Developmental Biology, Medical University of Vienna, Vienna, Austria
| | - Patrick Minx
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Paul O Mireji
- Department of Biochemistry, Biotechnology Research Institute - Kenya Agricultural and Livestock Research Organization, Kikuyu, Kenya.,Centre for Geographic Medicine Research Coast, Kenya Medical Research Institute, Kilifi, Kenya
| | - Lino Ometto
- Department of Sustainable Ecosystems and Bioresources, Research and Innovation Centre, Fondazione Edmund Mach, San Michele all'Adige, TN, Italy.,Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Andrew G Parker
- Insect Pest Control Laboratory, Joint FAO/IAEA Division of Nuclear Techniques in Food & Agriculture, Vienna, Vienna, Austria
| | - Rita Rio
- Department of Biology, West Virginia University, Morgantown, WV, USA
| | - Clair Rose
- Department of Vector Biology, Liverpool School of Tropical Medicine, Merseyside, Liverpool, UK
| | - Andrew J Rosendale
- Department of Biology, Mount St. Joseph University, Cincinnati, OH, USA.,Department of Biological Sciences, University of Cincinnati, Cincinnati, OH, USA
| | - Omar Rota-Stabelli
- Department of Sustainable Ecosystems and Bioresources, Research and Innovation Centre, Fondazione Edmund Mach, San Michele all'Adige, TN, Italy
| | - Grazia Savini
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Liliane Schoofs
- Department of Biology - Functional Genomics and Proteomics Group, KU Leuven, Leuven, Belgium
| | - Francesca Scolari
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Martin T Swain
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, Ceredigion, UK
| | - Peter Takáč
- Department of Animal Systematics, Ústav zoológie SAV; Scientica, Ltd, Bratislava, Slovakia
| | - Chad Tomlinson
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - George Tsiamis
- Department of Environmental and Natural Resources Management, University of Patras, Agrinio, Etoloakarnania, Greece
| | | | - Aurelien Vigneron
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Jingwen Wang
- School of Life Sciences, Fudan University, Shanghai, China
| | - Wesley C Warren
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA.,Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Robert M Waterhouse
- Department of Ecology & Evolution, Swiss Institute of Bioinformatics, University of Lausanne, Lausanne, Switzerland
| | - Matthew T Weirauch
- Center for Autoimmune Genomics and Etiology and Divisions of Biomedical Informatics and Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Brian L Weiss
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Richard K Wilson
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Xin Zhao
- CAS Center for Influenza Research and Early-warning (CASCIRE), Chinese Academy of Sciences, Beijing, China
| | - Serap Aksoy
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA.
| |
Collapse
|
26
|
Attardo GM, Abd-Alla AMM, Acosta-Serrano A, Allen JE, Bateta R, Benoit JB, Bourtzis K, Caers J, Caljon G, Christensen MB, Farrow DW, Friedrich M, Hua-Van A, Jennings EC, Larkin DM, Lawson D, Lehane MJ, Lenis VP, Lowy-Gallego E, Macharia RW, Malacrida AR, Marco HG, Masiga D, Maslen GL, Matetovici I, Meisel RP, Meki I, Michalkova V, Miller WJ, Minx P, Mireji PO, Ometto L, Parker AG, Rio R, Rose C, Rosendale AJ, Rota-Stabelli O, Savini G, Schoofs L, Scolari F, Swain MT, Takáč P, Tomlinson C, Tsiamis G, Van Den Abbeele J, Vigneron A, Wang J, Warren WC, Waterhouse RM, Weirauch MT, Weiss BL, Wilson RK, Zhao X, Aksoy S. Comparative genomic analysis of six Glossina genomes, vectors of African trypanosomes. Genome Biol 2019; 20:187. [PMID: 31477173 DOI: 10.1101/531749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 07/22/2019] [Indexed: 05/24/2023] Open
Abstract
BACKGROUND Tsetse flies (Glossina sp.) are the vectors of human and animal trypanosomiasis throughout sub-Saharan Africa. Tsetse flies are distinguished from other Diptera by unique adaptations, including lactation and the birthing of live young (obligate viviparity), a vertebrate blood-specific diet by both sexes, and obligate bacterial symbiosis. This work describes the comparative analysis of six Glossina genomes representing three sub-genera: Morsitans (G. morsitans morsitans, G. pallidipes, G. austeni), Palpalis (G. palpalis, G. fuscipes), and Fusca (G. brevipalpis) which represent different habitats, host preferences, and vectorial capacity. RESULTS Genomic analyses validate established evolutionary relationships and sub-genera. Syntenic analysis of Glossina relative to Drosophila melanogaster shows reduced structural conservation across the sex-linked X chromosome. Sex-linked scaffolds show increased rates of female-specific gene expression and lower evolutionary rates relative to autosome associated genes. Tsetse-specific genes are enriched in protease, odorant-binding, and helicase activities. Lactation-associated genes are conserved across all Glossina species while male seminal proteins are rapidly evolving. Olfactory and gustatory genes are reduced across the genus relative to other insects. Vision-associated Rhodopsin genes show conservation of motion detection/tracking functions and variance in the Rhodopsin detecting colors in the blue wavelength ranges. CONCLUSIONS Expanded genomic discoveries reveal the genetics underlying Glossina biology and provide a rich body of knowledge for basic science and disease control. They also provide insight into the evolutionary biology underlying novel adaptations and are relevant to applied aspects of vector control such as trap design and discovery of novel pest and disease control strategies.
Collapse
Affiliation(s)
- Geoffrey M Attardo
- Department of Entomology and Nematology, University of California, Davis, Davis, CA, USA.
| | - Adly M M Abd-Alla
- Insect Pest Control Laboratory, Joint FAO/IAEA Division of Nuclear Techniques in Food & Agriculture, Vienna, Vienna, Austria
| | - Alvaro Acosta-Serrano
- Department of Vector Biology, Liverpool School of Tropical Medicine, Merseyside, Liverpool, UK
| | - James E Allen
- VectorBase, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, Cambridgeshire, UK
| | - Rosemary Bateta
- Department of Biochemistry, Biotechnology Research Institute - Kenya Agricultural and Livestock Research Organization, Kikuyu, Kenya
| | - Joshua B Benoit
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH, USA
| | - Kostas Bourtzis
- Insect Pest Control Laboratory, Joint FAO/IAEA Division of Nuclear Techniques in Food & Agriculture, Vienna, Vienna, Austria
| | - Jelle Caers
- Department of Biology - Functional Genomics and Proteomics Group, KU Leuven, Leuven, Belgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene, University of Antwerp, Antwerp, Belgium
| | - Mikkel B Christensen
- VectorBase, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, Cambridgeshire, UK
| | - David W Farrow
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH, USA
| | - Markus Friedrich
- Department of Biological Sciences, Wayne State University, Detroit, MI, USA
| | - Aurélie Hua-Van
- Laboratoire Evolution, Genomes, Comportement, Ecologie, CNRS, IRD, Univ. Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Emily C Jennings
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH, USA
| | - Denis M Larkin
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Daniel Lawson
- Department of Life Sciences, Imperial College London, London, UK
| | - Michael J Lehane
- Department of Vector Biology, Liverpool School of Tropical Medicine, Merseyside, Liverpool, UK
| | - Vasileios P Lenis
- Schools of Medicine and Dentistry, University of Plymouth, Plymouth, UK
| | - Ernesto Lowy-Gallego
- VectorBase, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, Cambridgeshire, UK
| | - Rosaline W Macharia
- Molecular Biology and Bioinformatics Unit, International Center for Insect Physiology and Ecology, Nairobi, Kenya
- Centre for Biotechnology and Bioinformatics, University of Nairobi, Nairobi, Kenya
| | - Anna R Malacrida
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Heather G Marco
- Department of Biological Sciences, University of Cape Town, Rondebosch, South Africa
| | - Daniel Masiga
- Molecular Biology and Bioinformatics Unit, International Center for Insect Physiology and Ecology, Nairobi, Kenya
| | - Gareth L Maslen
- VectorBase, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, Cambridgeshire, UK
| | - Irina Matetovici
- Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Richard P Meisel
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Irene Meki
- Insect Pest Control Laboratory, Joint FAO/IAEA Division of Nuclear Techniques in Food & Agriculture, Vienna, Vienna, Austria
| | - Veronika Michalkova
- Department of Biological Sciences, Florida International University, Miami, Florida, USA
- Institute of Zoology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Wolfgang J Miller
- Department of Cell and Developmental Biology, Medical University of Vienna, Vienna, Austria
| | - Patrick Minx
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Paul O Mireji
- Department of Biochemistry, Biotechnology Research Institute - Kenya Agricultural and Livestock Research Organization, Kikuyu, Kenya
- Centre for Geographic Medicine Research Coast, Kenya Medical Research Institute, Kilifi, Kenya
| | - Lino Ometto
- Department of Sustainable Ecosystems and Bioresources, Research and Innovation Centre, Fondazione Edmund Mach, San Michele all'Adige, TN, Italy
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Andrew G Parker
- Insect Pest Control Laboratory, Joint FAO/IAEA Division of Nuclear Techniques in Food & Agriculture, Vienna, Vienna, Austria
| | - Rita Rio
- Department of Biology, West Virginia University, Morgantown, WV, USA
| | - Clair Rose
- Department of Vector Biology, Liverpool School of Tropical Medicine, Merseyside, Liverpool, UK
| | - Andrew J Rosendale
- Department of Biology, Mount St. Joseph University, Cincinnati, OH, USA
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH, USA
| | - Omar Rota-Stabelli
- Department of Sustainable Ecosystems and Bioresources, Research and Innovation Centre, Fondazione Edmund Mach, San Michele all'Adige, TN, Italy
| | - Grazia Savini
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Liliane Schoofs
- Department of Biology - Functional Genomics and Proteomics Group, KU Leuven, Leuven, Belgium
| | - Francesca Scolari
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Martin T Swain
- Institute of Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, Ceredigion, UK
| | - Peter Takáč
- Department of Animal Systematics, Ústav zoológie SAV; Scientica, Ltd, Bratislava, Slovakia
| | - Chad Tomlinson
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - George Tsiamis
- Department of Environmental and Natural Resources Management, University of Patras, Agrinio, Etoloakarnania, Greece
| | | | - Aurelien Vigneron
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Jingwen Wang
- School of Life Sciences, Fudan University, Shanghai, China
| | - Wesley C Warren
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
- Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Robert M Waterhouse
- Department of Ecology & Evolution, Swiss Institute of Bioinformatics, University of Lausanne, Lausanne, Switzerland
| | - Matthew T Weirauch
- Center for Autoimmune Genomics and Etiology and Divisions of Biomedical Informatics and Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Brian L Weiss
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - Richard K Wilson
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Xin Zhao
- CAS Center for Influenza Research and Early-warning (CASCIRE), Chinese Academy of Sciences, Beijing, China
| | - Serap Aksoy
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA.
