1
|
Nakayama K, Haraguchi A, Hakozaki J, Nakamura S, Kusakisako K, Ikadai H. The C-terminal region of the Plasmodium berghei gamete surface 184-kDa protein Pb184 contributes to fertilization and male gamete binding to the residual body. Parasit Vectors 2024; 17:304. [PMID: 39003498 PMCID: PMC11246575 DOI: 10.1186/s13071-024-06374-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/24/2024] [Indexed: 07/15/2024] Open
Abstract
BACKGROUND Malaria, a global health concern, is caused by parasites of the Plasmodium genus, which undergo gametogenesis in the midgut of mosquitoes after ingestion of an infected blood meal. The resulting male and female gametes fuse to form a zygote, which differentiates into a motile ookinete. After traversing the midgut epithelium, the ookinete differentiates into an oocyst on the basal side of the epithelium. METHODS Membrane proteins with increased gene expression levels from the gamete to oocyst stages in P. berghei were investigated utilizing PlasmoDB, the functional genomic database for Plasmodium spp. Based on this analysis, we selected the 184-kDa membrane protein, Pb184, for further study. The expression of Pb184 was further confirmed through immunofluorescence staining, following which we examined whether Pb184 is involved in fertilization using antibodies targeting the C-terminal region of Pb184 and biotin-labeled C-terminal region peptides of Pb184. RESULTS Pb184 is expressed on the surface of male and female gametes. The antibody inhibited zygote and ookinete formation in vitro. When mosquitoes were fed on parasite-infected blood containing the antibody, oocyst formation decreased on the second day after feeding. Synthesized biotin-labeled peptides matching the C-terminal region of Pb184 bound to the female gamete and the residual body of male gametes, and inhibited differentiation into ookinetes in the in vitro culture system. CONCLUSIONS These results may be useful for the further studying the fertilization mechanism of Plasmodium protozoa. There is also the potential for their application as future tools to prevent malaria transmission.
Collapse
Affiliation(s)
- Kazuhiko Nakayama
- Laboratory of Veterinary Parasitology, School of Veterinary Medicine, Kitasato University, Aomori, Towada, 034-8628, Japan
| | - Asako Haraguchi
- Laboratory of Veterinary Parasitology, School of Veterinary Medicine, Kitasato University, Aomori, Towada, 034-8628, Japan
| | - Jun Hakozaki
- Laboratory of Veterinary Parasitology, School of Veterinary Medicine, Kitasato University, Aomori, Towada, 034-8628, Japan
| | - Sakure Nakamura
- Laboratory of Veterinary Parasitology, School of Veterinary Medicine, Kitasato University, Aomori, Towada, 034-8628, Japan
| | - Kodai Kusakisako
- Laboratory of Veterinary Parasitology, School of Veterinary Medicine, Kitasato University, Aomori, Towada, 034-8628, Japan
| | - Hiromi Ikadai
- Laboratory of Veterinary Parasitology, School of Veterinary Medicine, Kitasato University, Aomori, Towada, 034-8628, Japan.
| |
Collapse
|
2
|
Ravindran J, Eapen A. A comparative study on the vector competence of Anopheles stephensi from geographically distinct malarious and non-malarious urban areas in India to the malarial parasite, Plasmodium vivax. J Vector Borne Dis 2024; 61:413-419. [PMID: 38634456 DOI: 10.4103/jvbd.jvbd_7_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 03/15/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND OBJECTIVES Anopheles stephensi is responsible for the transmission of malaria in urban areas. Vector competence of An. stephensi from a non-malarious (Coimbatore) and highly malarious (Chennai) urban area were investigated to find out the reason for the non-transmission of malaria in Coimbatore. METHODS Vector competence (Susceptibility/refractoriness) of An. stephensi mosquitoes from Chennai (Malarious) and Coimbatore (Non-malarious), Tamil Nadu, India to Plasmodium vivax (Chennai) were investigated. Bioassays were carried out concurrently in both these strains by artificial membrane feeding technique using the same malaria infected blood. An. stephensi were dissected to observe infection in the midgut and salivary glands. The parasite infection, oocyst and sporozoite positivity rate, the oocyst load, correlation between male-female gametocyte ratio and infection, and Survival Analysis of parasitic stages during sporogony were analyzed and compared. RESULTS The overall infection rate was 45.8 and 41.2 per cent in Chennai and Coimbatore. Oocyst count ranged from 1-80 and 1-208 respectively and not statistically significant. Oocyst positivity was high from Day 8-21in both strains. The Mean Survival Day (MSD) for oocyst was Day 14 in both strains. Sporozoite was observed in four experiments in each of the strains and the MSD for sporozoites was Day 20 and Day 17 in Chennai and Coimbatore. INTERPRETATION CONCLUSION An. stephensi of Chennai and Coimbatore are equally susceptible to P. vivax infection and the non-transmission of malaria in Coimbatore can be attributed to external factors such as the presence of preferential breeding habitat, vector density, vector survival, and weather. The only difference observed was the comparatively shortened oocyst maturation time in the Coimbatore strain which requires further investigation.
