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
|
Ombura FLO, Malele I, Abd-Alla AM, Akutse KS, Ajene IJ, Khamis FM. Potential of entomopathogenic fungi for Glossina austeni control: insights into microbiome alterations and implications on sustainable management of the pest. INSECT SCIENCE 2025. [PMID: 40287897 DOI: 10.1111/1744-7917.70052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/04/2025] [Accepted: 03/11/2025] [Indexed: 04/29/2025]
Abstract
Glossina austeni Newstead (Diptera: Glossinidae) is a competent vector of the trypanosomes causing human African trypanosomiasis and the African animal trypanosomosis. Management of this pest has primarily involved trapping methods, Sterile Insect Technique, and research into vector competence-symbiotic interactions. Nevertheless, the use of entomopathogenic fungi (EPF) in integrated pest management programs for G. austeni control remains limited. Moreover, different tsetse fly species exhibit varying susceptibility to different EPF strains, indicating that no single strain is universally effective. Therefore, our study aimed to identify candidate EPF isolates for G. austeni management, evaluate the effects of temperature on the radial growth of these potent isolates, and assess the impact of the candidate EPF on the gut microbiome of G. austeni. Consequently, 16 Metarhizium anisopliae (Metschn.) Sorokin isolates were screened against G. austeni using dry conidia in an infection chamber, with the most virulent isolates having LT50 values of 3.95-9.37 d. Temperature significantly influenced the radial growth, conidia germination, and yield of these strains. There were also significant differences in conidia acquisition, retention and transmission between male and female G. austeni flies. Furthermore, all conidia receivers carried sufficient conidia, 5 d post-interaction with EPF-challenged conidia donors. Microbiome analysis revealed Wigglesworthia, Serratia, Klebsiella, and Escherichia as the most abundant taxa. Among the M. anisopliae isolates, ICIPE 82 exhibited the fastest radial growth and highest thermostability, hence selected as a potential biopesticide candidate for managing G. austeni. This study demonstrates the efficacy and potential of M. anisopliae ICIPE 82 as a biopesticide for controlling G. austeni.
Collapse
Affiliation(s)
| | - Imna Malele
- Tanzania Veterinary Laboratory Agency, Dar es Salaam, Tanzania
| | - Adly Mm Abd-Alla
- Insect Pest Control Laboratory, Joint FAO/IAEA Centre of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency, Vienna, Austria
| | - Komivi Senyo Akutse
- International Centre of Insect Physiology and Ecology (icipe), Nairobi, Kenya
- Unit of Environmental Sciences and Management, North-West University, Potchefstroom, South Africa
| | - Inusa Jacob Ajene
- International Centre of Insect Physiology and Ecology (icipe), Nairobi, Kenya
| | - Fathiya Mbarak Khamis
- International Centre of Insect Physiology and Ecology (icipe), Nairobi, Kenya
- Department of Zoology and Entomology, University of Pretoria, Hatfield, South Africa
| |
Collapse
|
3
|
Omondi ZN, Caner A, Arserim SK. Trypanosomes and gut microbiota interactions in triatomine bugs and tsetse flies: A vectorial perspective. MEDICAL AND VETERINARY ENTOMOLOGY 2024; 38:253-268. [PMID: 38651684 DOI: 10.1111/mve.12723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 04/05/2024] [Indexed: 04/25/2024]
Abstract
Triatomines (kissing bugs) and tsetse flies (genus: Glossina) are natural vectors of Trypanosoma cruzi and Trypanosoma brucei, respectively. T. cruzi is the causative agent of Chagas disease, endemic in Latin America, while T. brucei causes African sleeping sickness disease in sub-Saharan Africa. Both triatomines and tsetse flies are host to a diverse community of gut microbiota that co-exist with the parasites in the gut. Evidence has shown that the gut microbiota of both vectors plays a key role in parasite development and transmission. However, knowledge on the mechanism involved in parasite-microbiota interaction remains limited and scanty. Here, we attempt to analyse Trypanosoma spp. and gut microbiota interactions in tsetse flies and triatomines, with a focus on understanding the possible mechanisms involved by reviewing published articles on the subject. We report that interactions between Trypanosoma spp. and gut microbiota can be both direct and indirect. In direct interactions, the gut microbiota directly affects the parasite via the formation of biofilms and the production of anti-parasitic molecules, while on the other hand, Trypanosoma spp. produces antimicrobial proteins to regulate gut microbiota of the vector. In indirect interactions, the parasite and gut bacteria affect each other through host vector-activated processes such as immunity and metabolism. Although we are beginning to understand how gut microbiota interacts with the Trypanosoma parasites, there is still a need for further studies on functional role of gut microbiota in parasite development to maximize the use of symbiotic bacteria in vector and parasite control.
Collapse
Affiliation(s)
- Zeph Nelson Omondi
- Department of Biology, Faculty of Science, Ege University, Izmir, Turkey
| | - Ayşe Caner
- Department of Parasitology, Faculty of Medicine, Ege University, Izmir, Turkey
- Department of Basic Oncology, Institute of Health Sciences, Ege University, Izmir, Turkey
| | - Suha Kenan Arserim
- Vocational School of Health Sciences, Manisa Celal Bayar University, Manisa, Turkey
| |
Collapse
|
4
|
Tsagmo JM, Njiokou F, Dziedziech A, Rofidal V, Hem S, Geiger A. Protein abundance in the midgut of wild tsetse flies (Glossina palpalis palpalis) naturally infected by Trypanosoma congolense s.l. MEDICAL AND VETERINARY ENTOMOLOGY 2023; 37:723-736. [PMID: 37357577 DOI: 10.1111/mve.12676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 06/08/2023] [Indexed: 06/27/2023]
Abstract
Tsetse flies (Glossina spp.) are major vectors of African trypanosomes, causing either Human or Animal African Trypanosomiasis (HAT or AAT). Several approaches have been developed to control the disease, among which is the anti-vector Sterile Insect Technique. Another approach to anti-vector strategies could consist of controlling the fly's vector competence through hitherto unidentified regulatory factors (genes, proteins, biological pathways, etc.). The present work aims to evaluate the protein abundance in the midgut of wild tsetse flies (Glossina palpalis palpalis) naturally infected by Trypanosoma congolense s.l. Infected and non-infected flies were sampled in two HAT/AAT foci in Southern Cameroon. After dissection, the proteomes from the guts of parasite-infected flies were compared to that of uninfected flies to identify quantitative and/or qualitative changes associated with infection. Among the proteins with increased abundance were fructose-1,6-biphosphatase, membrane trafficking proteins, death proteins (or apoptosis proteins) and SERPINs (inhibitor of serine proteases, enzymes considered as trypanosome virulence factors) that displayed the highest increased abundance. The present study, together with previous proteomic and transcriptomic studies on the secretome of trypanosomes from tsetse fly gut extracts, provides data to be explored in further investigations on, for example, mammal host immunisation or on fly vector competence modification via para-transgenic approaches.
Collapse
Affiliation(s)
- Jean Marc Tsagmo
- INTERTRYP, Institut de Recherche pour le Développement, University of Montpellier, Montpellier, France
- Faculty of Science, University of Yaoundé I, Yaounde, Cameroon
- Trypanosome Transmission Group, Trypanosome Cell Biology Unit, Department of Parasites and Insect Vectors and INSERM U1201, Institut Pasteur, Paris, France
| | - Flobert Njiokou
- Faculty of Science, University of Yaoundé I, Yaounde, Cameroon
| | - Alexis Dziedziech
- Biology of Host-Parasite Interactions Unit, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France
| | - Valerie Rofidal
- IPSiM, Univ Montpellier, CNRS, INRAE, Institut Agro, Montpellier, France
| | - Sonia Hem
- IPSiM, Univ Montpellier, CNRS, INRAE, Institut Agro, Montpellier, France
| | - Anne Geiger
- INTERTRYP, Institut de Recherche pour le Développement, University of Montpellier, Montpellier, France
- Faculty of Science, University of Yaoundé I, Yaounde, Cameroon
- Center for Research on Filariasis and Other Tropical Diseases (CRFilMT), Yaounde, Cameroon
| |
Collapse
|
5
|
Dieng MM, Dera KSM, Moyaba P, Ouedraogo GMS, Demirbas-Uzel G, Gstöttenmayer F, Mulandane FC, Neves L, Mdluli S, Rayaisse JB, Belem AMG, Pagabeleguem S, de Beer CJ, Parker AG, Van Den Abbeele J, Mach RL, Vreysen MJB, Abd-Alla AMM. Prevalence of Trypanosoma and Sodalis in wild populations of tsetse flies and their impact on sterile insect technique programmes for tsetse eradication. Sci Rep 2022; 12:3322. [PMID: 35228552 PMCID: PMC8885713 DOI: 10.1038/s41598-022-06699-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/03/2022] [Indexed: 11/24/2022] Open
Abstract
The sterile insect technique (SIT) is an environment friendly and sustainable method to manage insect pests of economic importance through successive releases of sterile irradiated males of the targeted species to a defined area. A mating of a sterile male with a virgin wild female will result in no offspring, and ultimately lead to the suppression or eradication of the targeted population. Tsetse flies, vectors of African Trypanosoma, have a highly regulated and defined microbial fauna composed of three bacterial symbionts that may have a role to play in the establishment of Trypanosoma infections in the flies and hence, may influence the vectorial competence of the released sterile males. Sodalis bacteria seem to interact with Trypanosoma infection in tsetse flies. Field-caught tsetse flies of ten different taxa and from 15 countries were screened using PCR to detect the presence of Sodalis and Trypanosoma species and analyse their interaction. The results indicate that the prevalence of Sodalis and Trypanosoma varied with country and tsetse species. Trypanosome prevalence was higher in east, central and southern African countries than in west African countries. Tsetse fly infection rates with Trypanosoma vivax and T. brucei sspp were higher in west African countries, whereas tsetse infection with T. congolense and T. simiae, T. simiae (tsavo) and T. godfreyi were higher in east, central and south African countries. Sodalis prevalence was high in Glossina morsitans morsitans and G. pallidipes but absent in G. tachinoides. Double and triple infections with Trypanosoma taxa and coinfection of Sodalis and Trypanosoma were rarely observed but it occurs in some taxa and locations. A significant Chi square value (< 0.05) seems to suggest that Sodalis and Trypanosoma infection correlate in G. palpalis gambiensis, G. pallidipes and G. medicorum. Trypanosoma infection seemed significantly associated with an increased density of Sodalis in wild G. m. morsitans and G. pallidipes flies, however, there was no significant impact of Sodalis infection on trypanosome density.
Collapse
Affiliation(s)
- Mouhamadou M Dieng
- Insect Pest Control Laboratory, Joint FAO/IAEA Centre of Nuclear Techniques in Food and Agriculture, 1400, Vienna, Austria
| | - Kiswend-Sida M Dera
- Insect Pest Control Laboratory, Joint FAO/IAEA Centre of Nuclear Techniques in Food and Agriculture, 1400, Vienna, Austria.,Insectarium de Bobo Dioulasso-Campagne d'Eradication de la mouche tsetse et de la Trypanosomose (IBD-CETT), 01 BP 1087, Bobo Dioulasso 01, Burkina Faso
| | - Percy Moyaba
- Epidemiology, Vectors and Parasites, Agricultural Research Council-Onderstepoort Veterinary Research, Pretoria, South Africa
| | - Gisele M S Ouedraogo
- Insectarium de Bobo Dioulasso-Campagne d'Eradication de la mouche tsetse et de la Trypanosomose (IBD-CETT), 01 BP 1087, Bobo Dioulasso 01, Burkina Faso
| | - Guler Demirbas-Uzel
- Insect Pest Control Laboratory, Joint FAO/IAEA Centre of Nuclear Techniques in Food and Agriculture, 1400, Vienna, Austria
| | - Fabian Gstöttenmayer
- Insect Pest Control Laboratory, Joint FAO/IAEA Centre of Nuclear Techniques in Food and Agriculture, 1400, Vienna, Austria
| | - Fernando C Mulandane
- University Eduardo Mondlane, Centro de Biotecnologia, Av. de Moçambique Km 1.5, Maputo, Mozambique
| | - Luis Neves
- University Eduardo Mondlane, Centro de Biotecnologia, Av. de Moçambique Km 1.5, Maputo, Mozambique.,Department of Veterinary Tropical Diseases, University of Pretoria, Private Bag X04, Onderstepoort, 0110, South Africa
| | - Sihle Mdluli
- Epidemiology Unit, Department of Veterinary Services, PO Box 4192, Manzini, Eswatini
| | - Jean-Baptiste Rayaisse
- Centre International de Recherche-Développement sur l'Elevage en zone Subhumide (CIRDES), 01 BP 454, Bobo-Dioulasso 01, Burkina Faso
| | | | - Soumaïla Pagabeleguem
- Insectarium de Bobo Dioulasso-Campagne d'Eradication de la mouche tsetse et de la Trypanosomose (IBD-CETT), 01 BP 1087, Bobo Dioulasso 01, Burkina Faso.,University of Dedougou, B.P. 176, Dédougou 01, Burkina Faso
| | - Chantel J de Beer
- Insect Pest Control Laboratory, Joint FAO/IAEA Centre of Nuclear Techniques in Food and Agriculture, 1400, Vienna, Austria.,Epidemiology, Vectors and Parasites, Agricultural Research Council-Onderstepoort Veterinary Research, Pretoria, South Africa
| | | | | | - Robert L Mach
- Institute of Chemical, Environmental, and Bioscience Engineering, Vienna University of Technology, Gumpendorfer Straße 1a, 1060, Vienna, Austria
| | - Marc J B Vreysen
- Insect Pest Control Laboratory, Joint FAO/IAEA Centre of Nuclear Techniques in Food and Agriculture, 1400, Vienna, Austria
| | - Adly M M Abd-Alla
- Insect Pest Control Laboratory, Joint FAO/IAEA Centre of Nuclear Techniques in Food and Agriculture, 1400, Vienna, Austria.