| |
Collapse
|
27
|
Matetovici I, De Vooght L, Van Den Abbeele J. Innate immunity in the tsetse fly (Glossina), vector of African trypanosomes. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 98:181-188. [PMID: 31075296 DOI: 10.1016/j.dci.2019.05.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 05/05/2019] [Accepted: 05/06/2019] [Indexed: 06/09/2023]
Abstract
Tsetse flies (Glossina sp.) are medically and veterinary important vectors of African trypanosomes, protozoan parasites that cause devastating diseases in humans and livestock in sub-Saharan Africa. These flies feed exclusively on vertebrate blood and harbor a limited diversity of obligate and facultative bacterial commensals. They have a well-developed innate immune system that plays a key role in protecting the fly against invading pathogens and in modulating the fly's ability to transmit African trypanosomes. In this review, we briefly summarize our current knowledge on the tsetse fly innate immune system and its interaction with the bacterial commensals and the trypanosome parasite.
Collapse
Affiliation(s)
- Irina Matetovici
- Department of Biomedical Sciences, Institute of Tropical Medicine Antwerp, Nationalestraat 155, B-2000, Antwerp, Belgium
| | - Linda De Vooght
- Department of Biomedical Sciences, Institute of Tropical Medicine Antwerp, Nationalestraat 155, B-2000, Antwerp, Belgium
| | - Jan Van Den Abbeele
- Department of Biomedical Sciences, Institute of Tropical Medicine Antwerp, Nationalestraat 155, B-2000, Antwerp, Belgium.
| |
Collapse
|
28
|
Weiss BL, Maltz MA, Vigneron A, Wu Y, Walter KS, O'Neill MB, Wang J, Aksoy S. Colonization of the tsetse fly midgut with commensal Kosakonia cowanii Zambiae inhibits trypanosome infection establishment. PLoS Pathog 2019; 15:e1007470. [PMID: 30817773 PMCID: PMC6394900 DOI: 10.1371/journal.ppat.1007470] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 12/27/2018] [Indexed: 11/18/2022] Open
Abstract
Tsetse flies (Glossina spp.) vector pathogenic trypanosomes (Trypanosoma spp.) in sub-Saharan Africa. These parasites cause human and animal African trypanosomiases, which are debilitating diseases that inflict an enormous socio-economic burden on inhabitants of endemic regions. Current disease control strategies rely primarily on treating infected animals and reducing tsetse population densities. However, relevant programs are costly, labor intensive and difficult to sustain. As such, novel strategies aimed at reducing tsetse vector competence require development. Herein we investigated whether Kosakonia cowanii Zambiae (Kco_Z), which confers Anopheles gambiae with resistance to Plasmodium, is able to colonize tsetse and induce a trypanosome refractory phenotype in the fly. Kco_Z established stable infections in tsetse's gut and exhibited no adverse effect on the fly's survival. Flies with established Kco_Z infections in their gut were significantly more refractory to infection with two distinct trypanosome species (T. congolense, 6% infection; T. brucei, 32% infection) than were age-matched flies that did not house the exogenous bacterium (T. congolense, 36% infected; T. brucei, 70% infected). Additionally, 52% of Kco_Z colonized tsetse survived infection with entomopathogenic Serratia marcescens, compared with only 9% of their wild-type counterparts. These parasite and pathogen refractory phenotypes result from the fact that Kco_Z acidifies tsetse's midgut environment, which inhibits trypanosome and Serratia growth and thus infection establishment. Finally, we determined that Kco_Z infection does not impact the fecundity of male or female tsetse, nor the ability of male flies to compete with their wild-type counterparts for mates. We propose that Kco_Z could be used as one component of an integrated strategy aimed at reducing the ability of tsetse to transmit pathogenic trypanosomes.
Collapse
Affiliation(s)
- Brian L Weiss
- Yale School of Public Health, Department of Epidemiology of Microbial Diseases, New Haven, Connecticut, United States of America
| | - Michele A Maltz
- Southern Connecticut State University, New Haven, Connecticut, United States of America
| | - Aurélien Vigneron
- Yale School of Public Health, Department of Epidemiology of Microbial Diseases, New Haven, Connecticut, United States of America
| | - Yineng Wu
- Yale School of Public Health, Department of Epidemiology of Microbial Diseases, New Haven, Connecticut, United States of America
| | - Katharine S Walter
- Yale School of Public Health, Department of Epidemiology of Microbial Diseases, New Haven, Connecticut, United States of America
| | - Michelle B O'Neill
- Yale School of Public Health, Department of Epidemiology of Microbial Diseases, New Haven, Connecticut, United States of America
| | - Jingwen Wang
- Yale School of Public Health, Department of Epidemiology of Microbial Diseases, New Haven, Connecticut, United States of America
| | - Serap Aksoy
- Yale School of Public Health, Department of Epidemiology of Microbial Diseases, New Haven, Connecticut, United States of America
| |
Collapse
|
29
|
Gibson W, Peacock L. Fluorescent proteins reveal what trypanosomes get up to inside the tsetse fly. Parasit Vectors 2019; 12:6. [PMID: 30609932 PMCID: PMC6320599 DOI: 10.1186/s13071-018-3204-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 11/19/2018] [Indexed: 12/12/2022] Open
Abstract
The discovery and development of fluorescent proteins for the investigation of living cells and whole organisms has been a major advance in biomedical research. This approach was quickly exploited by parasitologists, particularly those studying single-celled protists. Here we describe some of our experiments to illustrate how fluorescent proteins have helped to reveal what trypanosomes get up to inside the tsetse fly. Fluorescent proteins turned the tsetse fly from a "black box" into a bright showcase to track trypanosome migration and development within the insect. Crosses of genetically modified red and green fluorescent trypanosomes produced yellow fluorescent hybrids and established the "when" and "where" of trypanosome sexual reproduction inside the fly. Fluorescent-tagging endogenous proteins enabled us to identify the meiotic division stage and gametes inside the salivary glands of the fly and thus elucidate the mechanism of sexual reproduction in trypanosomes. Without fluorescent proteins we would still be in the "dark ages" of understanding what trypanosomes get up to inside the tsetse fly.
Collapse
Affiliation(s)
- Wendy Gibson
- School of Biological Sciences, University of Bristol, Bristol, BS8 1TQ UK
| | - Lori Peacock
- School of Biological Sciences, University of Bristol, Bristol, BS8 1TQ UK
- School of Clinical Veterinary Science, University of Bristol, Langford, Bristol, BS40 7DU UK
| |
Collapse
|
30
|
Calvo-Alvarez E, Cren-Travaillé C, Crouzols A, Rotureau B. A new chimeric triple reporter fusion protein as a tool for in vitro and in vivo multimodal imaging to monitor the development of African trypanosomes and Leishmania parasites. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2018; 63:391-403. [PMID: 29339220 DOI: 10.1016/j.meegid.2018.01.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 01/09/2018] [Accepted: 01/11/2018] [Indexed: 12/20/2022]
Abstract
Trypanosomiases and leishmaniases, caused by a group of related protist parasites, are Neglected Tropical Diseases currently threatening >500 million people worldwide. Reporter proteins have revolutionised the research on infectious diseases and have opened up new advances in the understanding of trypanosomatid-borne diseases in terms of both biology, pathogenesis and drug development. Here, we describe the generation and some applications of a new chimeric triple reporter fusion protein combining the red-shifted firefly luciferase PpyREH9 and the tdTomato red fluorescent protein, fused by the TY1 tag. Expressed in both Trypanosoma brucei brucei and Leishmania major transgenic parasites, this construct was successfully assessed on different state-of-the-art imaging technologies, at different scales ranging from whole organism to cellular level, both in vitro and in vivo in murine models. For T. b. brucei, the usefulness of this triple marker to monitor the entire parasite cycle in both tsetse flies and mice was further demonstrated. This stable reporter allows to qualitatively and quantitatively scrutinize in real-time several crucial aspects of the parasite's development, including the development of African trypanosomes in the dermis of the mammalian host. We briefly discuss developments in bio-imaging technologies and highlight how we could improve our understanding of parasitism by combining the genetic engineering of parasites to the one of the hosting organisms in which they complete their developmental program.