Collapse
Affiliation(s)
- John Ravindran
- ICMR-National Institute of Malaria Research, Field Unit, Chennai, India
| | | |
Collapse
|
3
|
Cardoso-Jaime V, Maya-Maldonado K, Tsutsumi V, Hernández-Martínez S. Mosquito pericardial cells upregulate Cecropin expression after an immune challenge. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 147:104745. [PMID: 37268262 DOI: 10.1016/j.dci.2023.104745] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/04/2023]
Abstract
Most mosquito-transmitted pathogens grow or replicate in the midgut before invading the salivary glands. Pathogens are exposed to several immunological factors along the way. Recently, it was shown that hemocytes gather near the periostial region of the heart to efficiently phagocytose pathogens circulating in the hemolymph. Nerveless, not all pathogens can be phagocyted by hemocytes and eliminated by lysis. Interestingly, some studies have shown that pericardial cells (PCs) surrounding periostial regions, may produce humoral factors, such as lysozymes. Our current work provides evidence that Anopheles albimanus PCs are a major producer of Cecropin 1 (Cec1). Furthermore, our findings reveal that after an immunological challenge, PCs upregulate Cec1 expression. We conclude that PCs are positioned in a strategic location that could allow releasing humoral components, such as cecropin, to lyse pathogens on the heart or circulating in the hemolymph, implying that PCs could play a significant role in the systemic immune response.
Collapse
Affiliation(s)
- Victor Cardoso-Jaime
- Centro de Investigaciones Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública. Av. Universidad 655, Santa María Ahuacatitlan, Cuernavaca, Morelos, C.P. 62100, Mexico; Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados, IPN. Av. Instituto Politécnico Nacional 2508, Gustavo A. Madero, Ciudad de México, C.P. 07360, Mexico
| | - Krystal Maya-Maldonado
- Centro de Investigaciones Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública. Av. Universidad 655, Santa María Ahuacatitlan, Cuernavaca, Morelos, C.P. 62100, Mexico; Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados, IPN. Av. Instituto Politécnico Nacional 2508, Gustavo A. Madero, Ciudad de México, C.P. 07360, Mexico
| | - Víctor Tsutsumi
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados, IPN. Av. Instituto Politécnico Nacional 2508, Gustavo A. Madero, Ciudad de México, C.P. 07360, Mexico.
| | - Salvador Hernández-Martínez
- Centro de Investigaciones Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública. Av. Universidad 655, Santa María Ahuacatitlan, Cuernavaca, Morelos, C.P. 62100, Mexico.