| |
Collapse
|
6
|
Gashururu S. R, Maingi N, Githigia SM, Gasana MN, Odhiambo PO, Getange DO, Habimana R, Cecchi G, Zhao W, Gashumba J, Bargul JL, Masiga DK. Occurrence, diversity and distribution of Trypanosoma infections in cattle around the Akagera National Park, Rwanda. PLoS Negl Trop Dis 2021; 15:e0009929. [PMID: 34910728 PMCID: PMC8726506 DOI: 10.1371/journal.pntd.0009929] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 01/04/2022] [Accepted: 10/19/2021] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND African Trypanosomiases threaten the life of both humans and animals. Trypanosomes are transmitted by tsetse and other biting flies. In Rwanda, the African Animal Trypanosomiasis (AAT) endemic area is mainly around the tsetse-infested Akagera National Park (NP). The study aimed to identify Trypanosoma species circulating in cattle, their genetic diversity and distribution around the Akagera NP. METHODOLOGY A cross-sectional study was carried out in four districts, where 1,037 cattle blood samples were collected. The presence of trypanosomes was determined by microscopy, immunological rapid test VerY Diag and PCR coupled with High-Resolution Melt (HRM) analysis. A parametric test (ANOVA) was used to compare the mean Packed cell Volume (PCV) and trypanosomes occurrence. The Cohen Kappa test was used to compare the level of agreement between the diagnostic methods. FINDINGS The overall prevalence of trypanosome infections was 5.6%, 7.1% and 18.7% by thin smear, Buffy coat technique and PCR/HRM respectively. Microscopy showed a low sensitivity while a low specificity was shown by the rapid test (VerY Diag). Trypanosoma (T.) congolense was found at a prevalence of 10.7%, T. vivax 5.2%, T. brucei brucei 2% and T. evansi 0.7% by PCR/HRM. This is the first report of T.evansi in cattle in Rwanda. The non-pathogenic T. theileri was also detected. Lower trypanosome infections were observed in Ankole x Friesian breeds than indigenous Ankole. No human-infective T. brucei rhodesiense was detected. There was no significant difference between the mean PCV of infected and non-infected animals (p>0.162). CONCLUSIONS Our study sheds light on the species of animal infective trypanosomes around the Akagera NP, including both pathogenic and non-pathogenic trypanosomes. The PCV estimation is not always an indication of trypanosome infection and the mechanical transmission should not be overlooked. The study confirms that the area around the Akagera NP is affected by AAT, and should, therefore, be targeted by the control activities. AAT impact assessment on cattle production and information on the use of trypanocides are needed to help policymakers prioritise target areas and optimize intervention strategies. Ultimately, these studies will allow Rwanda to advance in the Progressive Control Pathway (PCP) to reduce or eliminate the burden of AAT.
Collapse
Affiliation(s)
- Richard Gashururu S.
- School of Veterinary Medicine, University of Rwanda, Nyagatare, Rwanda
- Faculty of Veterinary Medicine, University of Nairobi, Nairobi, Kenya
- International Centre of Insect Physiology and Ecology (icipe), Nairobi, Kenya
| | - Ndichu Maingi
- Faculty of Veterinary Medicine, University of Nairobi, Nairobi, Kenya
| | | | | | - Peter O. Odhiambo
- International Centre of Insect Physiology and Ecology (icipe), Nairobi, Kenya
| | - Dennis O. Getange
- International Centre of Insect Physiology and Ecology (icipe), Nairobi, Kenya
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | - Richard Habimana
- School of Veterinary Medicine, University of Rwanda, Nyagatare, Rwanda
- Rwanda Food and Drugs Authority, Kigali, Rwanda
| | - Giuliano Cecchi
- Food and Agriculture Organization of the United Nations (FAO), Animal Production and Health Division, Rome, Italy
| | - Weining Zhao
- Food and Agriculture Organization of the United Nations (FAO), Animal Production and Health Division, Rome, Italy
| | | | - Joel L. Bargul
- International Centre of Insect Physiology and Ecology (icipe), Nairobi, Kenya
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | - Daniel K. Masiga
- International Centre of Insect Physiology and Ecology (icipe), Nairobi, Kenya
| |
Collapse
|
7
|
Temporão A, Sanches-Vaz M, Luís R, Nunes-Cabaço H, Smith TK, Prudêncio M, Figueiredo LM. Excreted Trypanosoma brucei proteins inhibit Plasmodium hepatic infection. PLoS Negl Trop Dis 2021; 15:e0009912. [PMID: 34714824 PMCID: PMC8580256 DOI: 10.1371/journal.pntd.0009912] [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: 05/31/2021] [Revised: 11/10/2021] [Accepted: 10/15/2021] [Indexed: 11/25/2022] Open
Abstract
Malaria, a disease caused by Plasmodium parasites, remains a major threat to public health globally. It is the most common disease in patients with sleeping sickness, another parasitic illness, caused by Trypanosoma brucei. We have previously shown that a T. brucei infection impairs a secondary P. berghei liver infection and decreases malaria severity in mice. However, whether this effect requires an active trypanosome infection remained unknown. Here, we show that Plasmodium liver infection can also be inhibited by the serum of a mouse previously infected by T. brucei and by total protein lysates of this kinetoplastid. Biochemical characterisation showed that the anti-Plasmodium activity of the total T. brucei lysates depends on its protein fraction, but is independent of the abundant variant surface glycoprotein. Finally, we found that the protein(s) responsible for the inhibition of Plasmodium infection is/are present within a fraction of ~350 proteins that are excreted to the bloodstream of the host. We conclude that the defence mechanism developed by trypanosomes against Plasmodium relies on protein excretion. This study opens the door to the identification of novel antiplasmodial intervention strategies.
Collapse
Affiliation(s)
- Adriana Temporão
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Margarida Sanches-Vaz
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Rafael Luís
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Helena Nunes-Cabaço
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Terry K. Smith
- Schools of Biology and Chemistry Biomedical Sciences Research Complex, The North Haugh, The University, St. Andrews, Scotland, United Kingdom
| | - Miguel Prudêncio
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Luisa M. Figueiredo
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
8
|
Kipkorir LW, John TK, Owino OB, John O, Robert S, Daniel M, Owino AV. Mouse experiments demonstrate differential pathogenicity and virulence of Trypanosoma brucei rhodesiense strains. Exp Parasitol 2021; 228:108135. [PMID: 34284027 PMCID: PMC7613321 DOI: 10.1016/j.exppara.2021.108135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 06/25/2021] [Accepted: 07/13/2021] [Indexed: 11/24/2022]
Abstract
Trypanosoma brucei rhodesiense is the causative agent for Rhodesian human African trypanosomiasis. The disease is considered acute, but varying clinical outcomes including chronic infections have been observed. The basis for these different clinical manifestations is thought to be associated with a combination of parasite and host factors. In the current study, Trypanosoma brucei rhodesiense strains responsible for varying infection outcomes were sought using mouse model. Clinical rHAT parasite isolates were subjected to PCR tests to confirm presence of the serum resistance associated (SRA) gene. Thereafter, four T. b. rhodesiense isolates were subjected to a comparative pathogenicity study using female Swiss white mice; the parasite strains were compared on the basis of parasitaemia, host survival time, clinical and postmortem biomarkers of infection severity. Isolates identified to cause acute and chronic disease were compared for establishment in insect vector, tsetse fly. The mouse survival time was significantly different (Log-rankp = 0.0001). With mice infected with strain KETRI 3801 exhibiting the shortest survival time (20 days) as compared to those infected with KETRI 3928 that, as controls, survived past the 60 days study period. In addition, development of anaemia was rapid in KETRI 3801 and least in KETRI 3928 infections, and followed the magnitude of survival time. Notably, hepatosplenomegaly was pronounced with longer survival. Mouse weight and feed intake reduced (KETRI 3801 > KETRI 2636 > EATRO 1762) except in KETRI 3928 infections which remained similar to controls. Comparatively, acute to chronic infection outcomes is in the order of KETRI 3801 > KETRI 2636 > EATRO 1762 > KETRI 3928, indicative of predominant role of strain dependent factors. Further, KETRI 3928 strain established better in tsetse as compared to KETRI 3801, suggesting that transmission of strains causing chronic infections could be common. In sum, we have identified Trypanosoma brucei rhodesiense strains that cause acute and chronic infections in mice, that will be valuable in investigating pathogen - host interactions responsible for varying disease outcomes and transmission in African trypanosomiasis.
Collapse
Affiliation(s)
- Limo William Kipkorir
- Department of Biological Sciences, Egerton University, P. O Box, 536-20115, Egerton, Kenya
| | - Thuita Kibuthu John
- Biotechnology Research Institute - Kenya Agricultural and Livestock Research Organisation, Chemotherapy Division, Primate Section, P.O Box, 362-00902, Kikuyu, Kenya; Department of Animal Sciences, Meru University of Science and Technology, P.O Box, 972-60200, Meru, Kenya
| | - Orindi Benedict Owino
- KEMRI-Wellcome Trust Research Programme, CGMRC, P. O Box, 230-80108, Kilifi, Kenya; Department of Public Health and Primary Care, Leuven Biostatistics and Statistical Bioinformatics Centre, Kapucijnenvoer 35, Blok D, Bus 7001, B-3000, Leuven, Belgium
| | - Oidho John
- Biotechnology Research Institute - Kenya Agricultural and Livestock Research Organisation, Chemotherapy Division, Primate Section, P.O Box, 362-00902, Kikuyu, Kenya
| | - Shivairo Robert
- Department of Veterinary and Clinical Studies, Egerton University, P. O Box, 536-20115, Egerton, Kenya
| | - Masiga Daniel
- International Centre of Insect Physiology and Ecology, P. O Box, 30772-000100, Nairobi, Kenya
| | - Adung'a Vincent Owino
- Department of Biochemistry and Molecular Biology, Egerton University, P. O Box, 536-20115, Egerton, Kenya; International Centre of Insect Physiology and Ecology, P. O Box, 30772-000100, Nairobi, Kenya.
| |
Collapse
|
9
|
An African Canine Trypanosomosis Case Import: Is There a Possibility of Creating a Secondary Focus of Trypanosoma congolense Infection in France? Pathogens 2020; 9:pathogens9090709. [PMID: 32867247 PMCID: PMC7558263 DOI: 10.3390/pathogens9090709] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 08/24/2020] [Accepted: 08/26/2020] [Indexed: 11/25/2022] Open
Abstract
African animal trypanosomosis are parasitic diseases caused by several protozoa of the genus Trypanosoma, transmitted by hematophagous insects, essentially tsetse flies, but also, less frequently by Tabanidae and Stomoxidae. They are geolocated in a part of the continent and affect livestock animals and carnivores; dogs are especially sensitive to them. They do not seem to present a zoonotic risk. Despite the chemical prevention with trypanocides for French military working dogs on mission in Côte d’Ivoire, a fatal case induced by Trypanosoma congolense in France after returning from Abidjan raises the question of an imported secondary focus. The clinical case was developed and the causative agent was confirmed by microscopy and PCR methods. The three necessary pillars to create a secondary potential focus are present: the parasite introduction in a new territory, the presence and the propagation vectors, and their proximity with sensitive species.
Collapse
|
10
|
Blood meal sources and bacterial microbiome diversity in wild-caught tsetse flies. Sci Rep 2020; 10:5005. [PMID: 32193415 PMCID: PMC7081217 DOI: 10.1038/s41598-020-61817-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 02/28/2020] [Indexed: 12/02/2022] Open
Abstract
Tsetse flies are the vectors of African trypanosomiasis affecting 36 sub-Saharan countries. Both wild and domestic animals play a crucial role in maintaining the disease-causing parasites (trypanosomes). Thus, the identification of animal reservoirs of trypanosomes is vital for the effective control of African trypanosomiasis. Additionally, the biotic and abiotic factors that drive gut microbiome diversity in tsetse flies are primarily unresolved, especially under natural, field conditions. In this study, we present a comprehensive DNA metabarcoding approach for individual tsetse fly analysis in the identification of mammalian blood meal sources and fly bacterial microbiome composition. We analyzed samples from two endemic foci, Kafue, Zambia collected in June 2017, and Hurungwe, Zimbabwe sampled in April 2014 (pilot study) and detected DNA of various mammals including humans, wild animals, domestic animals and small mammals (rat and bat). The bacterial diversity was relatively similar in flies with different mammalian species DNA, trypanosome infected and uninfected flies, and female and male flies. This study is the first report on bat DNA detection in wild tsetse flies. This study reveals that small mammals such as bats and rats are among the opportunistic blood meal sources for tsetse flies in the wild, and the implication on tsetse biology and ecology needs to be studied.
Collapse
|
11
|
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
|
12
|
Abstract
Parasites elicit several physiological changes in their host to enhance transmission. Little is known about the functional association between parasitism and microbiota-provisioned resources typically dedicated to animal hosts and how these goods may be rerouted to optimize parasite development. This study is the first to identify a specific symbiont-generated metabolite that impacts insect vector competence by facilitating parasite establishment and, thus, eventual transmission. Specifically, we demonstrate that the tsetse fly obligate mutualist Wigglesworthia provisions folate (vitamin B9) that pathogenic African trypanosomes exploit in an effort to successfully establish an infection in the vector’s MG. This process is essential for the parasite to complete its life cycle and be transmitted to a new vertebrate host. Disrupting metabolic contributions provided by the microbiota of arthropod disease vectors may fuel future innovative control strategies while also offering minimal nontarget effects. Many symbionts supplement their host’s diet with essential nutrients. However, whether these nutrients also enhance parasitism is unknown. In this study, we investigated whether folate (vitamin B9) production by the tsetse fly (Glossina spp.) essential mutualist, Wigglesworthia, aids auxotrophic African trypanosomes in completing their life cycle within this obligate vector. We show that the expression of Wigglesworthia folate biosynthesis genes changes with the progression of trypanosome infection within tsetse. The disruption of Wigglesworthia folate production caused a reduction in the percentage of flies that housed midgut (MG) trypanosome infections. However, decreased folate did not prevent MG trypanosomes from migrating to and establishing an infection in the fly’s salivary glands, thus suggesting that nutrient requirements vary throughout the trypanosome life cycle. We further substantiated that trypanosomes rely on symbiont-generated folate by feeding this vitamin to Glossina brevipalpis, which exhibits low trypanosome vector competency and houses Wigglesworthia incapable of producing folate. Folate-supplemented G. brevipalpis flies were significantly more susceptible to trypanosome infection, further demonstrating that this vitamin facilitates parasite infection establishment. Our cumulative results provide evidence that Wigglesworthia provides a key metabolite (folate) that is “hijacked” by trypanosomes to enhance their infectivity, thus indirectly impacting tsetse species vector competency. Parasite dependence on symbiont-derived micronutrients, which likely also occurs in other arthropod vectors, represents a relationship that may be exploited to reduce disease transmission.