Collapse
Affiliation(s)
- Estefania Calvo-Alvarez
- Trypanosome Transmission Group, Trypanosome Cell Biology Unit, Institut Pasteur and INSERM U1201, Paris, France.
| | - Christelle Cren-Travaillé
- Trypanosome Transmission Group, Trypanosome Cell Biology Unit, Institut Pasteur and INSERM U1201, Paris, France
| | - Aline Crouzols
- Trypanosome Transmission Group, Trypanosome Cell Biology Unit, Institut Pasteur and INSERM U1201, Paris, France
| | - Brice Rotureau
- Trypanosome Transmission Group, Trypanosome Cell Biology Unit, Institut Pasteur and INSERM U1201, Paris, France
| |
Collapse
|
31
|
Krafsur ES, Maudlin I. Tsetse fly evolution, genetics and the trypanosomiases - A review. INFECTION GENETICS AND EVOLUTION 2018; 64:185-206. [PMID: 29885477 DOI: 10.1016/j.meegid.2018.05.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 05/30/2018] [Accepted: 05/31/2018] [Indexed: 01/27/2023]
Abstract
This reviews work published since 2007. Relative efforts devoted to the agents of African trypanosomiasis and their tsetse fly vectors are given by the numbers of PubMed accessions. In the last 10 years PubMed citations number 3457 for Trypanosoma brucei and 769 for Glossina. The development of simple sequence repeats and single nucleotide polymorphisms afford much higher resolution of Glossina and Trypanosoma population structures than heretofore. Even greater resolution is offered by partial and whole genome sequencing. Reproduction in T. brucei sensu lato is principally clonal although genetic recombination in tsetse salivary glands has been demonstrated in T. b. brucei and T. b. rhodesiense but not in T. b. gambiense. In the past decade most genetic attention was given to the chief human African trypanosomiasis vectors in subgenus Nemorhina e.g., Glossina f. fuscipes, G. p. palpalis, and G. p. gambiense. The chief interest in Nemorhina population genetics seemed to be finding vector populations sufficiently isolated to enable efficient and long-lasting suppression. To this end estimates were made of gene flow, derived from FST and its analogues, and Ne, the size of a hypothetical population equivalent to that under study. Genetic drift was greater, gene flow and Ne typically lesser in savannah inhabiting tsetse (subgenus Glossina) than in riverine forms (Nemorhina). Population stabilities were examined by sequential sampling and genotypic analysis of nuclear and mitochondrial genomes in both groups and found to be stable. Gene frequencies estimated in sequential samplings differed by drift and allowed estimates of effective population numbers that were greater for Nemorhina spp than Glossina spp. Prospects are examined of genetic methods of vector control. The tsetse long generation time (c. 50 d) is a major contraindication to any suggested genetic method of tsetse population manipulation. Ecological and modelling research convincingly show that conventional methods of targeted insecticide applications and traps/targets can achieve cost-effective reduction in tsetse densities.
Collapse
Affiliation(s)
- E S Krafsur
- Department of Entomology, Iowa State University, Ames, IA 50011, USA.
| | - Ian Maudlin
- School of Biomedical Sciences, The University of Edinburgh, Scotland, UK
| |
Collapse
|
32
|
Matetovici I, Van Den Abbeele J. Thioester-containing proteins in the tsetse fly (Glossina) and their response to trypanosome infection. INSECT MOLECULAR BIOLOGY 2018; 27. [PMID: 29528164 PMCID: PMC5969219 DOI: 10.1111/imb.12382] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Thioester-containing proteins (TEPs) are conserved proteins with a role in innate immune immunity. In the current study, we characterized the TEP family in the genome of six tsetse fly species (Glossina spp.). Tsetse flies are the biological vectors of several African trypanosomes, which cause sleeping sickness in humans or nagana in livestock. The analysis of the tsetse TEP sequences revealed information about their structure, evolutionary relationships and expression profiles under both normal and trypanosome infection conditions. Phylogenetic analysis of the family showed that tsetse flies harbour a genomic expansion of specific TEPs that are not found in other dipterans. We found a general expression of all TEP genes in the alimentary tract, mouthparts and salivary glands. Glossina morsitans and Glossina palpalis TEP genes display a tissue-specific expression pattern with some that are markedly up-regulated when the fly is infected with the trypanosome parasite. A different TEP response was observed to infection with Trypanosoma brucei compared to Trypanosoma congolense, indicating that the tsetse TEP response is trypanosome-specific. These findings are suggestive for the involvement of the TEP family in tsetse innate immunity, with a possible role in the control of the trypanosome parasite.
Collapse
Affiliation(s)
- I. Matetovici
- Unit of Veterinary Protozoology, Department of Biomedical SciencesInstitute of Tropical Medicine Antwerp (ITM)AntwerpBelgium
| | - J. Van Den Abbeele
- Unit of Veterinary Protozoology, Department of Biomedical SciencesInstitute of Tropical Medicine Antwerp (ITM)AntwerpBelgium
| |
Collapse
|
33
|
Peacock L, Kay C, Bailey M, Gibson W. Shape-shifting trypanosomes: Flagellar shortening followed by asymmetric division in Trypanosoma congolense from the tsetse proventriculus. PLoS Pathog 2018; 14:e1007043. [PMID: 29772025 PMCID: PMC5957336 DOI: 10.1371/journal.ppat.1007043] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 04/18/2018] [Indexed: 11/18/2022] Open
Abstract
Trypanosomatids such as Leishmania and Trypanosoma are digenetic, single-celled, parasitic flagellates that undergo complex life cycles involving morphological and metabolic changes to fit them for survival in different environments within their mammalian and insect hosts. According to current consensus, asymmetric division enables trypanosomatids to achieve the major morphological rearrangements associated with transition between developmental stages. Contrary to this view, here we show that the African trypanosome Trypanosoma congolense, an important livestock pathogen, undergoes extensive cell remodelling, involving shortening of the cell body and flagellum, during its transition from free-swimming proventricular forms to attached epimastigotes in vitro. Shortening of the flagellum was associated with accumulation of PFR1, a major constituent of the paraflagellar rod, in the mid-region of the flagellum where it was attached to the substrate. However, the PFR1 depot was not essential for attachment, as it accumulated several hours after initial attachment of proventricular trypanosomes. Detergent and CaCl2 treatment failed to dislodge attached parasites, demonstrating the robust nature of flagellar attachment to the substrate; the PFR1 depot was also unaffected by these treatments. Division of the remodelled proventricular trypanosome was asymmetric, producing a small daughter cell. Each mother cell went on to produce at least one more daughter cell, while the daughter trypanosomes also proliferated, eventually resulting in a dense culture of epimastigotes. Here, by observing the synchronous development of the homogeneous population of trypanosomes in the tsetse proventriculus, we have been able to examine the transition from proventricular forms to attached epimastigotes in detail in T. congolense. This transition is difficult to observe in vivo as it happens inside the mouthparts of the tsetse fly. In T. brucei, this transition is achieved by asymmetric division of long trypomastigotes in the proventriculus, yielding short epimastigotes, which go on to colonise the salivary glands. Thus, despite their close evolutionary relationship and shared developmental route within the vector, T. brucei and T. congolense have evolved different ways of accomplishing the same developmental transition from proventricular form to attached epimastigote.
Collapse
Affiliation(s)
- Lori Peacock
- School of Biological Sciences, University of Bristol, Bristol, United Kingdom
- Bristol Veterinary School, University of Bristol, Langford, Bristol, United Kingdom
| | - Christopher Kay
- School of Biological Sciences, University of Bristol, Bristol, United Kingdom
| | - Mick Bailey
- Bristol Veterinary School, University of Bristol, Langford, Bristol, United Kingdom
| | - Wendy Gibson
- School of Biological Sciences, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
34
|
A fine-tuned vector-parasite dialogue in tsetse's cardia determines peritrophic matrix integrity and trypanosome transmission success. PLoS Pathog 2018; 14:e1006972. [PMID: 29614112 PMCID: PMC5898766 DOI: 10.1371/journal.ppat.1006972] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 04/13/2018] [Accepted: 03/13/2018] [Indexed: 01/19/2023] Open
Abstract
Arthropod vectors have multiple physical and immunological barriers that impede the development and transmission of parasites to new vertebrate hosts. These barriers include the peritrophic matrix (PM), a chitinous barrier that separates the blood bolus from the midgut epithelia and modulates vector-pathogens interactions. In tsetse flies, a sleeve-like PM is continuously produced by the cardia organ located at the fore- and midgut junction. African trypanosomes, Trypanosoma brucei, must bypass the PM twice; first to colonize the midgut and secondly to reach the salivary glands (SG), to complete their transmission cycle in tsetse. However, not all flies with midgut infections develop mammalian transmissible SG infections—the reasons for which are unclear. Here, we used transcriptomics, microscopy and functional genomics analyses to understand the factors that regulate parasite migration from midgut to SG. In flies with midgut infections only, parasites fail to cross the PM as they are eliminated from the cardia by reactive oxygen intermediates (ROIs)—albeit at the expense of collateral cytotoxic damage to the cardia. In flies with midgut and SG infections, expression of genes encoding components of the PM is reduced in the cardia, and structural integrity of the PM barrier is compromised. Under these circumstances trypanosomes traverse through the newly secreted and compromised PM. The process of PM attrition that enables the parasites to re-enter into the midgut lumen is apparently mediated by components of the parasites residing in the cardia. Thus, a fine-tuned dialogue between tsetse and trypanosomes at the cardia determines the outcome of PM integrity and trypanosome transmission success. Insects are responsible for transmission of parasites that cause deadly diseases in humans and animals. Understanding the key factors that enhance or interfere with parasite transmission processes can result in new control strategies. Here, we report that a proportion of tsetse flies with African trypanosome infections in their midgut can prevent parasites from migrating to the salivary glands, albeit at the expense of collateral damage. In a subset of flies with gut infections, the parasites manipulate the integrity of a midgut barrier, called the peritrophic matrix, and reach the salivary glands for transmission to the next mammal. Either targeting parasite manipulative processes or enhancing peritrophic matrix integrity could reduce parasite transmission.