| |
Collapse
|
4
|
Huang W, Rodrigues J, Bilgo E, Tormo JR, Challenger JD, De Cozar-Gallardo C, Pérez-Victoria I, Reyes F, Castañeda-Casado P, Gnambani EJ, Hien DFDS, Konkobo M, Urones B, Coppens I, Mendoza-Losana A, Ballell L, Diabate A, Churcher TS, Jacobs-Lorena M. Delftia tsuruhatensis TC1 symbiont suppresses malaria transmission by anopheline mosquitoes. Science 2023; 381:533-540. [PMID: 37535741 DOI: 10.1126/science.adf8141] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 06/22/2023] [Indexed: 08/05/2023]
Abstract
Malaria control demands the development of a wide range of complementary strategies. We describe the properties of a naturally occurring, non-genetically modified symbiotic bacterium, Delftia tsuruhatensis TC1, which was isolated from mosquitoes incapable of sustaining the development of Plasmodium falciparum parasites. D. tsuruhatensis TC1 inhibits early stages of Plasmodium development and subsequent transmission by the Anopheles mosquito through secretion of a small-molecule inhibitor. We have identified this inhibitor to be the hydrophobic molecule harmane. We also found that, on mosquito contact, harmane penetrates the cuticle, inhibiting Plasmodium development. D. tsuruhatensis TC1 stably populates the mosquito gut, does not impose a fitness cost on the mosquito, and inhibits Plasmodium development for the mosquito's life. Contained field studies in Burkina Faso and modeling showed that D. tsuruhatensis TC1 has the potential to complement mosquito-targeted malaria transmission control.
Collapse
Affiliation(s)
- Wei Huang
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | | | - Etienne Bilgo
- Institut de Recherche en Sciences de la Santé (IRSS), Bobo-Dioulasso BP: 545, Burkina Faso
| | | | - Joseph D Challenger
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London W2 1PG, UK
| | | | | | | | - Pablo Castañeda-Casado
- Drug Metabolism and Pharmacokinetics (DMPK) Discovery, In Vitro/In Vivo Translation (IVIVT), GSK, 28760 Tres Cantos, Madrid, Spain
| | | | | | - Maurice Konkobo
- Institut de Recherche en Sciences de la Santé (IRSS), Bobo-Dioulasso BP: 545, Burkina Faso
| | - Beatriz Urones
- Global Health Medicines R&D, GSK, Tres Cantos, 28760 Madrid, Spain
| | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | | | - Lluís Ballell
- Global Health Medicines R&D, GSK, Tres Cantos, 28760 Madrid, Spain
| | - Abdoulaye Diabate
- Institut de Recherche en Sciences de la Santé (IRSS), Bobo-Dioulasso BP: 545, Burkina Faso
| | - Thomas S Churcher
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London W2 1PG, UK
| | - Marcelo Jacobs-Lorena
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| |
Collapse
|
5
|
Salimo ZM, Barros AL, Adrião AAX, Rodrigues AM, Sartim MA, de Oliveira IS, Pucca MB, Baia-da-Silva DC, Monteiro WM, de Melo GC, Koolen HHF. Toxins from Animal Venoms as a Potential Source of Antimalarials: A Comprehensive Review. Toxins (Basel) 2023; 15:375. [PMID: 37368676 DOI: 10.3390/toxins15060375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/23/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
Malaria is an infectious disease caused by Plasmodium spp. and it is mainly transmitted to humans by female mosquitoes of the genus Anopheles. Malaria is an important global public health problem due to its high rates of morbidity and mortality. At present, drug therapies and vector control with insecticides are respectively the most commonly used methods for the treatment and control of malaria. However, several studies have shown the resistance of Plasmodium to drugs that are recommended for the treatment of malaria. In view of this, it is necessary to carry out studies to discover new antimalarial molecules as lead compounds for the development of new medicines. In this sense, in the last few decades, animal venoms have attracted attention as a potential source for new antimalarial molecules. Therefore, the aim of this review was to summarize animal venom toxins with antimalarial activity found in the literature. From this research, 50 isolated substances, 4 venom fractions and 7 venom extracts from animals such as anurans, spiders, scorpions, snakes, and bees were identified. These toxins act as inhibitors at different key points in the biological cycle of Plasmodium and may be important in the context of the resistance of Plasmodium to currently available antimalarial drugs.