Collapse
|
13
|
Odeniran PO, Macleod ET, Ademola IO, Welburn SC. Endosymbionts interaction with trypanosomes in Palpalis group of Glossina captured in southwest Nigeria. Parasitol Int 2019; 70:64-69. [DOI: 10.1016/j.parint.2019.01.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 01/23/2019] [Accepted: 01/31/2019] [Indexed: 12/31/2022]
|
14
|
Demirbas-Uzel G, Parker AG, Vreysen MJB, Mach RL, Bouyer J, Takac P, Abd-Alla AMM. Impact of Glossina pallidipes salivary gland hypertrophy virus (GpSGHV) on a heterologous tsetse fly host, Glossina fuscipes fuscipes. BMC Microbiol 2018; 18:161. [PMID: 30470172 PMCID: PMC6251146 DOI: 10.1186/s12866-018-1276-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Tsetse flies (Diptera: Glossinidae) are the vectors of African trypanosomosis, the causal agent of sleeping sickness in humans and nagana in animals. Glossina fuscipes fuscipes is one of the most important tsetse vectors of sleeping sickness, particularly in Central Africa. Due to the development of resistance of the trypanosomes to the commonly used trypanocidal drugs and the lack of effective vaccines, vector control approaches remain the most effective strategies for sustainable management of those diseases. The Sterile Insect Technique (SIT) is an effective, environment-friendly method for the management of tsetse flies in the context of area-wide integrated pest management programs (AW-IPM). This technique relies on the mass-production of the target insect, its sterilization with ionizing radiation and the release of sterile males in the target area where they will mate with wild females and induce sterility in the native population. It has been shown that Glossina pallidipes salivary gland hypertrophy virus (GpSGHV) infection causes a decrease in fecundity and fertility hampering the maintenance of colonies of the tsetse fly G. pallidipes. This virus has also been detected in different species of tsetse files. In this study, we evaluated the impact of GpSGHV on the performance of a colony of the heterologous host G. f. fuscipes, including the flies' productivity, mortality, survival, flight propensity and mating ability and insemination rates. RESULTS Even though GpSGHV infection did not induce SGH symptoms, it significantly reduced all examined parameters, except adult flight propensity and insemination rate. CONCLUSION These results emphasize the important role of GpSGHV management strategy in the maintenance of G. f. fuscipes colonies and the urgent need to implement measures to avoid virus infection, to ensure the optimal mass production of this tsetse species for use in AW-IPM programs with an SIT component.
Collapse
Affiliation(s)
- Güler Demirbas-Uzel
- Insect Pest Control Laboratory, Joint FAO/IAEA Division of Nuclear Techniques in Food & Agriculture, International Atomic Energy Agency, Vienna International Centre, P.O. Box 100, 1400, Vienna, Austria.,Institute of Chemical, Environmental and Biological Engineering, Research Area Biochemical Technology, Vienna University of Technology, Gumpendorfer Straße 1a, 1060, Vienna, Austria
| | - Andrew G Parker
- Insect Pest Control Laboratory, Joint FAO/IAEA Division of Nuclear Techniques in Food & Agriculture, International Atomic Energy Agency, Vienna International Centre, P.O. Box 100, 1400, Vienna, Austria
| | - Marc J B Vreysen
- Insect Pest Control Laboratory, Joint FAO/IAEA Division of Nuclear Techniques in Food & Agriculture, International Atomic Energy Agency, Vienna International Centre, P.O. Box 100, 1400, Vienna, Austria
| | - Robert L Mach
- Institute of Chemical, Environmental and Biological Engineering, Research Area Biochemical Technology, Vienna University of Technology, Gumpendorfer Straße 1a, 1060, Vienna, Austria
| | - Jeremy Bouyer
- Insect Pest Control Laboratory, Joint FAO/IAEA Division of Nuclear Techniques in Food & Agriculture, International Atomic Energy Agency, Vienna International Centre, P.O. Box 100, 1400, Vienna, Austria
| | - Peter Takac
- Section of Molecular and Applied Zoology, Institute of Zoology, Slovak Academy of Sciences, 845 06, Bratislava, SR, Slovakia.,Scientica, Ltd., Hybešova 33, 831 06, Bratislava, Slovakia
| | - Adly M M Abd-Alla
- Insect Pest Control Laboratory, Joint FAO/IAEA Division of Nuclear Techniques in Food & Agriculture, International Atomic Energy Agency, Vienna International Centre, P.O. Box 100, 1400, Vienna, Austria.
| |
Collapse
|
15
|
Griffith BC, Weiss BL, Aksoy E, Mireji PO, Auma JE, Wamwiri FN, Echodu R, Murilla G, Aksoy S. Analysis of the gut-specific microbiome from field-captured tsetse flies, and its potential relevance to host trypanosome vector competence. BMC Microbiol 2018; 18:146. [PMID: 30470178 PMCID: PMC6251097 DOI: 10.1186/s12866-018-1284-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Background The tsetse fly (Glossina sp.) midgut is colonized by maternally transmitted and environmentally acquired bacteria. Additionally, the midgut serves as a niche in which pathogenic African trypanosomes reside within infected flies. Tsetse’s bacterial microbiota impacts many aspects of the fly’s physiology. However, little is known about the structure of tsetse’s midgut-associated bacterial communities as they relate to geographically distinct fly habitats in east Africa and their contributions to parasite infection outcomes. We utilized culture dependent and independent methods to characterize the taxonomic structure and density of bacterial communities that reside within the midgut of tsetse flies collected at geographically distinct locations in Kenya and Uganda. Results Using culture dependent methods, we isolated 34 strains of bacteria from four different tsetse species (G. pallidipes, G. brevipalpis, G. fuscipes and G. fuscipleuris) captured at three distinct locations in Kenya. To increase the depth of this study, we deep sequenced midguts from individual uninfected and trypanosome infected G. pallidipes captured at two distinct locations in Kenya and one in Uganda. We found that tsetse’s obligate endosymbiont, Wigglesworthia, was the most abundant bacterium present in the midgut of G. pallidipes, and the density of this bacterium remained largely consistent regardless of whether or not its tsetse host was infected with trypanosomes. These fly populations also housed the commensal symbiont Sodalis, which was found at significantly higher densities in trypanosome infected compared to uninfected flies. Finally, midguts of field-captured G. pallidipes were colonized with distinct, low density communities of environmentally acquired microbes that differed in taxonomic structure depending on parasite infection status and the geographic location from which the flies were collected. Conclusions The results of this study will enhance our understanding of the tripartite relationship between tsetse, its microbiota and trypanosome vector competence. This information may be useful for developing novel disease control strategies or enhancing the efficacy of those already in use. Electronic supplementary material The online version of this article (10.1186/s12866-018-1284-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bridget C Griffith
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA.,Present Address: Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Brian L Weiss
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA.
| | - Emre Aksoy
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA.,Present Address: Department of Entomology, University of California Riverside, Riverside, CA, USA
| | - Paul O Mireji
- Biotechnology Research Institute, Kenya Agricultural and Livestock Research Organization, Kikuyu, Kenya
| | - Joana E Auma
- Biotechnology Research Institute, Kenya Agricultural and Livestock Research Organization, Kikuyu, Kenya
| | - Florence N Wamwiri
- Biotechnology Research Institute, Kenya Agricultural and Livestock Research Organization, Kikuyu, Kenya
| | - Richard Echodu
- Department of Biology, Faculty of Science, Gulu University, Gulu, Uganda
| | - Grace Murilla
- Biotechnology Research Institute, Kenya Agricultural and Livestock Research Organization, Kikuyu, Kenya
| | - Serap Aksoy
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA.
| |
Collapse
|
16
|
Geiger A, Malele I, Abd-Alla AM, Njiokou F. Blood feeding tsetse flies as hosts and vectors of mammals-pre-adapted African Trypanosoma: current and expected research directions. BMC Microbiol 2018; 18:162. [PMID: 30470183 PMCID: PMC6251083 DOI: 10.1186/s12866-018-1281-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Research on the zoo-anthropophilic blood feeding tsetse flies' biology conducted, by different teams, in laboratory settings and at the level of the ecosystems- where also co-perpetuate African Trypanosoma- has allowed to unveil and characterize key features of tsetse flies' bacterial symbionts on which rely both (a) the perpetuation of the tsetse fly populations and (b) the completion of the developmental program of the African Trypanosoma. Transcriptomic analyses have already provided much information on tsetse fly genes as well as on genes of the fly symbiotic partners Sodalis glossinidius and Wigglesworthia, which account for the successful onset or not of the African Trypanosoma developmental program. In parallel, identification of the non- symbiotic bacterial communities hosted in the tsetse fly gut has recently been initiated: are briefly introduced those bacteria genera and species common to tsetse flies collected from distinct ecosystems, that could be further studied as potential biologicals preventing the onset of the African Trypanosoma developmental program. Finally, future work will need to concentrate on how to render tsetse flies refractory, and the best means to disseminate them in the field in order to establish an overall refractory fly population.
Collapse
Affiliation(s)
- Anne Geiger
- INTERTRYP, Institut de Recherche pour le Développement, University of Montpellier, Montpellier, France
| | - Imna Malele
- Vector and Vector Borne Diseases Institute, Majani Mapana, Off Korogwe Road, Box, 1026 Tanga, Tanzania
| | - Adly M Abd-Alla
- Insect Pest Control Laboratory, Joint FAO/IAEA Division of Nuclear Techniques in Food and Agriculture, Vienna, Austria
| | - Flobert Njiokou
- Faculty of Science, University of Yaoundé I, P.O. Box 812, Yaoundé, Cameroon
| |
Collapse
|
17
|
Qiu Y, Milanes JE, Jones JA, Noorai RE, Shankar V, Morris JC. Glucose Signaling Is Important for Nutrient Adaptation during Differentiation of Pleomorphic African Trypanosomes. mSphere 2018; 3:e00366-18. [PMID: 30381351 PMCID: PMC6211221 DOI: 10.1128/msphere.00366-18] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 09/05/2018] [Indexed: 11/30/2022] Open
Abstract
The African trypanosome has evolved mechanisms to adapt to changes in nutrient availability that occur during its life cycle. During transition from mammalian blood to insect vector gut, parasites experience a rapid reduction in environmental glucose. Here we describe how pleomorphic parasites respond to glucose depletion with a focus on parasite changes in energy metabolism and growth. Long slender bloodstream form parasites were rapidly killed as glucose concentrations fell, while short stumpy bloodstream form parasites persisted to differentiate into the insect-stage procyclic form parasite. The rate of differentiation was lower than that triggered by other cues but reached physiological rates when combined with cold shock. Both differentiation and growth of resulting procyclic form parasites were inhibited by glucose and nonmetabolizable glucose analogs, and these parasites were found to have upregulated amino acid metabolic pathway component gene expression. In summary, glucose transitions from the primary metabolite of the blood-stage infection to a negative regulator of cell development and growth in the insect vector, suggesting that the hexose is not only a key metabolic agent but also an important signaling molecule.IMPORTANCE As the African trypanosome Trypanosoma brucei completes its life cycle, it encounters many different environments. Adaptation to these environments includes modulation of metabolic pathways to parallel the availability of nutrients. Here, we describe how the blood-dwelling life cycle stages of the African trypanosome, which consume glucose to meet their nutritional needs, respond differently to culture in the near absence of glucose. The proliferative long slender parasites rapidly die, while the nondividing short stumpy parasite remains viable and undergoes differentiation to the next life cycle stage, the procyclic form parasite. Interestingly, a sugar analog that cannot be used as an energy source inhibited the process. Furthermore, the growth of procyclic form parasite that resulted from the event was inhibited by glucose, a behavior that is similar to that of parasites isolated from tsetse flies. Our findings suggest that glucose sensing serves as an important modulator of nutrient adaptation in the parasite.
Collapse
Affiliation(s)
- Yijian Qiu
- Department of Genetics and Biochemistry, Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, South Carolina, USA
| | - Jillian E Milanes
- Department of Genetics and Biochemistry, Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, South Carolina, USA
| | - Jessica A Jones
- Department of Genetics and Biochemistry, Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, South Carolina, USA
| | - Rooksana E Noorai
- Clemson University Genomics & Computational Biology Laboratory, Clemson University, Clemson, South Carolina, USA
| | - Vijay Shankar
- Clemson University Genomics & Computational Biology Laboratory, Clemson University, Clemson, South Carolina, USA
| | - James C Morris
- Department of Genetics and Biochemistry, Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, South Carolina, USA
| |
Collapse
|
18
|
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
|
19
|
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
|
20
|
Schuster S, Krüger T, Subota I, Thusek S, Rotureau B, Beilhack A, Engstler M. Developmental adaptations of trypanosome motility to the tsetse fly host environments unravel a multifaceted in vivo microswimmer system. eLife 2017; 6. [PMID: 28807106 PMCID: PMC5570225 DOI: 10.7554/elife.27656] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 07/25/2017] [Indexed: 12/20/2022] Open
Abstract
The highly motile and versatile protozoan pathogen Trypanosoma brucei undergoes a complex life cycle in the tsetse fly. Here we introduce the host insect as an expedient model environment for microswimmer research, as it allows examination of microbial motion within a diversified, secluded and yet microscopically tractable space. During their week-long journey through the different microenvironments of the fly´s interior organs, the incessantly swimming trypanosomes cross various barriers and confined surroundings, with concurrently occurring major changes of parasite cell architecture. Multicolour light sheet fluorescence microscopy provided information about tsetse tissue topology with unprecedented resolution and allowed the first 3D analysis of the infection process. High-speed fluorescence microscopy illuminated the versatile behaviour of trypanosome developmental stages, ranging from solitary motion and near-wall swimming to collective motility in synchronised swarms and in confinement. We correlate the microenvironments and trypanosome morphologies to high-speed motility data, which paves the way for cross-disciplinary microswimmer research in a naturally evolved environment.