Collapse
|
35
|
Bateta R, Wang J, Wu Y, Weiss BL, Warren WC, Murilla GA, Aksoy S, Mireji PO. Tsetse fly (Glossina pallidipes) midgut responses to Trypanosoma brucei challenge. Parasit Vectors 2017; 10:614. [PMID: 29258576 PMCID: PMC5738168 DOI: 10.1186/s13071-017-2569-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 12/04/2017] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Tsetse flies (Glossina spp.) are the prominent vector of African trypanosome parasites (Trypanosoma spp.) in sub-Saharan Africa, and Glossina pallidipes is the most widely distributed species in Kenya. This species displays strong resistance to infection by parasites, which are typically eliminated in the midgut shortly after acquisition from the mammalian host. Although extensive molecular information on immunity for the related species Glossina morsitans morsitans exists, similar information is scarce for G. pallidipes. METHODS To determine temporal transcriptional responses of G. pallidipes to Trypanosoma brucei brucei challenge, we conducted Illumina based RNA-seq on midgut organ and carcass from teneral females G. pallidipes at 24 and 48 h post-challenge (hpc) with T. b. brucei relative to their respective controls that received normal blood meals (without the parasite). We used a suite of bioinformatics tools to determine differentially expressed and enriched transcripts between and among tissues, and to identify expanded transcripts in G. pallidipes relative to their orthologs G. m. morsitans. RESULTS Midgut transcripts induced at 24 hpc encoded proteins were associated with lipid remodelling, proteolysis, collagen metabolism, apoptosis, and cell growth. Midgut transcripts induced at 48 hpc encoded proteins linked to embryonic growth and development, serine endopeptidases and proteosomal degradation of the target protein, mRNA translation and neuronal development. Temporal expression of immune responsive transcripts at 48 relative to 24 hpc was pronounced, indicative of a gradual induction of host immune responses the following challenge. We also searched for G. m. morsitans orthologous groups that may have experienced expansions in the G. pallidipes genome. We identified ten expanded groups in G. pallidipes with putative immunity-related functions, which may play a role in the higher refractoriness exhibited by this species. CONCLUSIONS There appears to be a lack of strong immune responses elicited by gut epithelia of teneral adults. This in combination with a compromised peritrophic matrix at this stage during the initial phase of T. b. brucei challenge may facilitate the increased parasite infection establishment noted in teneral flies relative to older adults. Although teneral flies are more susceptible than older adults, the majority of tenerals are still able to eliminate parasite infections. Hence, robust responses elicited at a later time point, such as 72 hpc, may clear parasite infections from the majority of flies. The expanded G. m. morsitans orthologous groups in G. pallidipes may also be functionally associated with the enhanced refractoriness to trypanosome infections reported in G. pallidipes relative to G. m. morsitans.
Collapse
Affiliation(s)
- Rosemary Bateta
- Department of Biochemistry, Biotechnology Research Institute, Kenya Agricultural and Livestock Research Organization, P.O. Box 362, Kikuyu, Kenya
- Department of Biochemistry and Molecular Biology, Egerton University, P.O. Box 536, Njoro, Kenya
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT USA
| | - Jingwen Wang
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT USA
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200433 China
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200433 China
| | - Yineng Wu
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT USA
| | - Brian L. Weiss
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT USA
| | - Wesley C. Warren
- McDonnell Genome Institute, Washington University School of Medicine, 4444 Forest Park Ave., Campus Box 8501, St Louis, MO 63108 USA
| | - Grace A. Murilla
- Department of Biochemistry, Biotechnology Research Institute, Kenya Agricultural and Livestock Research Organization, P.O. Box 362, Kikuyu, Kenya
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT USA
| | - Serap Aksoy
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT USA
| | - Paul O. Mireji
- Department of Biochemistry, Biotechnology Research Institute, Kenya Agricultural and Livestock Research Organization, P.O. Box 362, Kikuyu, Kenya
- Department of Biochemistry and Molecular Biology, Egerton University, P.O. Box 536, Njoro, Kenya
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT USA
- Centre for Geographic Medicine Research - Coast, Kenya Medical Research Institute, P. O. Box 428-80108, Kilifi, Kenya
| |
Collapse
|
36
|
Bombaça ACS, Dias FDA, Ennes-Vidal V, Garcia-Gomes ADS, Sorgine MHF, d'Avila-Levy CM, Menna-Barreto RFS. Hydrogen peroxide resistance in Strigomonas culicis: Effects on mitochondrial functionality and Aedes aegypti interaction. Free Radic Biol Med 2017; 113:255-266. [PMID: 28993269 DOI: 10.1016/j.freeradbiomed.2017.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 10/04/2017] [Accepted: 10/05/2017] [Indexed: 12/27/2022]
Abstract
Reactive oxygen species (ROS) are toxic molecules involved in several biological processes such as cellular signaling, proliferation, differentiation and cell death. Adaptations to oxidative environments are crucial for the success of the colonization of insects by protozoa. Strigomonas culicis is a monoxenic trypanosomatid found in the midgut of mosquitoes and presenting a life cycle restricted to the epimastigote form. Among S. culicis peculiarities, there is an endosymbiotic bacterium in the cytoplasm, which completes essential biosynthetic routes of the host cell and may represent an intermediary evolutive step in organelle origin, thus constituting an interesting model for evolutive researches. In this work, we induced ROS resistance in wild type S. culicis epimastigotes by the incubation with increasing concentrations of hydrogen peroxide (H2O2), and compared the oxidative and energetic metabolisms among wild type, wild type-H2O2 resistant and aposymbiotic strains. Resistant protozoa were less sensitive to the oxidative challenge and more dependent on oxidative phosphorylation, which was demonstrated by higher oxygen consumption and mitochondrial membrane potential, increased activity of complexes II-III and IV, increased complex II gene expression and higher ATP production. Furthermore, the wild type-H2O2 resistant strain produced reduced ROS levels and showed lower lipid peroxidation, as well as an increase in gene expression of antioxidant enzymes and thiol-dependent peroxidase activity. On the other hand, the aposymbiotic strain showed impaired mitochondrial function, higher H2O2 production and deficient antioxidant response. The induction of H2O2 resistance also led to a remarkable increase in Aedes aegypti midgut binding in vitro and colonization in vivo, indicating that both the pro-oxidant environment in the mosquito gut and the oxidative stress susceptibility regulate S. culicis population in invertebrates.
Collapse
Affiliation(s)
| | - Felipe de Almeida Dias
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vitor Ennes-Vidal
- Laboratório de Estudos Integrados em Protozoologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Aline Dos Santos Garcia-Gomes
- Laboratório de Estudos Integrados em Protozoologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil; Laboratório de Microbiologia, Instituto Federal de Educação, Ciência e Tecnologia do Rio de Janeiro, Campus Rio de Janeiro, Brazil
| | - Marcos Henrique Ferreira Sorgine
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Claudia Masini d'Avila-Levy
- Laboratório de Estudos Integrados em Protozoologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | | |
Collapse
|
37
|
Gibson W, Peacock L, Hutchinson R. Microarchitecture of the tsetse fly proboscis. Parasit Vectors 2017; 10:430. [PMID: 28927459 PMCID: PMC5606065 DOI: 10.1186/s13071-017-2367-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 09/07/2017] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Tsetse flies (genus Glossina) are large blood-sucking dipteran flies that are important as vectors of human and animal trypanosomiasis in sub-Saharan Africa. Tsetse anatomy has been well described, including detailed accounts of the functional anatomy of the proboscis for piercing host skin and sucking up blood. The proboscis also serves as the developmental site for the infective metacyclic stages of several species of pathogenic livestock trypanosomes that are inoculated into the host with fly saliva. To understand the physical environment in which these trypanosomes develop, we have re-examined the microarchitecture of the tsetse proboscis. RESULTS We examined proboscises from male and female flies of Glossina pallidipes using light microscopy and scanning electron microscopy (SEM). Each proboscis was removed from the fly head and either examined intact or dissected into the three constituent components: Labrum, labium and hypopharynx. Our light and SEM images reaffirm earlier observations that the tsetse proboscis is a formidably armed weapon, well-adapted for piercing skin, and provide comparative data for G. pallidipes. In addition, the images reveal that the hypopharynx, the narrow tube that delivers saliva to the wound site, ends in a remarkably ornate and complex structure with around ten finger-like projections, each adorned with sucker-like protrusions, contradicting previous descriptions that show a simple, bevelled end like a hypodermic needle. The function of the finger-like projections is speculative; they appear to be flexible and may serve to protect the hypopharynx from influx of blood or microorganisms, or control the flow of saliva. Proboscises were examined after colonisation by Trypanosoma congolense savannah. Consistent with the idea that colonisation commences in the region nearest the foregut, the highest densities of trypanosomes were found in the region of the labrum proximal to the bulb, although high densities were also found in other regions of the labrum. Trypanosomes were visible through the thin wall of the hypopharynx by both light microscopy and SEM. CONCLUSIONS We highlight the remarkable architecture of the tsetse proboscis, in particular the intricate structure of the distal end of the hypopharynx. Further work is needed to elucidate the function of this intriguing structure.