Collapse
Affiliation(s)
- Zeca M Salimo
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus 69040-000, Brazil
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus 69040-000, Brazil
- Grupo de Pesquisa em Metabolômica e Espectrometria de Massas, Universidade do Estado do Amazonas, Manaus 69065-001, Brazil
| | - André L Barros
- Setor de Medicina Veterinária, Universidade Nilton Lins, Manaus 69058-030, Brazil
| | - Asenate A X Adrião
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus 69040-000, Brazil
- Grupo de Pesquisa em Metabolômica e Espectrometria de Massas, Universidade do Estado do Amazonas, Manaus 69065-001, Brazil
- Programa de Pós-Graduação em Biodiversidade e Biotecnologia-Rede BIONORTE, Universidade do Estado do Amazonas, Manaus 69065-001, Brazil
| | - Aline M Rodrigues
- Grupo de Pesquisa em Metabolômica e Espectrometria de Massas, Universidade do Estado do Amazonas, Manaus 69065-001, Brazil
| | - Marco A Sartim
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus 69040-000, Brazil
- Programa de Pós-Graduação em Biodiversidade e Biotecnologia-Rede BIONORTE, Universidade do Estado do Amazonas, Manaus 69065-001, Brazil
- Pro-Reitoria de Pesquisa e Pós-Graduação, Universidade Nilton Lins, Manaus 69058-030, Brazil
| | - Isadora S de Oliveira
- Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto 14040-903, Brazil
| | - Manuela B Pucca
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus 69040-000, Brazil
- Faculdade de Medicina, Universidade Federal de Roraima, Boa Vista 69317-810, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Roraima, Boa Vista 69317-810, Brazil
| | - Djane C Baia-da-Silva
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus 69040-000, Brazil
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus 69040-000, Brazil
- Faculdade de Farmácia, Universidade Nilton Lins, Manaus 69058-030, Brazil
- Instituto Leônidas e Maria Deane, Fundação Oswaldo Cruz, Manaus 69057-070, Brazil
- Programa de Pós Graduação em Ciências Farmacêuticas, Universidade Federal do Amazonas, Manaus 69080-900, Brazil
| | - Wuelton M Monteiro
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus 69040-000, Brazil
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus 69040-000, Brazil
| | - Gisely C de Melo
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus 69040-000, Brazil
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus 69040-000, Brazil
| | - Hector H F Koolen
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus 69040-000, Brazil
- Grupo de Pesquisa em Metabolômica e Espectrometria de Massas, Universidade do Estado do Amazonas, Manaus 69065-001, Brazil
- Programa de Pós-Graduação em Biodiversidade e Biotecnologia-Rede BIONORTE, Universidade do Estado do Amazonas, Manaus 69065-001, Brazil
| |
Collapse
|
6
|
Awasthi V, Gupta Y, Chauhan R, Kempaiah P, Das J. Growth inhibition of plasmodium falciparum by Nano-molar concentrations of 1-(4‑hydroxy-3-methoxyphenyl) decan-3-one (6-paradol); is a cure at hand? PHYTOMEDICINE PLUS 2022; 2:100208. [DOI: 10.1016/j.phyplu.2021.100208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2025]
|
7
|
Keleta Y, Ramelow J, Cui L, Li J. Molecular interactions between parasite and mosquito during midgut invasion as targets to block malaria transmission. NPJ Vaccines 2021; 6:140. [PMID: 34845210 PMCID: PMC8630063 DOI: 10.1038/s41541-021-00401-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 11/01/2021] [Indexed: 11/21/2022] Open
Abstract
Despite considerable effort, malaria remains a major public health burden. Malaria is caused by five Plasmodium species and is transmitted to humans via the female Anopheles mosquito. The development of malaria vaccines against the liver and blood stages has been challenging. Therefore, malaria elimination strategies advocate integrated measures, including transmission-blocking approaches. Designing an effective transmission-blocking strategy relies on a sophisticated understanding of the molecular mechanisms governing the interactions between the mosquito midgut molecules and the malaria parasite. Here we review recent advances in the biology of malaria transmission, focusing on molecular interactions between Plasmodium and Anopheles mosquito midgut proteins. We provide an overview of parasite and mosquito proteins that are either targets for drugs currently in clinical trials or candidates of promising transmission-blocking vaccines.