Collapse
Affiliation(s)
- Sarah Schuster
- Department of Cell and Developmental Biology, Biocentre, University of Würzburg, Würzburg, Germany
| | - Timothy Krüger
- Department of Cell and Developmental Biology, Biocentre, University of Würzburg, Würzburg, Germany
| | - Ines Subota
- Department of Cell and Developmental Biology, Biocentre, University of Würzburg, Würzburg, Germany
| | - Sina Thusek
- Department of Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Brice Rotureau
- Trypanosome Transmission Group, Trypanosome Cell Biology Unit, Department of Parasites and Insect Vectors, Institut Pasteur and INSERM U1201, Paris, France
| | - Andreas Beilhack
- Department of Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Markus Engstler
- Department of Cell and Developmental Biology, Biocentre, University of Würzburg, Würzburg, Germany
| |
Collapse
|
21
|
Tsagmo Ngoune JM, Njiokou F, Loriod B, Kame-Ngasse G, Fernandez-Nunez N, Rioualen C, van Helden J, Geiger A. Transcriptional Profiling of Midguts Prepared from Trypanosoma/T. congolense-Positive Glossina palpalis palpalis Collected from Two Distinct Cameroonian Foci: Coordinated Signatures of the Midguts' Remodeling As T. congolense-Supportive Niches. Front Immunol 2017; 8:876. [PMID: 28804485 PMCID: PMC5532377 DOI: 10.3389/fimmu.2017.00876] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 07/10/2017] [Indexed: 12/11/2022] Open
Abstract
Our previous transcriptomic analysis of Glossina palpalis gambiensis experimentally infected or not with Trypanosoma brucei gambiense aimed to detect differentially expressed genes (DEGs) associated with infection. Specifically, we selected candidate genes governing tsetse fly vector competence that could be used in the context of an anti-vector strategy, to control human and/or animal trypanosomiasis. The present study aimed to verify whether gene expression in field tsetse flies (G. p. palpalis) is modified in response to natural infection by trypanosomes (T. congolense), as reported when insectary-raised flies (G. p. gambiensis) are experimentally infected with T. b. gambiense. This was achieved using the RNA-seq approach, which identified 524 DEGs in infected vs. non-infected tsetse flies, including 285 downregulated genes and 239 upregulated genes (identified using DESeq2). Several of these genes were highly differentially expressed, with log2 fold change values in the vicinity of either +40 or −40. Downregulated genes were primarily involved in transcription/translation processes, whereas encoded upregulated genes governed amino acid and nucleotide biosynthesis pathways. The BioCyc metabolic pathways associated with infection also revealed that downregulated genes were mainly involved in fly immunity processes. Importantly, our study demonstrates that data on the molecular cross-talk between the host and the parasite (as well as the always present fly microbiome) recorded from an experimental biological model has a counterpart in field flies, which in turn validates the use of experimental host/parasite couples.
Collapse
Affiliation(s)
- Jean M Tsagmo Ngoune
- Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon.,UMR 177, IRD-CIRAD, CIRAD TA A-17/G, Campus International de Baillarguet, Montpellier, France
| | - Flobert Njiokou
- Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon
| | - Béatrice Loriod
- Aix-Marseille University, INSERM, TAGC, Technological Advances for Genomics and Clinics, UMR S 1090, Marseille, France
| | | | - Nicolas Fernandez-Nunez
- Aix-Marseille University, INSERM, TAGC, Technological Advances for Genomics and Clinics, UMR S 1090, Marseille, France
| | - Claire Rioualen
- Aix-Marseille University, INSERM, TAGC, Technological Advances for Genomics and Clinics, UMR S 1090, Marseille, France
| | - Jacques van Helden
- Aix-Marseille University, INSERM, TAGC, Technological Advances for Genomics and Clinics, UMR S 1090, Marseille, France
| | - Anne Geiger
- UMR 177, IRD-CIRAD, CIRAD TA A-17/G, Campus International de Baillarguet, Montpellier, France
| |
Collapse
|
22
|
Garcia HA, Rodrigues CMF, Rodrigues AC, Pereira DL, Pereira CL, Camargo EP, Hamilton PB, Teixeira MMG. Remarkable richness of trypanosomes in tsetse flies (Glossina morsitans morsitans and Glossina pallidipes) from the Gorongosa National Park and Niassa National Reserve of Mozambique revealed by fluorescent fragment length barcoding (FFLB). INFECTION GENETICS AND EVOLUTION 2017; 63:370-379. [PMID: 28688979 DOI: 10.1016/j.meegid.2017.07.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 07/03/2017] [Accepted: 07/04/2017] [Indexed: 11/25/2022]
Abstract
Trypanosomes of African wild ungulates transmitted by tsetse flies can cause human and livestock diseases. However, trypanosome diversity in wild tsetse flies remains greatly underestimated. We employed FFLB (fluorescent fragment length barcoding) for surveys of trypanosomes in tsetse flies (3086) from the Gorongosa National Park (GNP) and Niassa National Reserve (NNR) in Mozambique (MZ), identified as Glossina morsitans morsitans (GNP/NNR=77.6%/90.5%) and Glossina pallidipes (22.4%/9.5%). Trypanosomes were microscopically detected in 8.3% of tsetse guts. FFLB of gut samples revealed (GNP/NNR): Trypanosoma congolense of Savannah (27%/63%), Kilifi (16.7%/29.7%) and Forest (1.0%/0.3%) genetic groups; T. simiae Tsavo (36.5%/6.1%); T. simiae (22.2%/17.7%); T. godfreyi (18.2%/7.0%); subgenus Trypanozoon (20.2%/25.7%); T. vivax/T. vivax-like (1.5%/5.2%); T. suis/T. suis-like (9.4%/11.9%). Tsetse proboscises exhibited similar species composition, but most prevalent species were (GNP/NNR): T. simiae (21.9%/28%), T. b. brucei (19.2%/31.7%), and T. vivax/T. vivax-like (19.2%/28.6%). Flies harboring mixtures of trypanosomes were common (~ 64%), and combinations of more than four trypanosomes were especially abundant in the pristine NNR. The non-pathogenic T. theileri was found in 2.5% while FFLB profiles of unknown species were detected in 19% of flies examined. This is the first report on molecular diversity of tsetse flies and their trypanosomes in MZ; all trypanosomes pathogenic for ungulates were detected, but no human pathogens were detected. Overall, two species of tsetse flies harbor 12 species/genotypes of trypanosomes. This notable species richness was likely uncovered because flies were captured in wildlife reserves and surveyed using the method of FFLB able to identify, with high sensitivity and accuracy, known and novel trypanosomes. Our findings importantly improve the knowledge on trypanosome diversity in tsetse flies, revealed the greatest species richness so far reported in tsetse fly of any African country, and indicate the existence of a hidden trypanosome diversity to be discovered in African wildlife protected areas.
Collapse
Affiliation(s)
- Herakles A Garcia
- Department of Parasitology, Institute of Biomedical Sciences, University of Sao Paulo, Brazil
| | - Carla M F Rodrigues
- Department of Parasitology, Institute of Biomedical Sciences, University of Sao Paulo, Brazil
| | - Adriana C Rodrigues
- Department of Parasitology, Institute of Biomedical Sciences, University of Sao Paulo, Brazil
| | | | - Carlos L Pereira
- Ministry of Tourism of Mozambique, Wildlife Conservation Society, Mozambique
| | - Erney P Camargo
- Department of Parasitology, Institute of Biomedical Sciences, University of Sao Paulo, Brazil
| | - P B Hamilton
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, UK
| | - Marta M G Teixeira
- Department of Parasitology, Institute of Biomedical Sciences, University of Sao Paulo, Brazil.
| |
Collapse
|
23
|
Aksoy S, Buscher P, Lehane M, Solano P, Van Den Abbeele J. Human African trypanosomiasis control: Achievements and challenges. PLoS Negl Trop Dis 2017; 11:e0005454. [PMID: 28426685 PMCID: PMC5398477 DOI: 10.1371/journal.pntd.0005454] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Sleeping sickness, also known as human African trypanosomiasis (HAT), is a neglected disease that impacts 70 million people living in 1.55 million km2 in sub-Saharan Africa. Since the beginning of the 20th century, there have been multiple HAT epidemics in sub-Saharan Africa, with the most recent epidemic in the 1990s resulting in about half a million HAT cases reported between 1990 and 2015. Here we review the status of HAT disease at the current time and the toolbox available for its control. We also highlight future opportunities under development towards novel or improved interventions.
Collapse
Affiliation(s)
- Serap Aksoy
- Department of Epidemiology of Microbial Diseases, School of Public Health, Yale University, New Haven, Connecticut, United States of America
- * E-mail:
| | - Phillipe Buscher
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Mike Lehane
- Department of Vector Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Philippe Solano
- Institut de Recherche pour le Développement (IRD), Montpellier, France
| | - Jan Van Den Abbeele
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| |
Collapse
|
24
|
Patino LH, Ramírez JD. RNA-seq in kinetoplastids: A powerful tool for the understanding of the biology and host-pathogen interactions. INFECTION GENETICS AND EVOLUTION 2017; 49:273-282. [PMID: 28179142 DOI: 10.1016/j.meegid.2017.02.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 02/01/2017] [Accepted: 02/02/2017] [Indexed: 12/31/2022]
Abstract
The kinetoplastids include a large number of parasites responsible for serious diseases in humans and animals (Leishmania and Trypanosoma brucei) considered endemic in several regions of the world. These parasites are characterized by digenetic life cycles that undergo morphological and genetic changes that allow them to adapt to different microenvironments on their vertebrates and invertebrates hosts. Recent advances in ´omics´ technology, specifically transcriptomics have allowed to reveal aspects associated with such molecular changes. So far, different techniques have been used to evaluate the gene expression profile during the various stages of the life cycle of these parasites and during the host-parasite interactions. However, some of them have serious drawbacks that limit the precise study and full understanding of their transcriptomes. Therefore, recently has been implemented the latest technology (RNA-seq), which overcomes the drawbacks of traditional methods. In this review, studies that so far have used RNA-seq are presented and allowed to expand our knowledge regarding the biology of these parasites and their interactions with their hosts.
Collapse
Affiliation(s)
- Luz Helena Patino
- Grupo de Investigaciones Microbiológicas-UR (GIMUR), Programa de Biología, Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Carrera 24# 63C-69, Bogotá, Colombia
| | - Juan David Ramírez
- Grupo de Investigaciones Microbiológicas-UR (GIMUR), Programa de Biología, Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Carrera 24# 63C-69, Bogotá, Colombia.
| |
Collapse
|
25
|
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
|
26
|
Zhao X, Silva TLAE, Cronin L, Savage AF, O’Neill M, Nerima B, Okedi LM, Aksoy S. Immunogenicity and Serological Cross-Reactivity of Saliva Proteins among Different Tsetse Species. PLoS Negl Trop Dis 2015; 9:e0004038. [PMID: 26313460 PMCID: PMC4551805 DOI: 10.1371/journal.pntd.0004038] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 08/05/2015] [Indexed: 12/17/2022] Open
Abstract
Tsetse are vectors of pathogenic trypanosomes, agents of human and animal trypanosomiasis in Africa. Components of tsetse saliva (sialome) are introduced into the mammalian host bite site during the blood feeding process and are important for tsetse’s ability to feed efficiently, but can also influence disease transmission and serve as biomarkers for host exposure. We compared the sialome components from four tsetse species in two subgenera: subgenus Morsitans: Glossina morsitans morsitans (Gmm) and Glossina pallidipes (Gpd), and subgenus Palpalis: Glossina palpalis gambiensis (Gpg) and Glossina fuscipes fuscipes (Gff), and evaluated their immunogenicity and serological cross reactivity by an immunoblot approach utilizing antibodies from experimental mice challenged with uninfected flies. The protein and immune profiles of sialome components varied with fly species in the same subgenus displaying greater similarity and cross reactivity. Sera obtained from cattle from disease endemic areas of Africa displayed an immunogenicity profile reflective of tsetse species distribution. We analyzed the sialome fractions of Gmm by LC-MS/MS, and identified TAg5, Tsal1/Tsal2, and Sgp3 as major immunogenic proteins, and the 5'-nucleotidase family as well as four members of the Adenosine Deaminase Growth Factor (ADGF) family as the major non-immunogenic proteins. Within the ADGF family, we identified four closely related proteins (TSGF-1, TSGF-2, ADGF-3 and ADGF-4), all of which are expressed in tsetse salivary glands. We describe the tsetse species-specific expression profiles and genomic localization of these proteins. Using a passive-immunity approach, we evaluated the effects of rec-TSGF (TSGF-1 and TSGF-2) polyclonal antibodies on tsetse fitness parameters. Limited exposure of tsetse to mice with circulating anti-TSGF antibodies resulted in a slight detriment to their blood feeding ability as reflected by compromised digestion, lower weight gain and less total lipid reserves although these results were not statistically significant. Long-term exposure studies of tsetse flies to antibodies corresponding to the ADGF family of proteins are warranted to evaluate the role of this conserved family in fly biology. Insect saliva contains many proteins that are injected into the mammalian host during the blood feeding process. Saliva proteins enhance the blood feeding ability of insects, but they can also induce mammalian immune responses that inhibit successful feeding, or modulate the bite site to benefit pathogen transmission. Here we studied saliva from four different tsetse species that belong to two distant species groups. We show that the saliva protein profiles of different species groups vary. Experimental mice subjected to fly bites display varying immunological responses against the abundant saliva proteins and the antigenicity of the shared saliva proteins in different tsetse species differs. We show that one member of the ADGF family with adenosine deaminase motifs, TSGF-2, is non-immunogenic in Glossina morsitans in mice, while the same protein from Glossina fuscipes is highly immunogenic. Such species-specific immune responses could be exploited as biomarkers of host exposures in the field. We also show that short-term exposure of G. morsitans to mice passively immunized by anti-TSGF antibodies leads to slight but not statistically significant negative fitness effects. Thus, future investigations with non-antigenic saliva proteins are warranted as they can lead to novel mammalian vaccine targets to reduce tsetse populations in the field.