Collapse
Affiliation(s)
- Wendy Gibson
- School of Biological Sciences, University of Bristol, Bristol, BS8 1TQ UK
| | - Lori Peacock
- School of Biological Sciences, University of Bristol, Bristol, BS8 1TQ UK
- School of Clinical Veterinary Science, University of Bristol, Langford, Bristol, BS40 7DU UK
| | - Rachel Hutchinson
- School of Biological Sciences, University of Bristol, Bristol, BS8 1TQ UK
| |
Collapse
|
38
|
Jacob F, Melachio TT, Njitchouang GR, Gimonneau G, Njiokou F, Abate L, Christen R, Reveillaud J, Geiger A. Intestinal Bacterial Communities of Trypanosome-Infected and Uninfected Glossina palpalis palpalis from Three Human African Trypanomiasis Foci in Cameroon. Front Microbiol 2017; 8:1464. [PMID: 28824591 PMCID: PMC5541443 DOI: 10.3389/fmicb.2017.01464] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 07/20/2017] [Indexed: 11/27/2022] Open
Abstract
Glossina sp. the tsetse fly that transmits trypanosomes causing the Human or the Animal African Trypanosomiasis (HAT or AAT) can harbor symbiotic bacteria that are known to play a crucial role in the fly's vector competence. We hypothesized that other bacteria could be present, and that some of them could also influence the fly's vector competence. In this context the objectives of our work were: (a) to characterize the bacteria that compose the G. palpalis palpalis midgut bacteriome, (b) to evidence possible bacterial community differences between trypanosome-infected and non-infected fly individuals from a given AAT and HAT focus or from different foci using barcoded Illumina sequencing of the hypervariable V3-V4 region of the 16S rRNA gene. Forty G. p. palpalis flies, either infected by Trypanosoma congolense or uninfected were sampled from three trypanosomiasis foci in Cameroon. A total of 143 OTUs were detected in the midgut samples. Most taxa were identified at the genus level, nearly 50% at the species level; they belonged to 83 genera principally within the phyla Actinobacteria, Bacteroidetes, Firmicutes, and Proteobacteria. Prominent representatives included Wigglesworthia (the fly's obligate symbiont), Serratia, and Enterobacter hormaechei. Wolbachia was identified for the first time in G. p. palpalis. The average number of bacterial species per tsetse sample was not significantly different regarding the fly infection status, and the hierarchical analysis based on the differences in bacterial community structure did not provide a clear clustering between infected and non-infected flies. Finally, the most important result was the evidence of the overall very large diversity of intestinal bacteria which, except for Wigglesworthia, were unevenly distributed over the sampled flies regardless of their geographic origin and their trypanosome infection status.
Collapse
Affiliation(s)
- Franck Jacob
- UMR INTERTRYP, Institut de Recherche pour le Développement-CIRAD, CIRAD TA A-17/GMontpellier, France
| | - Trésor T Melachio
- Parasitology and Ecology Laboratory, Department of Animal Biology and Physiology, Faculty of Science, University of Yaounde 1Yaounde, Cameroon
| | - Guy R Njitchouang
- Parasitology and Ecology Laboratory, Department of Animal Biology and Physiology, Faculty of Science, University of Yaounde 1Yaounde, Cameroon
| | - Geoffrey Gimonneau
- UMR INTERTRYP, Institut de Recherche pour le Développement-CIRAD, CIRAD TA A-17/GMontpellier, France
| | - Flobert Njiokou
- Parasitology and Ecology Laboratory, Department of Animal Biology and Physiology, Faculty of Science, University of Yaounde 1Yaounde, Cameroon
| | - Luc Abate
- UMR MIVEGEC, Institut de Recherche pour le Développement 224-Centre National de la Recherche Scientifique 5290Montpellier, France
| | - Richard Christen
- UMR 7138, Systématique Adaptation Evolution, Université de Nice-Sophia AntipolisNice, France
| | - Julie Reveillaud
- Institut National de la Recherche Agronomique, UMR 1309 ASTREMontpellier, France.,CIRAD, UMR ASTREMontpellier, France
| | - Anne Geiger
- UMR INTERTRYP, Institut de Recherche pour le Développement-CIRAD, CIRAD TA A-17/GMontpellier, France
| |
Collapse
|
39
|
Azambuja P, Garcia ES, Waniek PJ, Vieira CS, Figueiredo MB, Gonzalez MS, Mello CB, Castro DP, Ratcliffe NA. Rhodnius prolixus: from physiology by Wigglesworth to recent studies of immune system modulation by Trypanosoma cruzi and Trypanosoma rangeli. JOURNAL OF INSECT PHYSIOLOGY 2017; 97:45-65. [PMID: 27866813 DOI: 10.1016/j.jinsphys.2016.11.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 11/04/2016] [Accepted: 11/16/2016] [Indexed: 06/06/2023]
Abstract
This review is dedicated to the memory of Professor Sir Vincent B. Wigglesworth (VW) in recognition of his many pioneering contributions to insect physiology which, even today, form the basis of modern-day research in this field. Insects not only make vital contributions to our everyday lives by their roles in pollination, balancing eco-systems and provision of honey and silk products, but they are also outstanding models for studying the pathogenicity of microorganisms and the functioning of innate immunity in humans. In this overview, the immune system of the triatomine bug, Rhodnius prolixus, is considered which is most appropriate to this dedication as this insect species was the favourite subject of VW's research. Herein are described recent developments in knowledge of the functioning of the R. prolixus immune system. Thus, the roles of the cellular defences, such as phagocytosis and nodule formation, as well as the role of eicosanoids, ecdysone, antimicrobial peptides, reactive oxygen and nitrogen radicals, and the gut microbiota in the immune response of R. prolixus are described. The details of many of these were unknown to VW although his work gives indications of his awareness of the importance to R. prolixus of cellular immunity, antibacterial activity, prophenoloxidase and the gut microbiota. This description of R. prolixus immunity forms a backdrop to studies on the interaction of the parasitic flagellates, Trypanosoma cruzi and Trypanosoma rangeli, with the host defences of this important insect vector. These parasites remarkably utilize different strategies to avoid/modulate the triatomine immune response in order to survive in the extremely hostile host environments present in the vector gut and haemocoel. Much recent information has also been gleaned on the remarkable diversity of the immune system in the R. prolixus gut and its interaction with trypanosome parasites. This new data is reviewed and gaps in our knowledge of R. prolixus immunity are identified as subjects for future endeavours. Finally, the publication of the T. cruzi, T. rangeli and R. prolixus genomes, together with the use of modern molecular techniques, should lead to the enhanced identification of the determinants of infection derived from both the vector and the parasites which, in turn, could form targets for new molecular-based control strategies.
Collapse
Affiliation(s)
- P Azambuja
- Laboratório de Bioquímica e Fisiologia de Insetos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/FIOCRUZ), Rio de Janeiro, RJ, Brazil; Departamento de Entomologia Molecular, Instituto Nacional de Entomologia Molecular (INCT-EM), Rio de Janeiro, RJ, Brazil.
| | - E S Garcia
- Laboratório de Bioquímica e Fisiologia de Insetos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/FIOCRUZ), Rio de Janeiro, RJ, Brazil; Departamento de Entomologia Molecular, Instituto Nacional de Entomologia Molecular (INCT-EM), Rio de Janeiro, RJ, Brazil.
| | - P J Waniek
- Laboratório de Bioquímica e Fisiologia de Insetos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/FIOCRUZ), Rio de Janeiro, RJ, Brazil.
| | - C S Vieira
- Laboratório de Bioquímica e Fisiologia de Insetos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/FIOCRUZ), Rio de Janeiro, RJ, Brazil.
| | - M B Figueiredo
- Laboratório de Bioquímica e Fisiologia de Insetos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/FIOCRUZ), Rio de Janeiro, RJ, Brazil.
| | - M S Gonzalez
- Laboratório de Biologia de Insetos, Universidade Federal Fluminense, Niterói, RJ, Brazil.
| | - C B Mello
- Laboratório de Biologia de Insetos, Universidade Federal Fluminense, Niterói, RJ, Brazil.