Collapse
Affiliation(s)
- Yacob Keleta
- Department of Biological Sciences, Florida International University, Miami, FL, 33199, USA
| | - Julian Ramelow
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Liwang Cui
- College of Public Health, University of South Florida, Tampa, FL, 33612, USA
| | - Jun Li
- Department of Biological Sciences, Florida International University, Miami, FL, 33199, USA.
- Biomolecular Science Institute, Florida International University, Miami, FL, 33199, USA.
| |
Collapse
|
8
|
Nakayama K, Kimura Y, Kitahara Y, Soga A, Haraguchi A, Hakozaki J, Sugiyama M, Kusakisako K, Fukumoto S, Ikadai H. Role of Plasmodium berghei ookinete surface and oocyst capsule protein, a novel oocyst capsule-associated protein, in ookinete motility. Parasit Vectors 2021; 14:373. [PMID: 34289894 PMCID: PMC8296654 DOI: 10.1186/s13071-021-04868-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 06/30/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Plasmodium sp., which causes malaria, must first develop in mosquitoes before being transmitted. Upon ingesting infected blood, gametes form in the mosquito lumen, followed by fertilization and differentiation of the resulting zygotes into motile ookinetes. Within 24 h of blood ingestion, these ookinetes traverse mosquito epithelial cells and lodge below the midgut basal lamina, where they differentiate into sessile oocysts that are protected by a capsule. METHODS We identified an ookinete surface and oocyst capsule protein (OSCP) that is involved in ookinete motility as well as oocyst capsule formation. RESULTS We found that knockout of OSCP in parasite decreases ookinete gliding motility and gradually reduces the number of oocysts. On day 15 after blood ingestion, the oocyst wall was significantly thinner. Moreover, adding anti-OSCP antibodies decreased the gliding speed of wild-type ookinetes in vitro. Adding anti-OSCP antibodies to an infected blood meal also resulted in decreased oocyst formation. CONCLUSION These findings may be useful for the development of a transmission-blocking tool for malaria.
Collapse
Affiliation(s)
- Kazuhiko Nakayama
- Laboratory of Veterinary Parasitology, School of Veterinary Medicine, Kitasato University, Towada, Aomori, 034-8628, Japan
| | - Yuta Kimura
- Laboratory of Veterinary Parasitology, School of Veterinary Medicine, Kitasato University, Towada, Aomori, 034-8628, Japan
| | - Yu Kitahara
- Laboratory of Veterinary Parasitology, School of Veterinary Medicine, Kitasato University, Towada, Aomori, 034-8628, Japan
| | - Akira Soga
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada, Obihiro, 080-8555, Japan
| | - Asako Haraguchi
- Laboratory of Veterinary Parasitology, School of Veterinary Medicine, Kitasato University, Towada, Aomori, 034-8628, Japan
| | - Jun Hakozaki
- Laboratory of Veterinary Parasitology, School of Veterinary Medicine, Kitasato University, Towada, Aomori, 034-8628, Japan
| | - Makoto Sugiyama
- Laboratory of Veterinary Anatomy, School of Veterinary Medicine, Kitasato University, Towada, Aomori, 034-8628, Japan
| | - Kodai Kusakisako
- Laboratory of Veterinary Parasitology, School of Veterinary Medicine, Kitasato University, Towada, Aomori, 034-8628, Japan
| | - Shinya Fukumoto
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada, Obihiro, 080-8555, Japan
| | - Hiromi Ikadai
- Laboratory of Veterinary Parasitology, School of Veterinary Medicine, Kitasato University, Towada, Aomori, 034-8628, Japan.