Collapse
Affiliation(s)
- Xin Zhao
- Yale School of Public Health, Department of Epidemiology of Microbial Diseases, New Haven, Connecticut, United States of America
| | - Thiago Luiz Alves e Silva
- Yale School of Public Health, Department of Epidemiology of Microbial Diseases, New Haven, Connecticut, United States of America
| | - Laura Cronin
- Yale School of Public Health, Department of Epidemiology of Microbial Diseases, New Haven, Connecticut, United States of America
| | - Amy F. Savage
- Yale School of Public Health, Department of Epidemiology of Microbial Diseases, New Haven, Connecticut, United States of America
| | - Michelle O’Neill
- 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
- * E-mail:
| |
Collapse
|
27
|
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
|
28
|
Alizadehrad D, Krüger T, Engstler M, Stark H. Simulating the complex cell design of Trypanosoma brucei and its motility. PLoS Comput Biol 2015; 11:e1003967. [PMID: 25569823 PMCID: PMC4288712 DOI: 10.1371/journal.pcbi.1003967] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 10/06/2014] [Indexed: 11/19/2022] Open
Abstract
The flagellate Trypanosoma brucei, which causes the sleeping sickness when infecting a mammalian host, goes through an intricate life cycle. It has a rather complex propulsion mechanism and swims in diverse microenvironments. These continuously exert selective pressure, to which the trypanosome adjusts with its architecture and behavior. As a result, the trypanosome assumes a diversity of complex morphotypes during its life cycle. However, although cell biology has detailed form and function of most of them, experimental data on the dynamic behavior and development of most morphotypes is lacking. Here we show that simulation science can predict intermediate cell designs by conducting specific and controlled modifications of an accurate, nature-inspired cell model, which we developed using information from live cell analyses. The cell models account for several important characteristics of the real trypanosomal morphotypes, such as the geometry and elastic properties of the cell body, and their swimming mechanism using an eukaryotic flagellum. We introduce an elastic network model for the cell body, including bending rigidity and simulate swimming in a fluid environment, using the mesoscale simulation technique called multi-particle collision dynamics. The in silico trypanosome of the bloodstream form displays the characteristic in vivo rotational and translational motility pattern that is crucial for survival and virulence in the vertebrate host. Moreover, our model accurately simulates the trypanosome's tumbling and backward motion. We show that the distinctive course of the attached flagellum around the cell body is one important aspect to produce the observed swimming behavior in a viscous fluid, and also required to reach the maximal swimming velocity. Changing details of the flagellar attachment generates less efficient swimmers. We also simulate different morphotypes that occur during the parasite's development in the tsetse fly, and predict a flagellar course we have not been able to measure in experiments so far. Typanosoma brucei is a uni-cellular parasite that causes the sleeping sickness, a deadly disease for humans that also occurs in livestock. Injected into the mammalian host by the tsetse fly, the trypanosome travels through the blood stream, where it proliferates, and ultimately can be taken up again by a fly during a bloodmeal. In the tsetse fly, it continues its development with several morphological changes to the cell body plan. During its life cycle, the trypanosome meets different microenvironments, such as the mammalian's bloodstream and the tsetse fly's midgut, proventriculus, foregut, and salivary gland. The cell body of the trypanosome has the shape of a spindle along which an eukaryotic flagellum is attached. We have developed an accurate, in silico model trypanosome using information from live cell analyses. Performing computer simulations, we are able to reproduce all motility patterns of the blood-stream form in typical cell culture medium. Modifying the cell design, we show that the helical course of the flagellar attachment optimizes the trypanosome's swimming speed. We also design trypanosomal morphotypes that occur in the tsetse fly. Simulation science thereby provides an investigative tool to systematically explore the morphologcial diversity during the trypanosome's life cycle even beyond experimental capabilities.
Collapse
Affiliation(s)
- Davod Alizadehrad
- Institute of Theoretical Physics, Technische Universität Berlin, Berlin, Germany
- * E-mail:
| | - Timothy Krüger
- Department of Cell and Developmental Biology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Markus Engstler
- Department of Cell and Developmental Biology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Holger Stark
- Institute of Theoretical Physics, Technische Universität Berlin, Berlin, Germany
| |
Collapse
|
29
|
Geiger A, Ponton F, Simo G. Adult blood-feeding tsetse flies, trypanosomes, microbiota and the fluctuating environment in sub-Saharan Africa. ISME JOURNAL 2014; 9:1496-507. [PMID: 25500509 DOI: 10.1038/ismej.2014.236] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2014] [Revised: 11/03/2014] [Accepted: 11/06/2014] [Indexed: 01/01/2023]
Abstract
The tsetse fly vector transmits the protozoan Trypanosoma brucei, responsible for Human African Trypanosomiasis, one of the most neglected tropical diseases. Despite a recent decline in new cases, it is still crucial to develop alternative strategies to combat this disease. Here, we review the literature on the factors that influence trypanosome transmission from the fly vector to its vertebrate host (particularly humans). These factors include climate change effects to pathogen and vector development (in particular climate warming), as well as the distribution of host reservoirs. Finally, we present reports on the relationships between insect vector nutrition, immune function, microbiota and infection, to demonstrate how continuing research on the evolving ecology of these complex systems will help improve control strategies. In the future, such studies will be of increasing importance to understand how vector-borne diseases are spread in a changing world.
Collapse
Affiliation(s)
- Anne Geiger
- UMR 177, IRD-CIRAD, CIRAD TA A-17/G, Campus International de Baillarguet, Montpellier Cedex 5, France
| | - Fleur Ponton
- 1] School of Biological Sciences, The University of Sydney, Sydney, New South Wales, Australia [2] The Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Gustave Simo
- Molecular Parasitology and Entomology Unit, Department of Biochemistry, Faculty of Science, University of Dschang, Dschang, Cameroon
| |
Collapse
|
30
|
Hamidou Soumana I, Tchicaya B, Chuchana P, Geiger A. Midgut expression of immune-related genes in Glossina palpalis gambiensis challenged with Trypanosoma brucei gambiense. Front Microbiol 2014; 5:609. [PMID: 25426112 PMCID: PMC4226161 DOI: 10.3389/fmicb.2014.00609] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Accepted: 10/26/2014] [Indexed: 11/13/2022] Open
Abstract
Tsetse flies from the subspecies Glossina morsitans morsitans and Glossina palpalis gambiensis, respectively, transmit Trypanosoma brucei rhodesiense and Trypanosoma brucei gambiense. The former causes the acute form of sleeping sickness, and the latter provokes the chronic form. Although several articles have reported G. m. morsitans gene expression following trypanosome infection, no comparable investigation has been performed for G. p. gambiensis. This report presents results on the differential expression of immune-related genes in G. p. gambiensis challenged with T. b. gambiense. The aim was to characterize transcriptomic events occurring in the tsetse gut during the parasite establishment step, which is the crucial first step in the parasite development cycle within its vector. The selected genes were chosen from those previously shown to be highly expressed in G. m. morsitans, to allow further comparison of gene expression in both Glossina species. Using quantitative PCR, genes were amplified from the dissected midguts of trypanosome-stimulated, infected, non-infected, and self-cleared flies at three sampling timepoints (3, 10, and 20 days) after a bloodmeal. At the 3-day sampling point, transferrin transcripts were significantly up-regulated in trypanosome-challenged flies versus flies fed on non-infected mice. In self-cleared flies, serpin-2 and thioredoxin peroxidase-3 transcripts were significantly up-regulated 10 days after trypanosome challenge, whereas nitric oxide synthase and chitin-binding protein transcripts were up-regulated after 20 days. Although the expression levels of the other genes were highly variable, the expression of immune-related genes in G. p. gambiensis appears to be a time-dependent process. The possible biological significance of these findings is discussed, and the results are compared with previous reports for G. m. morsitans.
Collapse
Affiliation(s)
| | | | - Paul Chuchana
- Inserm, U844, Hôpital Saint-Eloi Montpellier, France
| | | |
Collapse
|
31
|
Balaña-Fouce R, Alvarez-Velilla R, Fernández-Prada C, García-Estrada C, Reguera RM. Trypanosomatids topoisomerase re-visited. New structural findings and role in drug discovery. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2014; 4:326-37. [PMID: 25516844 PMCID: PMC4266802 DOI: 10.1016/j.ijpddr.2014.07.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
There is an urgent need of new treatments against trypanosomatids-borne diseases. DNA topoisomerases are pointed as potential drug targets against unicellular parasites. Trypanosomatids have a full set of DNA topoisomerases in both nucleus and kinetoplast. TopII and TopIII are located in the kinetoplast and fully involved in kDNA replication. Tritryps TopIB differ in structure from mammalian’s pointing to an attractive target.
The Trypanosomatidae family, composed of unicellular parasites, causes severe vector-borne diseases that afflict human populations worldwide. Chagas disease, sleeping sickness, as well as different sorts of leishmaniases are amongst the most important infectious diseases produced by Trypanosoma cruzi, Trypanosoma brucei and Leishmania spp., respectively. All these infections are closely related to weak health care services in low-income populations of less developed and least economically developed countries. Search for new therapeutic targets in order to hit these pathogens is of paramount priority, as no effective vaccine is currently in use against any of these parasites. Furthermore, present-day chemotherapy comprises old-fashioned drugs full of important side effects. Besides, they are prone to produce tolerance and resistance as a consequence of their continuous use for decades. DNA topoisomerases (Top) are ubiquitous enzymes responsible for solving the torsional tensions caused during replication and transcription processes, as well as in maintaining genomic stability during DNA recombination. As the inhibition of these enzymes produces cell arrest and triggers cell death, Top inhibitors are among the most effective and most widely used drugs in both cancer and antibacterial therapies. Top relaxation and decatenation activities, which are based on a common nicking–closing cycle involving one or both DNA strands, have been pointed as a promising drug target. Specific inhibitors that bind to the interface of DNA-Top complexes can stabilize Top-mediated transient DNA breaks. In addition, important structural differences have been found between Tops from the Trypanosomatidae family members and Tops from the host. Such dissimilarities make these proteins very interesting for drug design and molecular intervention. The present review is a critical update of the last findings regarding trypanosomatid’s Tops, their new structural features, their involvement both in the physiology and virulence of these parasites, as well as their use as promising targets for drug discovery.
Collapse
Affiliation(s)
- Rafael Balaña-Fouce
- Departamento de Ciencias Biomédicas, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Raquel Alvarez-Velilla
- Departamento de Ciencias Biomédicas, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | | | - Carlos García-Estrada
- Departamento de Ciencias Biomédicas, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| | - Rosa M Reguera
- Departamento de Ciencias Biomédicas, Universidad de León, Campus de Vegazana s/n, 24071 León, Spain
| |
Collapse
|
32
|
Abstract
Human African trypanosomiasis (HAT), or sleeping sickness, is caused by Trypanosoma brucei gambiense, which is a chronic form of the disease present in western and central Africa, and by Trypanosoma brucei rhodesiense, which is an acute disease located in eastern and southern Africa. The rhodesiense form is a zoonosis, with the occasional infection of humans, but in the gambiense form, the human being is regarded as the main reservoir that plays a key role in the transmission cycle of the disease. The gambiense form currently assumes that 98% of the cases are declared; the Democratic Republic of the Congo is the most affected country, with more than 75% of the gambiense cases declared. The epidemiology of the disease is mediated by the interaction of the parasite (trypanosome) with the vectors (tsetse flies), as well as with the human and animal hosts within a particular environment. Related to these interactions, the disease is confined in spatially limited areas called “foci”, which are located in Sub-Saharan Africa, mainly in remote rural areas. The risk of contracting HAT is, therefore, determined by the possibility of contact of a human being with an infected tsetse fly. Epidemics of HAT were described at the beginning of the 20th century; intensive activities have been set up to confront the disease, and it was under control in the 1960s, with fewer than 5,000 cases reported in the whole continent. The disease resurged at the end of the 1990s, but renewed efforts from endemic countries, cooperation agencies, and nongovernmental organizations led by the World Health Organization succeeded to raise awareness and resources, while reinforcing national programs, reversing the trend of the cases reported, and bringing the disease under control again. In this context, sustainable elimination of the gambiense HAT, defined as the interruption of the transmission of the disease, was considered as a feasible target for 2030. Since rhodesiense HAT is a zoonosis, where the animal reservoir plays a key role, the interruption of the disease’s transmission is not deemed feasible.