| | - D P Castro
- Laboratório de Bioquímica e Fisiologia de Insetos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (IOC/FIOCRUZ), Rio de Janeiro, RJ, Brazil; Departamento de Entomologia Molecular, Instituto Nacional de Entomologia Molecular (INCT-EM), Rio de Janeiro, RJ, Brazil.
| | - N A Ratcliffe
- Laboratório de Biologia de Insetos, Universidade Federal Fluminense, Niterói, RJ, Brazil; Department of Biosciences, College of Science, Swansea University, Singleton Park, Swansea, Wales, United Kingdom.
| |
Collapse
|
40
|
Clayton AL, Enomoto S, Su Y, Dale C. The regulation of antimicrobial peptide resistance in the transition to insect symbiosis. Mol Microbiol 2017; 103:958-972. [PMID: 27987256 DOI: 10.1111/mmi.13598] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2016] [Indexed: 01/02/2023]
Abstract
Many bacteria utilize two-component systems consisting of a sensor kinase and a transcriptional response regulator to detect environmental signals and modulate gene expression for adaptation. The response regulator PhoP and its cognate sensor kinase PhoQ compose a two-component system known for its role in responding to low levels of Mg2+ , Ca2+ , pH and to the presence of antimicrobial peptides and activating the expression of genes involved in adaptation to host association. Compared with their free-living relatives, mutualistic insect symbiotic bacteria inhabit a static environment where the requirement for sensory functions is expected to be relaxed. The insect symbiont, Sodalis glossinidius, requires PhoP to resist killing by host derived antimicrobial peptides. However, the S. glossinidius PhoQ was found to be insensitive to Mg2+ , Ca2+ and pH. Here they show that Sodalis praecaptivus, a close non host-associated relative of S. glossinidius, utilizes a magnesium sensing PhoP-PhoQ and an uncharacterized MarR-like transcriptional regulator (Sant_4061) to control antimicrobial peptide resistance in vitro. While the inactivation of phoP, phoQ or Sant_4061 completely retards the growth of S. praecaptivus in the presence of an antimicrobial peptide in vitro, inactivation of both phoP and Sant_4061 is necessary to abrogate growth of this bacterium in an insect host.
Collapse
Affiliation(s)
- Adam L Clayton
- Department of Biology, University of Utah, Salt Lake City, UT, USA
| | | | - Yinghua Su
- Department of Biology, University of Utah, Salt Lake City, UT, USA
| | - Colin Dale
- Department of Biology, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
41
|
Caljon G, Van Reet N, De Trez C, Vermeersch M, Pérez-Morga D, Van Den Abbeele J. The Dermis as a Delivery Site of Trypanosoma brucei for Tsetse Flies. PLoS Pathog 2016; 12:e1005744. [PMID: 27441553 PMCID: PMC4956260 DOI: 10.1371/journal.ppat.1005744] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 06/15/2016] [Indexed: 12/13/2022] Open
Abstract
Tsetse flies are the sole vectors of Trypanosoma brucei parasites that cause sleeping sickness. Our knowledge on the early interface between the infective metacyclic forms and the mammalian host skin is currently highly limited. Glossina morsitans flies infected with fluorescently tagged T. brucei parasites were used in this study to initiate natural infections in mice. Metacyclic trypanosomes were found to be highly infectious through the intradermal route in sharp contrast with blood stream form trypanosomes. Parasite emigration from the dermal inoculation site resulted in detectable parasite levels in the draining lymph nodes within 18 hours and in the peripheral blood within 42 h. A subset of parasites remained and actively proliferated in the dermis. By initiating mixed infections with differentially labeled parasites, dermal parasites were unequivocally shown to arise from the initial inoculum and not from a re-invasion from the blood circulation. Scanning electron microscopy demonstrated intricate interactions of these skin-residing parasites with adipocytes in the connective tissue, entanglement by reticular fibers of the periadipocytic baskets and embedment between collagen bundles. Experimental transmission experiments combined with molecular parasite detection in blood fed flies provided evidence that dermal trypanosomes can be acquired from the inoculation site immediately after the initial transmission. High resolution thermographic imaging also revealed that intradermal parasite expansion induces elevated skin surface temperatures. Collectively, the dermis represents a delivery site of the highly infective metacyclic trypanosomes from which the host is systemically colonized and where a proliferative subpopulation remains that is physically constrained by intricate interactions with adipocytes and collagen fibrous structures. Sleeping sickness is caused by trypanosomes that are transmitted by the blood feeding tsetse flies. The present study has established an experimental transmission model with fluorescently labeled parasites in mice that allows us to study their fate following natural transmission by a tsetse fly bite. Parasites that arise in the tsetse salivary glands were found to be highly infective following inoculation in the mammalian skin in contrast with previous observations made for trypanosomes purified from the blood stream. This study unveiled that a proportion of parasites is retained in the skin and actively proliferates close to the initial inoculation site resulting in significantly elevated skin temperatures. This retention was linked to interaction with fat cells and collagen fibrous structures. Experimental transmission experiments were able to demonstrate that parasites can be acquired from the inoculation site immediately after the initial transmission.
Collapse
Affiliation(s)
- Guy Caljon
- Unit of Veterinary Protozoology, Department of Biomedical Sciences, Institute of Tropical Medicine Antwerp (ITM), Antwerp, Belgium
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Wilrijk, Belgium
- Laboratory of Myeloid Cell Immunology, VIB Inflammation Research Center, Ghent, Belgium
- * E-mail: (GC); (JVDA)
| | - Nick Van Reet
- Unit of Parasite Diagnostics, Department of Biomedical Sciences, Institute of Tropical Medicine Antwerp (ITM), Antwerp, Belgium
| | - Carl De Trez
- Unit of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Structural Biology Research Center (SBRC), VIB, Brussels, Belgium
| | - Marjorie Vermeersch
- Center for Microscopy and Molecular Imaging (CMMI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - David Pérez-Morga
- Center for Microscopy and Molecular Imaging (CMMI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
- Laboratory of Molecular Parasitology, Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | - Jan Van Den Abbeele
- Unit of Veterinary Protozoology, Department of Biomedical Sciences, Institute of Tropical Medicine Antwerp (ITM), Antwerp, Belgium
- * E-mail: (GC); (JVDA)
| |
Collapse
|
42
|
Peacock L, Bailey M, Gibson W. Dynamics of gamete production and mating in the parasitic protist Trypanosoma brucei. Parasit Vectors 2016; 9:404. [PMID: 27439767 PMCID: PMC4955137 DOI: 10.1186/s13071-016-1689-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 07/10/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Sexual reproduction in Plasmodium falciparum and Trypanosoma brucei occurs in the insect vector and is important in generating hybrid strains with different combinations of parental characteristics. Production of hybrid parasite genotypes depends on the likelihood of co-infection of the vector with multiple strains. In mosquitoes, existing infection with Plasmodium facilitates the establishment of a second infection, although the asynchronicity of gamete production subsequently prevents mating. In the trypanosome/tsetse system, flies become increasingly refractory to infection as they age, so the likelihood of a fly acquiring a second infection also decreases. This effectively restricts opportunities for trypanosome mating to co-infections picked up by the fly on its first feed, unless an existing infection increases the chance of successful second infection as in the Plasmodium/mosquito system. RESULTS Using green and red fluorescent trypanosomes, we compared the rates of trypanosome infection and hybrid production in flies co-infected on the first feed, co-infected on a subsequent feed 18 days after emergence, or fed sequentially with each trypanosome clone 18 days apart. Infection rates were highest in the midguts and salivary glands (SG) of flies that received both trypanosome clones in their first feed, and were halved when the infected feed was delayed to day 18. In flies fed the two trypanosome clones sequentially, the second clone often failed to establish a midgut infection and consequently was not present in the SG. Nevertheless, hybrids were recovered from all three groups of infected flies. Meiotic stages and gametes were produced continuously from day 11 to 42 after the infective feed, and in sequentially infected flies, the co-occurrence of gametes led to hybrid formation. CONCLUSIONS We found that a second trypanosome strain can establish infection in the tsetse SG 18 days after the first infected feed, with co-mingling of gametes and production of trypanosome hybrids. Establishment of the second strain was severely compromised by the strong immune response of the fly to the existing infection. Although sequential infection provides an opportunity for trypanosome mating, the easiest way for a tsetse fly to acquire a mixed infection is by feeding on a co-infected host.
Collapse
Affiliation(s)
- Lori Peacock
- School of Biological Sciences, University of Bristol, Bristol, BS8 1TQ, UK.,School of Clinical Veterinary Science, University of Bristol, Langford, Bristol, BS40 7DU, UK
| | - Mick Bailey
- School of Clinical Veterinary Science, University of Bristol, Langford, Bristol, BS40 7DU, UK
| | - Wendy Gibson
- School of Biological Sciences, University of Bristol, Bristol, BS8 1TQ, UK.
| |
Collapse
|
43
|
Caljon G, De Muylder G, Durnez L, Jennes W, Vanaerschot M, Dujardin JC. Alice in microbes' land: adaptations and counter-adaptations of vector-borne parasitic protozoa and their hosts. FEMS Microbiol Rev 2016; 40:664-85. [PMID: 27400870 DOI: 10.1093/femsre/fuw018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2016] [Indexed: 12/24/2022] Open
Abstract
In the present review, we aim to provide a general introduction to different facets of the arms race between pathogens and their hosts/environment, emphasizing its evolutionary aspects. We focus on vector-borne parasitic protozoa, which have to adapt to both invertebrate and vertebrate hosts. Using Leishmania, Trypanosoma and Plasmodium as main models, we review successively (i) the adaptations and counter-adaptations of parasites and their invertebrate host, (ii) the adaptations and counter-adaptations of parasites and their vertebrate host and (iii) the impact of human interventions (chemotherapy, vaccination, vector control and environmental changes) on these adaptations. We conclude by discussing the practical impact this knowledge can have on translational research and public health.