| |
Collapse
|
9
|
A natural symbiotic bacterium drives mosquito refractoriness to Plasmodium infection via secretion of an antimalarial lipase. Nat Microbiol 2021; 6:806-817. [PMID: 33958765 PMCID: PMC9793891 DOI: 10.1038/s41564-021-00899-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 03/29/2021] [Indexed: 02/02/2023]
Abstract
The stalling global progress in the fight against malaria prompts the urgent need to develop new intervention strategies. Whilst engineered symbiotic bacteria have been shown to confer mosquito resistance to parasite infection, a major challenge for field implementation is to address regulatory concerns. Here, we report the identification of a Plasmodium-blocking symbiotic bacterium, Serratia ureilytica Su_YN1, isolated from the midgut of wild Anopheles sinensis in China that inhibits malaria parasites via secretion of an antimalarial lipase. Analysis of Plasmodium vivax epidemic data indicates that local malaria cases in Tengchong (Yunnan province, China) are significantly lower than imported cases and importantly, that the local vector A. sinensis is more resistant to infection by P. vivax than A. sinensis from other regions. Analysis of the gut symbiotic bacteria of mosquitoes from Yunnan province led to the identification of S. ureilytica Su_YN1. This bacterium renders mosquitoes resistant to infection by the human parasite Plasmodium falciparum or the rodent parasite Plasmodium berghei via secretion of a lipase that selectively kills parasites at various stages. Importantly, Su_YN1 rapidly disseminates through mosquito populations by vertical and horizontal transmission, providing a potential tool for blocking malaria transmission in the field.
Collapse
|
10
|
Hixson B, Taracena ML, Buchon N. Midgut Epithelial Dynamics Are Central to Mosquitoes' Physiology and Fitness, and to the Transmission of Vector-Borne Disease. Front Cell Infect Microbiol 2021; 11:653156. [PMID: 33842397 PMCID: PMC8027260 DOI: 10.3389/fcimb.2021.653156] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/23/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Bretta Hixson
- Department of Entomology. Cornell Institute of Host-Microbe Interactions and Disease, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, United States
| | - Mabel Laline Taracena
- Department of Entomology. Cornell Institute of Host-Microbe Interactions and Disease, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, United States
| | - Nicolas Buchon
- Department of Entomology. Cornell Institute of Host-Microbe Interactions and Disease, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, United States
| |
Collapse
|
11
|
Zhou Y, Grieser AM, Do J, Itsara LS, Vaughan AM, Ghosh AK. Purification and production of Plasmodium falciparum zygotes from in vitro culture using magnetic column and Percoll density gradient. Malar J 2020; 19:192. [PMID: 32450861 PMCID: PMC7249376 DOI: 10.1186/s12936-020-03237-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 04/16/2020] [Indexed: 01/20/2023] Open
Abstract
Background Plasmodium falciparum zygotes develop in the mosquito midgut after an infectious blood meal containing mature male and female gametocytes. Studies of mosquito-produced P. falciparum zygotes to elucidate their biology and development have been hampered by high levels of contaminating mosquito proteins and macromolecules present in zygote preparations. Thus, no zygote-specific surface markers have been identified to date. Here, a methodology is developed to obtain large quantities of highly purified zygotes using in vitro culture, including purification methods that include magnetic column cell separation (MACS) followed by Percoll density gradient centrifugation. This straightforward and effective approach provides ample material for studies to enhance understanding of zygote biology and identify novel zygote surface marker candidates that can be tested as transmission blocking vaccine (TBV) candidates. Methods Plasmodium falciparum gametocyte cultures were established and maintained from asexual cultures. Gametocytes were matured for 14 days, then transferred into zygote media for 6 h at 27 ± 2 °C to promote gamete formation and fertilization. Zygotes were then purified using a combination of MACS column separation and Percoll density gradient centrifugation. Purity of the zygotes was determined through morphological studies: the parasite body and nuclear diameter were measured, and zygotes were further transformed into ookinetes. Immunofluorescence assays (IFA) were also performed using the ookinete surface marker, Pfs28. Results After stimulation, the culture consisted of transformed zygotes and a large number of uninfected red blood cells (RBCs), as well as infected RBCs with parasites at earlier developmental stages, including gametes, gametocytes, and asexual stages. The use of two MACS columns removed the vast majority of the RBCs and gametocytes. Subsequent use of two Percoll density gradients enabled isolation of a pure population of zygotes. These zygotes transformed into viable ookinetes that expressed Pfs28. Conclusion The combined approach of using two MACS columns and two Percoll density gradients yielded zygotes with very high purity (45-fold enrichment and a pure population of zygotes [approximately 100%]) that was devoid of contamination by other parasite stages and uninfected RBCs. These enriched zygotes, free from earlier parasites stages and mosquito-derived macromolecules, can be used to further elucidate the biology and developmental processes of Plasmodium.