Collapse
Affiliation(s)
- Jose R Franco
- World Health Organization, Control of Neglected Tropical Diseases, Innovative and Intensified Disease Management, Geneva, Switzerland
| | - Pere P Simarro
- World Health Organization, Control of Neglected Tropical Diseases, Innovative and Intensified Disease Management, Geneva, Switzerland
| | - Abdoulaye Diarra
- World Health Organization, Inter Country Support Team for Central Africa, Regional Office for Africa, Libreville, Gabon
| | - Jean G Jannin
- World Health Organization, Control of Neglected Tropical Diseases, Innovative and Intensified Disease Management, Geneva, Switzerland
| |
Collapse
|
33
|
Analysis of multiple tsetse fly populations in Uganda reveals limited diversity and species-specific gut microbiota. Appl Environ Microbiol 2014; 80:4301-12. [PMID: 24814785 DOI: 10.1128/aem.00079-14] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The invertebrate microbiome contributes to multiple aspects of host physiology, including nutrient supplementation and immune maturation processes. We identified and compared gut microbial abundance and diversity in natural tsetse flies from Uganda using five genetically distinct populations of Glossina fuscipes fuscipes and multiple tsetse species (Glossina morsitans morsitans, G. f. fuscipes, and Glossina pallidipes) that occur in sympatry in one location. We used multiple approaches, including deep sequencing of the V4 hypervariable region of the 16S rRNA gene, 16S rRNA gene clone libraries, and bacterium-specific quantitative PCR (qPCR), to investigate the levels and patterns of gut microbial diversity from a total of 151 individuals. Our results show extremely limited diversity in field flies of different tsetse species. The obligate endosymbiont Wigglesworthia dominated all samples (>99%), but we also observed wide prevalence of low-density Sodalis (tsetse's commensal endosymbiont) infections (<0.05%). There were also several individuals (22%) with high Sodalis density, which also carried coinfections with Serratia. Albeit in low density, we noted differences in microbiota composition among the genetically distinct G. f. fuscipes flies and between different sympatric species. Interestingly, Wigglesworthia density varied in different species (10(4) to 10(6) normalized genomes), with G. f. fuscipes having the highest levels. We describe the factors that may be responsible for the reduced diversity of tsetse's gut microbiota compared to those of other insects. Additionally, we discuss the implications of Wigglesworthia and Sodalis density variations as they relate to trypanosome transmission dynamics and vector competence variations associated with different tsetse species.
Collapse
|
34
|
Telleria EL, Benoit JB, Zhao X, Savage AF, Regmi S, e Silva TLA, O'Neill M, Aksoy S. Insights into the trypanosome-host interactions revealed through transcriptomic analysis of parasitized tsetse fly salivary glands. PLoS Negl Trop Dis 2014; 8:e2649. [PMID: 24763140 PMCID: PMC3998935 DOI: 10.1371/journal.pntd.0002649] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 12/03/2013] [Indexed: 12/12/2022] Open
Abstract
The agents of sleeping sickness disease, Trypanosoma brucei complex parasites, are transmitted to mammalian hosts through the bite of an infected tsetse. Information on tsetse-trypanosome interactions in the salivary gland (SG) tissue, and on mammalian infective metacyclic (MC) parasites present in the SG, is sparse. We performed RNA-seq analyses from uninfected and T. b. brucei infected SGs of Glossina morsitans morsitans. Comparison of the SG transcriptomes to a whole body fly transcriptome revealed that only 2.7% of the contigs are differentially expressed during SG infection, and that only 263 contigs (0.6%) are preferentially expressed in the SGs (SG-enriched). The expression of only 37 contigs (0.08%) and 27 SG-enriched contigs (10%) were suppressed in infected SG. These suppressed contigs accounted for over 55% of the SG transcriptome, and included the most abundant putative secreted proteins with anti-hemostatic functions present in saliva. In contrast, expression of putative host proteins associated with immunity, stress, cell division and tissue remodeling were enriched in infected SG suggesting that parasite infections induce host immune and stress response(s) that likely results in tissue renewal. We also performed RNA-seq analysis from mouse blood infected with the same parasite strain, and compared the transcriptome of bloodstream form (BSF) cells with that of parasites obtained from the infected SG. Over 30% of parasite transcripts are differentially regulated between the two stages, and reflect parasite adaptations to varying host nutritional and immune ecology. These differences are associated with the switch from an amino acid based metabolism in the SG to one based on glucose utilization in the blood, and with surface coat modifications that enable parasite survival in the different hosts. This study provides a foundation on the molecular aspects of the trypanosome dialogue with its tsetse and mammalian hosts, necessary for future functional investigations. Tsetse flies transmit the causative agents of African sleeping sickness and nagana in sub-Saharan Africa. The parasites are acquired when tsetse flies feed on an infected host, undergo multiplication in the fly gut and migrate to the salivary glands (SG). The cycle resumes once this infected fly transmits the parasites in conjunction with saliva to another host when feeding. We compared gene expression changes between parasitized and uninfected tsetse SG. We also assessed changes in parasite gene expression in the tsetse SG in relation to those present within vertebrate blood. We found that parasite infections increase expression of host proteins associated with stress and cell division, indicative of extensive cellular damage in SG. We also found that parasite infections reduce expression of the most highly expressed SG-specific secreted proteins, suggesting modification of saliva composition. The parasite transcriptome reveals changes in specific cell surface proteins and in metabolism related to glucose-amino acid utilization in the different host environments. This study provides information for critical understanding of tsetse-trypanosome interactions, and transcriptional changes that likely enable the parasite to persist in the varying environment of its insect and vertebrate hosts.
Collapse
Affiliation(s)
- Erich Loza Telleria
- Yale School of Public Health, Department of Epidemiology of Microbial Diseases, LEPH, New Haven, Connecticut, United States of America
| | - Joshua B. Benoit
- Yale School of Public Health, Department of Epidemiology of Microbial Diseases, LEPH, New Haven, Connecticut, United States of America
| | - Xin Zhao
- Yale School of Public Health, Department of Epidemiology of Microbial Diseases, LEPH, New Haven, Connecticut, United States of America
| | - Amy F. Savage
- Yale School of Public Health, Department of Epidemiology of Microbial Diseases, LEPH, New Haven, Connecticut, United States of America
| | - Sandesh Regmi
- Yale School of Public Health, Department of Epidemiology of Microbial Diseases, LEPH, New Haven, Connecticut, United States of America
| | - Thiago Luiz Alves e Silva
- Yale School of Public Health, Department of Epidemiology of Microbial Diseases, LEPH, New Haven, Connecticut, United States of America
| | - Michelle O'Neill
- Yale School of Public Health, Department of Epidemiology of Microbial Diseases, LEPH, New Haven, Connecticut, United States of America
| | - Serap Aksoy
- Yale School of Public Health, Department of Epidemiology of Microbial Diseases, LEPH, New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
35
|
An investigation into the protein composition of the teneral Glossina morsitans morsitans peritrophic matrix. PLoS Negl Trop Dis 2014; 8:e2691. [PMID: 24763256 PMCID: PMC3998921 DOI: 10.1371/journal.pntd.0002691] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Accepted: 12/24/2013] [Indexed: 11/23/2022] Open
Abstract
Background Tsetse flies serve as biological vectors for several species of African trypanosomes. In order to survive, proliferate and establish a midgut infection, trypanosomes must cross the tsetse fly peritrophic matrix (PM), which is an acellular gut lining surrounding the blood meal. Crossing of this multi-layered structure occurs at least twice during parasite migration and development, but the mechanism of how trypanosomes do so is not understood. In order to better comprehend the molecular events surrounding trypanosome penetration of the tsetse PM, a mass spectrometry-based approach was applied to investigate the PM protein composition using Glossina morsitans morsitans as a model organism. Methods PMs from male teneral (young, unfed) flies were dissected, solubilised in urea/SDS buffer and the proteins precipitated with cold acetone/TCA. The PM proteins were either subjected to an in-solution tryptic digestion or fractionated on 1D SDS-PAGE, and the resulting bands digested using trypsin. The tryptic fragments from both preparations were purified and analysed by LC-MS/MS. Results Overall, nearly 300 proteins were identified from both analyses, several of those containing signature Chitin Binding Domains (CBD), including novel peritrophins and peritrophin-like glycoproteins, which are essential in maintaining PM architecture and may act as trypanosome adhesins. Furthermore, 27 proteins from the tsetse secondary endosymbiont, Sodalis glossinidius, were also identified, suggesting this bacterium is probably in close association with the tsetse PM. Conclusion To our knowledge this is the first report on the protein composition of teneral G. m. morsitans, an important vector of African trypanosomes. Further functional analyses of these proteins will lead to a better understanding of the tsetse physiology and may help identify potential molecular targets to block trypanosome development within the tsetse. African trypanosomes are transmitted by the haematophagous tsetse vector. For transmission to occur, bloodmeal ingested trypanosomes must overcome numerous barriers imposed by the fly. The first obstacle is the crossing of peritrophic matrix (PM), a cell-free structure that protects the midgut epithelial cells from coming under attack by the hosts' digestive enzymes, aids in water retention and helps prevent harmful pathogens from establishing a systemic infection. Trypanosomes cross the tsetse PM at least twice in their development but how they do so remains to be elucidated. Despite being a recognised barrier to trypanosome infections, there is limited knowledge of the molecular components of the tsetse PM. In this study we identified nearly 300 PM proteins using two mass spectrometry approaches. Several of the identified components were peritrophins, which are a key group of glycoproteins essential for PM integrity. In addition, we detected proteins from Sodalis glossinidius, a commensal bacterium linked to increased susceptibility to trypanosome infection in tsetse. Our study provides the first comprehensive identification of proteins from the tsetse PM, which provides a starting point for research into potential targets for vector control.
Collapse
|
36
|
Proto WR, Jones NG, Coombs GH, Mottram JC. Tracking autophagy during proliferation and differentiation of Trypanosoma brucei. MICROBIAL CELL (GRAZ, AUSTRIA) 2014; 1:9-20. [PMID: 28357206 PMCID: PMC5349162 DOI: 10.15698/mic2014.01.120] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 12/13/2013] [Indexed: 11/13/2022]
Abstract
Autophagy is a lysosome-dependent degradation mechanism that sequesters target cargo into autophagosomal vesicles. The Trypanosoma brucei genome contains apparent orthologues of several autophagy-related proteins including an ATG8 family. These ubiquitin-like proteins are required for autophagosome membrane formation, but our studies show that ATG8.3 is atypical. To investigate the function of other ATG proteins, RNAi compatible T. brucei were modified to function as autophagy reporter lines by expressing only either YFP-ATG8.1 or YFP-ATG8.2. In the insect procyclic lifecycle stage, independent RNAi down-regulation of ATG3 or ATG7 generated autophagy-defective mutants and confirmed a pro-survival role for autophagy in the procyclic form nutrient starvation response. Similarly, RNAi depletion of ATG5 or ATG7 in the bloodstream form disrupted autophagy, but did not impede proliferation. Further characterisation showed bloodstream form autophagy mutants retain the capacity to undergo the complex cellular remodelling that occurs during differentiation to the procyclic form and are equally susceptible to dihydroxyacetone-induced cell death as wild type parasites, not supporting a role for autophagy in this cell death mechanism. The RNAi reporter system developed, which also identified TOR1 as a negative regulator controlling YFP-ATG8.2 but not YFP-ATG8.1 autophagosome formation, will enable further targeted analysis of the mechanisms and function of autophagy in the medically relevant bloodstream form of T. brucei.
Collapse
Affiliation(s)
- William R. Proto
- Wellcome Trust Centre for Molecular Parasitology, Institute of
Infection, Immunity and Inflammation, College of Medical, Veterinary and Life
Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Nathaniel G. Jones
- Wellcome Trust Centre for Molecular Parasitology, Institute of
Infection, Immunity and Inflammation, College of Medical, Veterinary and Life
Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Graham H. Coombs
- Strathclyde Institute of Pharmacy and Biomedical Sciences,
University of Strathclyde, Glasgow, G4 0RE, UK
| | - Jeremy C. Mottram
- Wellcome Trust Centre for Molecular Parasitology, Institute of
Infection, Immunity and Inflammation, College of Medical, Veterinary and Life
Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| |
Collapse
|
37
|
Rodrigues JCF, Godinho JLP, de Souza W. Biology of human pathogenic trypanosomatids: epidemiology, lifecycle and ultrastructure. Subcell Biochem 2014; 74:1-42. [PMID: 24264239 DOI: 10.1007/978-94-007-7305-9_1] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Leishmania and Trypanosoma belong to the Trypanosomatidae family and cause important human infections such as leishmaniasis, Chagas disease, and sleeping sickness. Leishmaniasis, caused by protozoa belonging to Leishmania, affects about 12 million people worldwide and can present different clinical manifestations, i.e., visceral leishmaniasis (VL), cutaneous leishmaniasis (CL), mucocutaneous leishmaniasis (MCL), diffuse cutaneous leishmaniasis (DCL), and post-kala-azar dermal leishmaniasis (PKDL). Chagas disease, also known as American trypanosomiasis, is caused by Trypanosoma cruzi and is mainly prevalent in Latin America but is increasingly occurring in the United States, Canada, and Europe. Sleeping sickness or human African trypanosomiasis (HAT), caused by two sub-species of Trypanosoma brucei (i.e., T. b. rhodesiense and T. b. gambiense), occurs only in sub-Saharan Africa countries. These pathogenic trypanosomatids alternate between invertebrate and vertebrate hosts throughout their lifecycles, and different developmental stages can live inside the host cells and circulate in the bloodstream or in the insect gut. Trypanosomatids have a classical eukaryotic ultrastructural organization with some of the same main organelles found in mammalian host cells, while also containing special structures and organelles that are absent in other eukaryotic organisms. For example, the mitochondrion is ramified and contains a region known as the kinetoplast, which houses the mitochondrial DNA. Also, the glycosomes are specialized peroxisomes containing glycolytic pathway enzymes. Moreover, a layer of subpellicular microtubules confers mechanic rigidity to the cell. Some of these structures have been investigated to determine their function and identify potential enzymes and metabolic pathways that may constitute targets for new chemotherapeutic drugs.