Collapse
Affiliation(s)
- Guy Caljon
- Institute of Tropical Medicine, Department of Biomedical Sciences, Nationalestraat 155, B-2000 Antwerp, Belgium University of Antwerp, Department of Biomedical Sciences, Laboratory of Microbiology, Parasitology and Health, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| | - Géraldine De Muylder
- Institute of Tropical Medicine, Department of Biomedical Sciences, Nationalestraat 155, B-2000 Antwerp, Belgium
| | - Lies Durnez
- Institute of Tropical Medicine, Department of Biomedical Sciences, Nationalestraat 155, B-2000 Antwerp, Belgium
| | - Wim Jennes
- Institute of Tropical Medicine, Department of Biomedical Sciences, Nationalestraat 155, B-2000 Antwerp, Belgium
| | - Manu Vanaerschot
- Institute of Tropical Medicine, Department of Biomedical Sciences, Nationalestraat 155, B-2000 Antwerp, Belgium Columbia University, College of Physicians and Surgeons, Department of Microbiology and Immunology, Fidock Lab, New York, NY 10032, USA
| | - Jean-Claude Dujardin
- Institute of Tropical Medicine, Department of Biomedical Sciences, Nationalestraat 155, B-2000 Antwerp, Belgium University of Antwerp, Department of Biomedical Sciences, Laboratory of Microbiology, Parasitology and Health, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| |
Collapse
|
44
|
Aksoy E, Vigneron A, Bing X, Zhao X, O'Neill M, Wu YN, Bangs JD, Weiss BL, Aksoy S. Mammalian African trypanosome VSG coat enhances tsetse's vector competence. Proc Natl Acad Sci U S A 2016; 113:6961-6. [PMID: 27185908 PMCID: PMC4922192 DOI: 10.1073/pnas.1600304113] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Tsetse flies are biological vectors of African trypanosomes, the protozoan parasites responsible for causing human and animal trypanosomiases across sub-Saharan Africa. Currently, no vaccines are available for disease prevention due to antigenic variation of the Variant Surface Glycoproteins (VSG) that coat parasites while they reside within mammalian hosts. As a result, interference with parasite development in the tsetse vector is being explored to reduce disease transmission. A major bottleneck to infection occurs as parasites attempt to colonize tsetse's midgut. One critical factor influencing this bottleneck is the fly's peritrophic matrix (PM), a semipermeable, chitinous barrier that lines the midgut. The mechanisms that enable trypanosomes to cross this barrier are currently unknown. Here, we determined that as parasites enter the tsetse's gut, VSG molecules released from trypanosomes are internalized by cells of the cardia-the tissue responsible for producing the PM. VSG internalization results in decreased expression of a tsetse microRNA (mir-275) and interferes with the Wnt-signaling pathway and the Iroquois/IRX transcription factor family. This interference reduces the function of the PM barrier and promotes parasite colonization of the gut early in the infection process. Manipulation of the insect midgut homeostasis by the mammalian parasite coat proteins is a novel function and indicates that VSG serves a dual role in trypanosome biology-that of facilitating transmission through its mammalian host and insect vector. We detail critical steps in the course of trypanosome infection establishment that can serve as novel targets to reduce the tsetse's vector competence and disease transmission.
Collapse
Affiliation(s)
- Emre Aksoy
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, CT 06520
| | - Aurélien Vigneron
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, CT 06520
| | - XiaoLi Bing
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, CT 06520
| | - Xin Zhao
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, CT 06520
| | - Michelle O'Neill
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, CT 06520
| | - Yi-Neng Wu
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, CT 06520
| | - James D Bangs
- Department of Microbiology and Immunology, University at Buffalo (SUNY), Buffalo, NY 14214
| | - Brian L Weiss
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, CT 06520;
| | - Serap Aksoy
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, CT 06520;
| |
Collapse
|
45
|
Lane-Serff H, MacGregor P, Peacock L, Macleod OJ, Kay C, Gibson W, Higgins MK, Carrington M. Evolutionary diversification of the trypanosome haptoglobin-haemoglobin receptor from an ancestral haemoglobin receptor. eLife 2016; 5. [PMID: 27083048 PMCID: PMC4889325 DOI: 10.7554/elife.13044] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Accepted: 04/14/2016] [Indexed: 01/27/2023] Open
Abstract
The haptoglobin-haemoglobin receptor of the African trypanosome species, Trypanosoma brucei, is expressed when the parasite is in the bloodstream of the mammalian host, allowing it to acquire haem through the uptake of haptoglobin-haemoglobin complexes. Here we show that in Trypanosoma congolense this receptor is instead expressed in the epimastigote developmental stage that occurs in the tsetse fly, where it acts as a haemoglobin receptor. We also present the structure of the T. congolense receptor in complex with haemoglobin. This allows us to propose an evolutionary history for this receptor, charting the structural and cellular changes that took place as it adapted from a role in the insect to a new role in the mammalian host. DOI:http://dx.doi.org/10.7554/eLife.13044.001 Trypanosomes are single-celled parasites that infect a range of animal hosts. These parasites need a molecule called haem to grow properly and are mostly spread by insects that feed on the blood of mammals. Most haem in mammals is found in red blood cells and is bound to a protein called haemoglobin. When it is released from these cells, haemoglobin forms a complex with another protein called haptoglobin as well. The best-studied trypanosomes from Africa have a receptor protein on their surface that recognizes the haptoglobin-haemoglobin complex and allows the parasites to obtain haem from their hosts. An African trypanosome called T. brucei causes sleeping sickness in humans, and has a receptor that can only recognize haemoglobin when it is in complex with haptoglobin. However, few trypanosome receptors have been studied to date, and so it was not clear if they all work in the same way. Trypanosoma congolense is a trypanosome that has a big impact on livestock farmers in sub-Saharan Africa and infects cattle, pigs and goats. Lane-Serff, MacGregor et al. now report that the receptor protein from T. congolense can bind to haemoglobin on its own. A technique called X-ray crystallography was used to reveal the three-dimensional structure of the T. congolense receptor and haemoglobin in fine detail. Further experiments then confirmed that the receptor actually binds more strongly to haemoglobin than it does to the haptoglobin-haemoglobin complex. Experiments with living parasites showed that T. congolense produces its receptor when it is in the mouthparts of its insect host, the tsetse fly. This is unlike what occurs in T. brucei, which only produces its receptor while it is in the bloodstream of its mammalian host. Lane-Serff, MacGregor et al. suggest that T. congolense’s receptor is more like the receptor found in ancestor of the trypanosomes. This means that, at least once during the evolution of these parasites, this receptor evolved from being a haemoglobin receptor produced in the tsetse fly to a haptoglobin-haemoglobin receptor produced in an infected mammal. The next step is to investigate the details of the role played by the T. congolense receptor when the parasite is in the tsetse fly. It will also be important to understand how this parasite is still able to grow in the mammalian host’s bloodstream even though it does not produce much of the receptor during this stage. DOI:http://dx.doi.org/10.7554/eLife.13044.002
Collapse
Affiliation(s)
- Harriet Lane-Serff
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Paula MacGregor
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Lori Peacock
- School of Veterinary Science, University of Bristol, Bristol, United Kingdom.,School of Biological Sciences, University of Bristol, Bristol, United Kingdom
| | - Olivia Js Macleod
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Christopher Kay
- School of Biological Sciences, University of Bristol, Bristol, United Kingdom
| | - Wendy Gibson
- School of Biological Sciences, University of Bristol, Bristol, United Kingdom
| | - Matthew K Higgins
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Mark Carrington
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
46
|
Avila CCDC, Peacock L, Machado FC, Gibson W, Schenkman S, Carrington M, Castilho BA. Phosphorylation of eIF2α on Threonine 169 is not required for Trypanosoma brucei cell cycle arrest during differentiation. Mol Biochem Parasitol 2016; 205:16-21. [PMID: 26996431 PMCID: PMC4850487 DOI: 10.1016/j.molbiopara.2016.03.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 03/15/2016] [Accepted: 03/16/2016] [Indexed: 11/17/2022]
Abstract
Pleomorphic T. brucei expressing an eIF2α phosphorylation site mutant were made. The mutation did not prevent normal arrest and differentiation into stumpy forms. Mutants differentiate into procyclic forms in vitro and in tsetse flies.