Collapse
Affiliation(s)
- Yaxian Zhou
- MalarVx, Inc, 1616 Eastlake Ave, E. Suite 285, Seattle, WA, 98102, USA
| | - Alexis M Grieser
- MalarVx, Inc, 1616 Eastlake Ave, E. Suite 285, Seattle, WA, 98102, USA
| | - Julie Do
- MalarVx, Inc, 1616 Eastlake Ave, E. Suite 285, Seattle, WA, 98102, USA
| | - Leslie S Itsara
- MalarVx, Inc, 1616 Eastlake Ave, E. Suite 285, Seattle, WA, 98102, USA
| | - Ashley M Vaughan
- Seattle Children's Research Institute, 307 Westlake Ave, N., Suite 500, Seattle, WA, 98109, USA
| | - Anil K Ghosh
- MalarVx, Inc, 1616 Eastlake Ave, E. Suite 285, Seattle, WA, 98102, USA.
| |
Collapse
|
12
|
|
13
|
Yang J, Schleicher TR, Dong Y, Park HB, Lan J, Cresswell P, Crawford J, Dimopoulos G, Fikrig E. Disruption of mosGILT in Anopheles gambiae impairs ovarian development and Plasmodium infection. J Exp Med 2020; 217:e20190682. [PMID: 31658986 PMCID: PMC7037243 DOI: 10.1084/jem.20190682] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 08/06/2019] [Accepted: 10/25/2019] [Indexed: 11/04/2022] Open
Abstract
Plasmodium infection in Anopheles is influenced by mosquito-derived factors. We previously showed that a protein in saliva from infected Anopheles, mosquito gamma-interferon-inducible lysosomal thiol reductase (mosGILT), inhibits the ability of sporozoites to traverse cells and readily establish infection of the vertebrate host. To determine whether mosGILT influences Plasmodium within the mosquito, we generated Anopheles gambiae mosquitoes carrying mosaic mutations in the mosGILT gene using CRISPR/CRISPR associated protein 9 (Cas9). Here, we show that female mosaic mosGILT mutant mosquitoes display defects in ovarian development and refractoriness to Plasmodium. Following infection by either Plasmodium berghei or Plasmodium falciparum, mutant mosquitoes have significantly reduced oocyst numbers as a result of increased thioester-containing protein 1 (TEP1)-dependent parasite killing. Expression of vitellogenin (Vg), the major yolk protein that can reduce the parasite-killing efficiency of TEP1, is severely impaired in mutant mosquitoes. MosGILT is a mosquito factor that is essential for ovarian development and indirectly protects both human and rodent Plasmodium species from mosquito immunity.