Collapse
Affiliation(s)
- Juliany Cola Fernandes Rodrigues
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil,
| | | | | |
Collapse
|
38
|
Abstract
The complex life cycle of Trypanosoma brucei provides an excellent model system to understand signalling pathways that regulate development. We described previously the classical functions of TOR (target of rapamycin) 1 and TOR2 in T. brucei. In a more recent study, we described a novel TOR kinase, named TOR4, which regulates differentiation from the proliferative infective form to the quiescent form. In contrast with TOR1 loss-of-function, down-regulation of TOR4 triggers an irreversible differentiation process through the development of the insect pre-adapted quiescent form. TOR4 governs a signalling pathway distinct from those controlled by the conventional TOR complexes TORC1 and TORC2. Depletion of TOR4 induces all well-known characteristics of the quiescent developmental stage in trypanosomes, including expression of the PAD (proteins associated with differentiation) surface proteins and transcriptional down-regulation of the VSG (variant surface glycoprotein) gene. TOR4 kinase forms a structurally and functionally distinct complex named TORC4. TOR4 associates with LST8 (lethal with sec-13 protein 8) and other factors including an armadillo-domain-containing protein and the major vault protein, which probably serves as a scaffold for this kinase. Research in T. brucei, a protozoan parasite that diverged from the eukaryotic tree early in evolution, may help to uncover new functions of TOR kinases.
Collapse
|
39
|
Rotureau B, Van Den Abbeele J. Through the dark continent: African trypanosome development in the tsetse fly. Front Cell Infect Microbiol 2013; 3:53. [PMID: 24066283 PMCID: PMC3776139 DOI: 10.3389/fcimb.2013.00053] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 08/29/2013] [Indexed: 11/13/2022] Open
Abstract
African trypanosomes are unicellular flagellated parasites causing trypanosomiases in Africa, a group of severe diseases also known as sleeping sickness in human and nagana in cattle. These parasites are almost exclusively transmitted by the bite of the tsetse fly. In this review, we describe and compare the three developmental programs of the main trypanosome species impacting human and animal health, with focus on the most recent observations. From here, some reflections are made on research issues concerning trypanosome developmental biology in the tsetse fly that are to be addressed in the future.
Collapse
Affiliation(s)
- Brice Rotureau
- Trypanosome Cell Biology Unit, Institut Pasteur and CNRS URA 2581, Paris, France.
| | | |
Collapse
|
40
|
Wamwiri FN, Alam U, Thande PC, Aksoy E, Ngure RM, Aksoy S, Ouma JO, Murilla GA. Wolbachia, Sodalis and trypanosome co-infections in natural populations of Glossina austeni and Glossina pallidipes. Parasit Vectors 2013; 6:232. [PMID: 23924682 PMCID: PMC3751944 DOI: 10.1186/1756-3305-6-232] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 07/31/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Tsetse flies harbor at least three bacterial symbionts: Wigglesworthia glossinidia, Wolbachia pipientis and Sodalis glossinidius. Wigglesworthia and Sodalis reside in the gut in close association with trypanosomes and may influence establishment and development of midgut parasite infections. Wolbachia has been shown to induce reproductive effects in infected tsetse. This study was conducted to determine the prevalence of these endosymbionts in natural populations of G. austeni and G. pallidipes and to assess the degree of concurrent infections with trypanosomes. METHODS Fly samples analyzed originated from Kenyan coastal forests (trapped in 2009-2011) and South African G. austeni collected in 2008. The age structure was estimated by standard methods. G. austeni (n=298) and G. pallidipes (n= 302) were analyzed for infection with Wolbachia and Sodalis using PCR. Trypanosome infection was determined either by microscopic examination of dissected organs or by PCR amplification. RESULTS Overall we observed that G. pallidipes females had a longer lifespan (70 d) than G. austeni (54 d) in natural populations. Wolbachia infections were present in all G. austeni flies analysed, while in contrast, this symbiont was absent from G. pallidipes. The density of Wolbachia infections in the Kenyan G. austeni population was higher than that observed in South African flies. The infection prevalence of Sodalis ranged from 3.7% in G. austeni to about 16% in G. pallidipes. Microscopic examination of midguts revealed an overall trypanosome infection prevalence of 6% (n = 235) and 5% (n = 552), while evaluation with ITS1 primers indicated a prevalence of about 13% (n = 296) and 10% (n = 302) in G. austeni and G. pallidipes, respectively. The majority of infections (46%) were with T. congolense. Co-infection with all three organisms was observed at 1% and 3.3% in G. austeni and G. pallidipes, respectively. Eleven out of the thirteen (85%) co-infected flies harboured T. congolense and T. simiae parasites. While the association between trypanosomes and Sodalis infection was statistically significant in G. pallidipes (P = 0.0127), the number of co-infected flies was too few for a definite conclusion. CONCLUSIONS The tsetse populations analyzed differed in the prevalence of symbionts, despite being sympatric and therefore exposed to identical environmental factors. The density of infections with Wolbachia also differed between G. austeni populations. There were too few natural co-infections detected with the Sodalis and trypanosomes to suggest extensive inter-relations between these infections in natural populations. We discuss these findings in the context of potential symbiont-mediated control interventions to reduce parasite infections and/or fly populations.
Collapse
|
41
|
Weiss BL, Wang J, Maltz MA, Wu Y, Aksoy S. Trypanosome infection establishment in the tsetse fly gut is influenced by microbiome-regulated host immune barriers. PLoS Pathog 2013; 9:e1003318. [PMID: 23637607 PMCID: PMC3630092 DOI: 10.1371/journal.ppat.1003318] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 03/05/2013] [Indexed: 12/22/2022] Open
Abstract
Tsetse flies (Glossina spp.) vector pathogenic African trypanosomes, which cause sleeping sickness in humans and nagana in domesticated animals. Additionally, tsetse harbors 3 maternally transmitted endosymbiotic bacteria that modulate their host's physiology. Tsetse is highly resistant to infection with trypanosomes, and this phenotype depends on multiple physiological factors at the time of challenge. These factors include host age, density of maternally-derived trypanolytic effector molecules present in the gut, and symbiont status during development. In this study, we investigated the molecular mechanisms that result in tsetse's resistance to trypanosomes. We found that following parasite challenge, young susceptible tsetse present a highly attenuated immune response. In contrast, mature refractory flies express higher levels of genes associated with humoral (attacin and pgrp-lb) and epithelial (inducible nitric oxide synthase and dual oxidase) immunity. Additionally, we discovered that tsetse must harbor its endogenous microbiome during intrauterine larval development in order to present a parasite refractory phenotype during adulthood. Interestingly, mature aposymbiotic flies (Gmm(Apo)) present a strong immune response earlier in the infection process than do WT flies that harbor symbiotic bacteria throughout their entire lifecycle. However, this early response fails to confer significant resistance to trypanosomes. Gmm(Apo) adults present a structurally compromised peritrophic matrix (PM), which lines the fly midgut and serves as a physical barrier that separates luminal contents from immune responsive epithelial cells. We propose that the early immune response we observe in Gmm(Apo) flies following parasite challenge results from the premature exposure of gut epithelia to parasite-derived immunogens in the absence of a robust PM. Thus, tsetse's PM appears to regulate the timing of host immune induction following parasite challenge. Our results document a novel finding, which is the existence of a positive correlation between tsetse's larval microbiome and the integrity of the emerging adult PM gut immune barrier.
Collapse
Affiliation(s)
- Brian L Weiss
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, United States of America.
| | | | | | | | | |
Collapse
|
42
|
Dyer NA, Rose C, Ejeh NO, Acosta-Serrano A. Flying tryps: survival and maturation of trypanosomes in tsetse flies. Trends Parasitol 2013; 29:188-96. [PMID: 23507033 DOI: 10.1016/j.pt.2013.02.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Revised: 02/05/2013] [Accepted: 02/06/2013] [Indexed: 01/30/2023]
Abstract
Survival in and colonization of the tsetse fly midgut are essential steps in the transmission of many species of African trypanosomes. In the fly, bloodstream trypanosomes transform into the procyclic stage within the gut lumen and later migrate to the ectoperitrophic space, where they multiply, establishing an infection. Progression of the parasite infection in the fly depends on factors inherent to the biology of trypanosomes, tsetse, and the bloodmeal. Flies usually eradicate infection early on with both pre-existing and inducible factors. Parasites, in contrast, respond to these stimuli by undergoing developmental changes, allowing a few to both survive and migrate within the tsetse. Here we discuss parasite and fly factors determining trypanosome colonization of the tsetse, focusing mainly on the midgut.
Collapse
Affiliation(s)
- Naomi A Dyer
- Parasitology Department, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK.
| | | | | | | |
Collapse
|
43
|
Chitanga S, Namangala B, De Deken R, Marcotty T. Shifting from wild to domestic hosts: the effect on the transmission of Trypanosoma congolense to tsetse flies. Acta Trop 2013; 125:32-6. [PMID: 23000543 DOI: 10.1016/j.actatropica.2012.08.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Revised: 07/24/2012] [Accepted: 08/24/2012] [Indexed: 10/27/2022]
Abstract
The epidemiology and impact of animal African trypanosomosis are influenced by the transmissibility and the pathogenicity of the circulating trypanosome strains in a particular biotope. The transmissibility of 22 Trypanosoma congolense strains isolated from domestic and wild animals was evaluated in a total of 1213 flies. Multivariate mixed models were used to compare infection and maturation rates in function of trypanosome origin (domestic or sylvatic) and pathogenicity. Both trypanosome pathogenicity and origin significantly affected the ability to establish a midgut infection in tsetse flies but not the maturation rates. The interaction between pathogenicity and origin was not significant. Since being pathogenic and having a domestic origin both increased transmissibility, dominant lowly pathogenic trypanosomes from domestic environments and highly pathogenic trypanosomes from sylvatic environments presented similar levels of transmissibility: 12% and 15%, respectively. Blood meals with parasite concentration ranging from 0.05 to 50trypanosomes/μl blood for 3 strains of T. congolense were provided to different batches of tsetse flies to evaluate the relationship between the parasite load in blood meals and the likelihood for a fly to become infected. A linear relationship between parasite load and transmissibility was observed at low parasitaemia and a plateau was observed for meals containing more than 5trypanosomes/μl. Maximum transmission was reached with 12.5trypanosomes/μl blood. About 50% of the flies were refractory to T. congolense, whatever their concentration in the blood meal. The results suggest that the dose-transmissibility relationship presents a similar profile for different T. congolense isolates.
Collapse
|
44
|
Okwor I, Onyilagha C, Kuriakose S, Mou Z, Jia P, Uzonna JE. Regulatory T cells enhance susceptibility to experimental Trypanosoma congolense infection independent of mouse genetic background. PLoS Negl Trop Dis 2012; 6:e1761. [PMID: 22860150 PMCID: PMC3409116 DOI: 10.1371/journal.pntd.0001761] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Accepted: 06/20/2012] [Indexed: 12/22/2022] Open
Abstract
Background BALB/c mice are highly susceptible while C57BL/6 are relatively resistant to experimental Trypanosoma congolense infection. Although regulatory T cells (Tregs) have been shown to regulate the pathogenesis of experimental T. congolense infection, their exact role remains controversial. We wished to determine whether Tregs contribute to distinct phenotypic outcomes in BALB/c and C57BL/6 mice and if so how they operate with respect to control of parasitemia and production of disease-exacerbating proinflammatory cytokines. Methodology/Findings BALB/c and C57BL/6 mice were infected intraperitoneally (i.p) with 103T. congolense clone TC13 and both the kinetics of Tregs expansion and intracellular cytokine profiles in the spleens and livers were monitored directly ex vivo by flow cytometry. In some experiments, mice were injected with anti-CD25 mAb prior or post T. congolense infection or adoptively (by intravenous route) given highly enriched naïve CD25+ T lymphocytes prior to T. congolense infection and the inflammatory cytokine/chemokine levels and survival were monitored. In contrast to a transient and non significant increase in the percentages and absolute numbers of CD4+CD25+Foxp3+ T cells (Tregs) in C57BL/6 mouse spleens and livers, a significant increase in the percentage and absolute numbers of Tregs was observed in spleens of infected BALB/c mice. Ablation or increasing the number of CD25+ cells in the relatively resistant C57BL/6 mice by anti-CD25 mAb treatment or by adoptive transfer of CD25+ T cells, respectively, ameliorates or exacerbates parasitemia and production of proinflammatory cytokines. Conclusion Collectively, our results show that regulatory T cells contribute to susceptibility in experimental murine trypanosomiasis in both the highly susceptible BALB/c and relatively resistant C57BL/6 mice. BALB/c mice are highly susceptible while C57BL/6 is relatively resistant to experimental Trypanosoma congolense infection. Acute death observed in infected BALB/c mice is usually associated with the excessive production of pro-inflammatory cytokines. Regulatory T cells (Tregs) have been shown to play a significant role in the pathogenesis of many diseases including those caused by parasites. However, the role of Tregs in the pathogenesis of T. congolense infection remains unclear. We were interested in addressing the following questions: Do Tregs contribute to the distinct phenotypic outcomes observed in T. congolense-infected BALB/c and C57BL/6 mice? If so, where and how do they operate with respect to parasitemia and cytokine response? By selectively altering the numbers of these cells either by targeted depletion with monoclonal antibody or adoptive transfer of highly enriched naïve CD25+ cells prior to infection, we show that Tregs impairs efficient parasite control and impacts on production of disease-exacerbating proinflammatory cytokines. Collectively, our findings suggest that Tregs contribute to enhanced susceptibility to experimental T. congolense infection in mice.