The trypanosome life cycle consists of a series of developmental forms each adapted to an environment in the relevant insect and/or mammalian host. The differentiation process from the mammalian bloodstream form to the insect-midgut procyclic form in Trypanosoma brucei occurs in two steps in vivo. First proliferating ‘slender' bloodstream forms differentiate to non-dividing ‘stumpy' forms arrested in G1. Second, in response to environmental cues, stumpy bloodstream forms re-enter the cell cycle and start to proliferate as procyclic forms after a lag during which both cell morphology and gene expression are modified. Nearly all arrested cells have lower rates of protein synthesis when compared to the proliferating equivalent. In eukaryotes, one mechanism used to regulate the overall rate of protein synthesis involves phosphorylation of the alpha subunit of initiation factor eIF2 (eIF2α). The effect of eIF2α phosphorylation is to prevent the action of eIF2B, the guanine nucleotide exchange factor that activates eIF2 for the next rounds of initiation. To investigate the role of the phosphorylation of eIF2α in the life cycle of T. brucei, a cell line was made with a single eIF2α gene that contained the phosphorylation site, threonine 169, mutated to alanine. These cells were capable of differentiating from proliferating bloodstream form cells into arrested stumpy forms in mice and into procyclic forms in vitro and in tsetse flies. These results indicate that translation attenuation mediated by the phosphorylation of eIF2α on threonine 169 is not necessary for the cell cycle arrest associated with these differentiation processes.
Collapse
Affiliation(s)
- Carla Cristi D C Avila
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Lori Peacock
- Department of Clinical Veterinary Science, University of Bristol, Langford, Bristol BS40 5DU, UK; School of Biological Sciences, University of Bristol, Bristol BS8 1TQ, UK
| | - Fabricio Castro Machado
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Wendy Gibson
- School of Biological Sciences, University of Bristol, Bristol BS8 1TQ, UK
| | - Sergio Schenkman
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Mark Carrington
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1QW, UK.
| | - Beatriz A Castilho
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
47
|
Machado-Silva A, Cerqueira PG, Grazielle-Silva V, Gadelha FR, Peloso EDF, Teixeira SMR, Machado CR. How Trypanosoma cruzi deals with oxidative stress: Antioxidant defence and DNA repair pathways. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2016; 767:8-22. [DOI: 10.1016/j.mrrev.2015.12.003] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 12/22/2015] [Accepted: 12/23/2015] [Indexed: 02/06/2023]
|
48
|
Hamidou Soumana I, Klopp C, Ravel S, Nabihoudine I, Tchicaya B, Parrinello H, Abate L, Rialle S, Geiger A. RNA-seq de novo Assembly Reveals Differential Gene Expression in Glossina palpalis gambiensis Infected with Trypanosoma brucei gambiense vs. Non-Infected and Self-Cured Flies. Front Microbiol 2015; 6:1259. [PMID: 26617594 PMCID: PMC4643127 DOI: 10.3389/fmicb.2015.01259] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 10/29/2015] [Indexed: 12/24/2022] Open
Abstract
Trypanosoma brucei gambiense (Tbg), causing the sleeping sickness chronic form, completes its developmental cycle within the tsetse fly vector Glossina palpalis gambiensis (Gpg) before its transmission to humans. Within the framework of an anti-vector disease control strategy, a global gene expression profiling of trypanosome infected (susceptible), non-infected, and self-cured (refractory) tsetse flies was performed, on their midguts, to determine differential genes expression resulting from in vivo trypanosomes, tsetse flies (and their microbiome) interactions. An RNAseq de novo assembly was achieved. The assembled transcripts were mapped to reference sequences for functional annotation. Twenty-four percent of the 16,936 contigs could not be annotated, possibly representing untranslated mRNA regions, or Gpg- or Tbg-specific ORFs. The remaining contigs were classified into 65 functional groups. Only a few transposable elements were present in the Gpg midgut transcriptome, which may represent active transpositions and play regulatory roles. One thousand three hundred and seventy three genes differentially expressed (DEGs) between stimulated and non-stimulated flies were identified at day-3 post-feeding; 52 and 1025 between infected and self-cured flies at 10 and 20 days post-feeding, respectively. The possible roles of several DEGs regarding fly susceptibility and refractoriness are discussed. The results provide new means to decipher fly infection mechanisms, crucial to develop anti-vector control strategies.
Collapse
Affiliation(s)
| | - Christophe Klopp
- Institut National de la Recherche Agronomique, GenoToul, UR875 Castanet-Tolosan, France
| | - Sophie Ravel
- UMR 177, Institut de Recherche Pour le Développement-CIRAD, CIRAD TA A-17/G Montpellier, France
| | | | - Bernadette Tchicaya
- UMR 177, Institut de Recherche Pour le Développement-CIRAD, CIRAD TA A-17/G Montpellier, France
| | - Hugues Parrinello
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5203, Institut de Génomique Fonctionnelle Montpellier, France ; Institut National de la Santé et de la Recherche Médicale U661 Montpellier, France ; Universités de Montpellier 1 and 2, UMR 5203 Montpellier, France ; Montpellier GenomiX, Institut de Génomique Fonctionnelle Montpellier, France
| | - Luc Abate
- UMR MIVEGEC (Institut de Recherche pour le Développement 224-Centre National de la Recherche Scientifique 5290-UM1-UM2), Institut de Recherche pour le Développement Montpellier, France
| | - Stéphanie Rialle
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5203, Institut de Génomique Fonctionnelle Montpellier, France ; Institut National de la Santé et de la Recherche Médicale U661 Montpellier, France ; Universités de Montpellier 1 and 2, UMR 5203 Montpellier, France ; Montpellier GenomiX, Institut de Génomique Fonctionnelle Montpellier, France
| | - Anne Geiger
- UMR 177, Institut de Recherche Pour le Développement-CIRAD, CIRAD TA A-17/G Montpellier, France
| |
Collapse
|
49
|
Liaisons dangereuses: sexual recombination among pathogenic trypanosomes. Res Microbiol 2015; 166:459-66. [DOI: 10.1016/j.resmic.2015.05.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 05/18/2015] [Accepted: 05/18/2015] [Indexed: 11/20/2022]
|
50
|
Geiger A, Hamidou Soumana I, Tchicaya B, Rofidal V, Decourcelle M, Santoni V, Hem S. Differential expression of midgut proteins in Trypanosoma brucei gambiense-stimulated vs. non-stimulated Glossina palpalis gambiensis flies. Front Microbiol 2015; 6:444. [PMID: 26029185 PMCID: PMC4428205 DOI: 10.3389/fmicb.2015.00444] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 04/24/2015] [Indexed: 11/13/2022] Open
Abstract
The unicellular pathogenic protozoan Trypanosoma brucei gambiense is responsible for the chronic form of sleeping sickness. This vector-borne disease is transmitted to humans by the tsetse fly of the group Glossina palpalis, including the subspecies G. p. gambiensis, in which the parasite completes its developmental cycle. Sleeping sickness control strategies can therefore target either the human host or the fly vector. Indeed, suppression of one step in the parasite developmental cycle could abolish parasite transmission to humans, with consequences on the spreading of the disease. In order to develop this type of approach, we have identified, at the proteome level, events resulting from the tripartite interaction between the tsetse fly G. p. gambiensis, its microbiome, and the trypanosome. Proteomes were analyzed from four biological replicates of midguts from flies sampled 3 days post-feeding on either a trypanosome-infected (stimulated flies) or a non-infected (non-stimulated flies) bloodmeal. Over 500 proteins were identified in the midguts of flies from both feeding groups, 13 of which were shown to be differentially expressed in trypanosome-stimulated vs. non-stimulated flies. Functional annotation revealed that several of these proteins have important functions that could be involved in modulating the fly infection process by trypanosomes (and thus fly vector competence), including anti-oxidant and anti-apoptotic, cellular detoxifying, trypanosome agglutination, and immune stimulating or depressive effects. The results show a strong potential for diminishing or even disrupting fly vector competence, and their application holds great promise for improving the control of sleeping sickness.
Collapse
Affiliation(s)
- Anne Geiger
- UMR 177, Institut de Recherche pour le Développement-CIRAD, CIRAD TA A-17/G Montpellier, France
| | | | - Bernadette Tchicaya
- UMR 177, Institut de Recherche pour le Développement-CIRAD, CIRAD TA A-17/G Montpellier, France
| | - Valérie Rofidal
- Plateforme de Spectrométrie de Masse Protéomique - MSPP, Biochimie et Physiologie Moléculaire des Plantes - UMR 5004 Centre National de la Recherche Scientifique/UMR 0386 INRA/Montpellier SupAgro/Université Montpellier II Montpellier, France
| | - Mathilde Decourcelle
- Plateforme de Spectrométrie de Masse Protéomique - MSPP, Biochimie et Physiologie Moléculaire des Plantes - UMR 5004 Centre National de la Recherche Scientifique/UMR 0386 INRA/Montpellier SupAgro/Université Montpellier II Montpellier, France
| | - Véronique Santoni
- Plateforme de Spectrométrie de Masse Protéomique - MSPP, Biochimie et Physiologie Moléculaire des Plantes - UMR 5004 Centre National de la Recherche Scientifique/UMR 0386 INRA/Montpellier SupAgro/Université Montpellier II Montpellier, France
| | - Sonia Hem
- Plateforme de Spectrométrie de Masse Protéomique - MSPP, Biochimie et Physiologie Moléculaire des Plantes - UMR 5004 Centre National de la Recherche Scientifique/UMR 0386 INRA/Montpellier SupAgro/Université Montpellier II Montpellier, France
| |
Collapse
|