Collapse
Affiliation(s)
- Ifeoma Okwor
- Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Chukwunonso Onyilagha
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Shiby Kuriakose
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Zhirong Mou
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ping Jia
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jude E. Uzonna
- Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- * E-mail:
| |
Collapse
|
45
|
Savage AF, Cerqueira GC, Regmi S, Wu Y, El Sayed NM, Aksoy S. Transcript expression analysis of putative Trypanosoma brucei GPI-anchored surface proteins during development in the tsetse and mammalian hosts. PLoS Negl Trop Dis 2012; 6:e1708. [PMID: 22724039 PMCID: PMC3378594 DOI: 10.1371/journal.pntd.0001708] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Accepted: 05/11/2012] [Indexed: 11/26/2022] Open
Abstract
Human African Trypanosomiasis is a devastating disease caused by the parasite Trypanosoma brucei. Trypanosomes live extracellularly in both the tsetse fly and the mammal. Trypanosome surface proteins can directly interact with the host environment, allowing parasites to effectively establish and maintain infections. Glycosylphosphatidylinositol (GPI) anchoring is a common posttranslational modification associated with eukaryotic surface proteins. In T. brucei, three GPI-anchored major surface proteins have been identified: variant surface glycoproteins (VSGs), procyclic acidic repetitive protein (PARP or procyclins), and brucei alanine rich proteins (BARP). The objective of this study was to select genes encoding predicted GPI-anchored proteins with unknown function(s) from the T. brucei genome and characterize the expression profile of a subset during cyclical development in the tsetse and mammalian hosts. An initial in silico screen of putative T. brucei proteins by Big PI algorithm identified 163 predicted GPI-anchored proteins, 106 of which had no known functions. Application of a second GPI-anchor prediction algorithm (FragAnchor), signal peptide and trans-membrane domain prediction software resulted in the identification of 25 putative hypothetical proteins. Eighty-one gene products with hypothetical functions were analyzed for stage-regulated expression using semi-quantitative RT-PCR. The expression of most of these genes were found to be upregulated in trypanosomes infecting tsetse salivary gland and proventriculus tissues, and 38% were specifically expressed only by parasites infecting salivary gland tissues. Transcripts for all of the genes specifically expressed in salivary glands were also detected in mammalian infective metacyclic trypomastigotes, suggesting a possible role for these putative proteins in invasion and/or establishment processes in the mammalian host. These results represent the first large-scale report of the differential expression of unknown genes encoding predicted T. brucei surface proteins during the complete developmental cycle. This knowledge may form the foundation for the development of future novel transmission blocking strategies against metacyclic parasites. Human African Trypanosomiasis (HAT) is a fatal disease caused by African trypanosomes and transmitted by an infected tsetse fly. Presently, there are no vaccines to prevent mammalian infections. Proteins expressed on the trypanosome surface can influence the host environment and allow for their transmission. Potentially accessible to the adaptive immune systems of vertebrate hosts, these proteins could serve as future vaccine targets. Identification and characterization of these currently unknown proteins can help us develop strategies to alter the host environment, making it inhospitable for the parasite, thereby reducing disease transmission. While there is extensive knowledge about trypanosome development in the mammalian host, less is known about the molecular events in the tsetse fly, particularly the salivary gland stages. We used an in silico approach to identify putative surface proteins from the known genome sequence of Trypanosoma brucei, and we describe the stage specific expression of these genes during development in the tsetse fly and mammalian host. Our findings show that a majority of unknown transcripts encoding predicted surface proteins are expressed by the parasites infecting tsetse salivary glands. These data will help focus future investigations into transmission-blocking approaches targeting the expressed antigens of trypanosomes infecting tsetse salivary glands.
Collapse
Affiliation(s)
- Amy F. Savage
- Division of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, Connecticut, United States of America
| | - Gustavo C. Cerqueira
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute (MPRI), University of Maryland, College Park, Maryland, United States of America
| | - Sandesh Regmi
- Division of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, Connecticut, United States of America
| | - Yineng Wu
- Division of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, Connecticut, United States of America
| | - Najib M. El Sayed
- Department of Cell Biology and Molecular Genetics, Maryland Pathogen Research Institute (MPRI), University of Maryland, College Park, Maryland, United States of America
- Center for Bioinformatics and Computational Biology, College of Chemical & Life Sciences, University of Maryland, College Park, Maryland, United States of America
| | - Serap Aksoy
- Division of Epidemiology of Microbial Diseases, Yale School of Public Health, Yale University, New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
46
|
Rotureau B, Subota I, Buisson J, Bastin P. A new asymmetric division contributes to the continuous production of infective trypanosomes in the tsetse fly. Development 2012; 139:1842-50. [PMID: 22491946 DOI: 10.1242/dev.072611] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
African trypanosomes are flagellated protozoan parasites that cause sleeping sickness and are transmitted by the bite of the tsetse fly. To complete their life cycle in the insect, trypanosomes reach the salivary glands and transform into the metacyclic infective form. The latter are expelled with the saliva at each blood meal during the whole life of the insect. Here, we reveal a means by which the continuous production of infective parasites could be ensured. Dividing trypanosomes present in the salivary glands of infected tsetse flies were monitored by live video-microscopy and by quantitative immunofluorescence analysis using molecular markers for the cytoskeleton and for surface antigens. This revealed the existence of two distinct modes of trypanosome proliferation occurring simultaneously in the salivary glands. The first cycle produces two equivalent cells that are not competent for infection and are attached to the epithelium. This mode of proliferation is predominant at the early steps of infection, ensuring a rapid colonization of the glands. The second mode is more frequent at later stages of infection and involves an asymmetric division. It produces a daughter cell that matures into the infective metacyclic form that is released in the saliva, as demonstrated by the expression of specific molecular markers - the calflagins. The levels of these calcium-binding proteins increase exclusively in the new flagellum during the asymmetric division, showing the commitment of the future daughter cell to differentiation. The coordination of these two alternative cell cycles contributes to the continuous production of infective parasites, turning the tsetse fly into an efficient and long-lasting vector for African trypanosomes.
Collapse
Affiliation(s)
- Brice Rotureau
- Trypanosome Cell Biology Unit, Institut Pasteur & CNRS, URA 2581, 25 rue du Docteur Roux, 75015 Paris, France
| | | | | | | |
Collapse
|
47
|
Motloang M, Masumu J, Mans B, Van den Bossche P, Latif A. Vector competence of Glossina austeni and Glossina brevipalpis for Trypanosoma congolense in KwaZulu-Natal, South Africa. ACTA ACUST UNITED AC 2012; 79:E1-6. [PMID: 23327306 DOI: 10.4102/ojvr.v79i1.353] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Revised: 10/19/2011] [Accepted: 10/19/2011] [Indexed: 01/11/2023]
Abstract
Tsetse-transmitted trypanosomosis (nagana) has been the cause of stock losses in the recent past and still presents a major problem to livestock owners in certain areas of KwaZulu- Natal, South Africa. Over 10 000 cattle mortalities were reported in the 1990 nagana outbreak. Although information on the distribution and abundance of the tsetse flies Glossina brevipalpis and Glossina austeni in KwaZulu-Natal exists, data on their vector competence are lacking. This study aimed to determine the rate of natural Trypanosoma congolense infection by field-collected as well as colony-reared flies of these species. A total of 442 field-collected G. brevipalpis and 40 G. austeni flies were dissected immediately after collection to determine their infection rates, whilst 699 G. brevipalpis and 49 G. austeni flies were fed on susceptible animals in 10 and four batches, respectively, for use in xenodiagnosis experiments. Teneral colony flies were fed on infected animals and dissected 21 days post infection to confirm their infectivity testing. Glossina austeni harboured 8% immature and mature infections. In G. brevipalpis, the infection with the immature stages was lower (1%) and no mature infections were observed. Although all four batches of G. austeni transmitted T. congolense to four susceptible animals, no transmission resulted from 10 batches of G. brevipalpis fed on susceptible cattle. Colony-derived G. austeni (534) and G. brevipalpis (882) were fed on four bovines infected with different T. congolense isolates. Both G. austeni and G. brevipalpis acquired trypanosome infection from the bovines, with immature infection ranges of 20% - 33% and 1% - 4%, respectively. Parasites, however, only matured in G. austeni (average = 4%). Glossina austeni plays a larger role in the epidemiology of animal trypanosomosis in KwaZulu-Natal than G. brevipalpis and therefore more focus should be aimed at the former when control measures are implemented.
Collapse
Affiliation(s)
- Makhosazana Motloang
- Parasites, Vectors & Vector-borne Diseases Programme, ARC-Onderstepoort Veterinary Institute, Onderstepoort.
| | | | | | | | | |
Collapse
|
48
|
Peacock L, Ferris V, Bailey M, Gibson W. The influence of sex and fly species on the development of trypanosomes in tsetse flies. PLoS Negl Trop Dis 2012; 6:e1515. [PMID: 22348165 PMCID: PMC3279344 DOI: 10.1371/journal.pntd.0001515] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Accepted: 12/19/2011] [Indexed: 01/01/2023] Open
Abstract
Unlike other dipteran disease vectors, tsetse flies of both sexes feed on blood and transmit pathogenic African trypanosomes. During transmission, Trypanosoma brucei undergoes a complex cycle of proliferation and development inside the tsetse vector, culminating in production of infective forms in the saliva. The insect manifests robust immune defences throughout the alimentary tract, which eliminate many trypanosome infections. Previous work has shown that fly sex influences susceptibility to trypanosome infection as males show higher rates of salivary gland (SG) infection with T. brucei than females. To investigate sex-linked differences in the progression of infection, we compared midgut (MG), proventriculus, foregut and SG infections in male and female Glossina morsitans morsitans. Initially, infections developed in the same way in both sexes: no difference was observed in numbers of MG or proventriculus infections, or in the number and type of developmental forms produced. Female flies tended to produce foregut migratory forms later than males, but this had no detectable impact on the number of SG infections. The sex difference was not apparent until the final stage of SG invasion and colonisation, showing that the SG environment differs between male and female flies. Comparison of G. m. morsitans with G. pallidipes showed a similar, though less pronounced, sex difference in susceptibility, but additionally revealed very different levels of trypanosome resistance in the MG and SG. While G. pallidipes was more refractory to MG infection, a very high proportion of MG infections led to SG infection in both sexes. It appears that the two fly species use different strategies to block trypanosome infection: G. pallidipes heavily defends against initial establishment in the MG, while G. m. morsitans has additional measures to prevent trypanosomes colonising the SG, particularly in female flies. We conclude that the tsetse-trypanosome interface works differently in G. m. morsitans and G. pallidipes. In tropical Africa human and livestock diseases caused by parasitic trypanosomes are transmitted by bloodsucking tsetse flies. In the fly, trypanosomes undergo a complex cycle of proliferation and development during their remarkable journey from the midgut to the salivary glands. At every step of the way, the flies mount robust immune defences against trypanosome infection and consequently most flies fail to develop a transmissible infection. Previous work has shown a sex difference in the numbers of salivary gland infections with Trypanosoma brucei: male flies are more susceptible to salivary gland infection than females. Here we explored possible reasons for this. Infections developed in the same way in both male and female flies until the final stage of salivary gland invasion and colonisation. We conclude that the salivary gland environment in the female fly is much more inhospitable for trypanosomes, perhaps because of a greater immune response. Comparison of two different tsetse species showed very different levels of trypanosome resistance in the midgut and salivary glands.
Collapse
Affiliation(s)
- Lori Peacock
- School of Biological Sciences, University of Bristol, Bristol, United Kingdom
- School of Clinical Veterinary Science, University of Bristol, Bristol, United Kingdom
| | - Vanessa Ferris
- School of Biological Sciences, University of Bristol, Bristol, United Kingdom
- School of Clinical Veterinary Science, University of Bristol, Bristol, United Kingdom
| | - Mick Bailey
- School of Clinical Veterinary Science, University of Bristol, Bristol, United Kingdom
| | - Wendy Gibson
- School of Biological Sciences, University of Bristol, Bristol, United Kingdom
- * E-mail:
| |
Collapse
|
49
|
Subota I, Rotureau B, Blisnick T, Ngwabyt S, Durand-Dubief M, Engstler M, Bastin P. ALBA proteins are stage regulated during trypanosome development in the tsetse fly and participate in differentiation. Mol Biol Cell 2011; 22:4205-19. [PMID: 21965287 PMCID: PMC3216647 DOI: 10.1091/mbc.e11-06-0511] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The protozoan parasite Trypanosoma brucei is responsible for sleeping sickness and alternates between mammal and tsetse fly hosts. Two proteins of the ALBA family associate to mRNA in cytoplasmic granules during starvation stress, are stage regulated, and contribute to trypanosome development in the tsetse fly. The protozoan parasite Trypanosoma brucei is responsible for sleeping sickness and alternates between mammal and tsetse fly hosts, where it has to adapt to different environments. We investigated the role of two members of the ALBA family, which encodes hypothetical RNA-binding proteins conserved in most eukaryotes. We show that ALBA3/4 proteins colocalize with the DHH1 RNA-binding protein and with a subset of poly(A+) RNA in stress granules upon starvation. Depletion of ALBA3/4 proteins by RNA interference in the cultured procyclic stage produces cell modifications mimicking several morphogenetic aspects of trypanosome differentiation that usually take place in the fly midgut. A combination of immunofluorescence data and videomicroscopy analysis of live trypanosomes expressing endogenously ALBA fused with fluorescent proteins revealed that ALBA3/4 are present throughout the development of the parasite in the tsetse fly, with the striking exception of the transition stages found in the proventriculus region. This involves migration of the nucleus toward the posterior end of the cell, a phenomenon that is perturbed upon forced expression of ALBA3 during the differentiation process, showing for the first time the involvement of an RNA-binding protein in trypanosome development in vivo.
Collapse
Affiliation(s)
- Ines Subota
- Trypanosome Cell Biology Unit, Centre National de la Recherche Scientifique, Parasitology and Mycology Department, Institut Pasteur, 75015 Paris, France
| | | | | | | | | | | | | |
Collapse
|
50
|
Microbiome influences on insect host vector competence. Trends Parasitol 2011; 27:514-22. [PMID: 21697014 DOI: 10.1016/j.pt.2011.05.001] [Citation(s) in RCA: 264] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Revised: 04/29/2011] [Accepted: 05/09/2011] [Indexed: 11/23/2022]
Abstract
Insect symbioses lack the complexity and diversity of those associated with higher eukaryotic hosts. Symbiotic microbiomes are beneficial to their insect hosts in many ways, including dietary supplementation, tolerance to environmental perturbations and maintenance and/or enhancement of host immune system homeostasis. Recent studies have also highlighted the importance of the microbiome in the context of host pathogen transmission processes. Here we provide an overview of the relationship between insect disease vectors, such as tsetse flies and mosquitoes, and their associated microbiome. Several mechanisms are discussed through which symbiotic microbes can influence the ability of their host to transmit pathogens, as well as potential disease control strategies that harness symbiotic microbes to reduce pathogen transmission through an insect vector.
Collapse
|