Collapse
Affiliation(s)
- Jing Yang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| | - Tyler R. Schleicher
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| | - Yuemei Dong
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
| | - Hyun Bong Park
- Department of Chemistry, Yale University, New Haven, CT
- Chemical Biology Institute, Yale University, West Haven, CT
| | - Jiangfeng Lan
- College of Fisheries, Huazhong Agricultural University, Wuhan, China
| | - Peter Cresswell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
| | - Jason Crawford
- Department of Chemistry, Yale University, New Haven, CT
- Chemical Biology Institute, Yale University, West Haven, CT
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT
| | - George Dimopoulos
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
- Howard Hughes Medical Institute, Chevy Chase, MD
| |
Collapse
|
14
|
Wells MB, Andrew DJ. Anopheles Salivary Gland Architecture Shapes Plasmodium Sporozoite Availability for Transmission. mBio 2019; 10:e01238-19. [PMID: 31387905 PMCID: PMC6686039 DOI: 10.1128/mbio.01238-19] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 07/11/2019] [Indexed: 12/18/2022] Open
Abstract
Plasmodium sporozoites (SPZs) must traverse the mosquito salivary glands (SGs) to reach a new vertebrate host and continue the malaria disease cycle. Although SGs can harbor thousands of sporozoites, only 10 to 100 are deposited into a host during probing. To determine how the SGs might function as a bottleneck in SPZ transmission, we have characterized Anopheles stephensi SGs infected with the rodent malaria parasite Plasmodium berghei using immunofluorescence confocal microscopy. Our analyses corroborate findings from previous electron microscopy studies and provide new insights into the invasion process. We identified sites of SPZ accumulation within SGs across a range of infection intensities. Although SPZs were most often seen in the distal lateral SG lobes, they were also observed in the medial and proximal lateral lobes. Most parasites were associated with either the basement membrane or secretory cavities. SPZs accumulated at physical barriers, including fused salivary ducts and extensions of the chitinous salivary duct wall into the distal lumen. SPZs were observed only rarely within salivary ducts. SPZs appeared to contact each other in many different quantities, not just in the previously described large bundles. Within parasite bundles, all of the SPZs were oriented in the same direction. We found that moderate levels of infection did not necessarily correlate with major SG disruptions or abundant SG cell death. Altogether, our findings suggest that SG architecture largely acts as a barrier to SPZ transmission.IMPORTANCE Malaria continues to have a devastating impact on human health. With growing resistance to insecticides and antimalarial drugs, as well as climate change predictions indicating expansion of vector territories, the impact of malaria is likely to increase. Additional insights regarding pathogen migration through vector mosquitoes are needed to develop novel methods to prevent transmission to new hosts. Pathogens, including the microbes that cause malaria, must invade the salivary glands (SGs) for transmission. Since SG traversal is required for parasite transmission, SGs are ideal targets for transmission-blocking strategies. The work presented here highlights the role that mosquito SG architecture plays in limiting parasite traversal, revealing how the SG transmission bottleneck is imposed. Further, our data provide unprecedented detail about SG-sporozoite interactions and gland-to-gland variation not provided in previous studies.
Collapse
Affiliation(s)
- Michael B Wells
- Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Deborah J Andrew
- Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
15
|
Bai L, Wang L, Vega-Rodríguez J, Wang G, Wang S. A Gut Symbiotic Bacterium Serratia marcescens Renders Mosquito Resistance to Plasmodium Infection Through Activation of Mosquito Immune Responses. Front Microbiol 2019; 10:1580. [PMID: 31379768 PMCID: PMC6657657 DOI: 10.3389/fmicb.2019.01580] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 06/25/2019] [Indexed: 01/04/2023] Open
Abstract
The malaria development in the mosquito midgut is a complex process that results in considerable parasite losses. The mosquito gut microbiota influences the outcome of pathogen infection in mosquitoes, but the underlying mechanisms through which gut symbiotic bacteria affect vector competence remain elusive. Here, we identified two Serratia strains (Y1 and J1) isolated from field-caught female Anopheles sinensis from China and assessed their effect on Plasmodium development in An. stephensi. Colonization of An. stephensi midgut by Serratia Y1 significantly renders the mosquito resistant to Plasmodium berghei infection, while Serratia J1 has no impact on parasite development. Parasite inhibition by Serratia Y1 is induced by the activation of the mosquito immune system. Genome-wide transcriptomic analysis by RNA-seq shows a similar pattern of midgut gene expression in response to Serratia Y1 and J1 in sugar-fed mosquitoes. However, 24 h after blood ingestion, Serratia Y1 modulates more midgut genes than Serratia J1 including the c-type lectins (CTLs), CLIP serine proteases and other immune effectors. Furthermore, silencing of several Serratia Y1-induced anti-Plasmodium factors like the thioester-containing protein 1 (TEP1), fibrinogen immunolectin 9 (FBN9) or leucine-rich repeat protein LRRD7 can rescue parasite oocyst development in the presence of Serratia Y1, suggesting that these factors modulate the Serratia Y1-mediated anti-Plasmodium effect. This study enhances our understanding of how gut bacteria influence mosquito-Plasmodium interactions.
Collapse
Affiliation(s)
- Liang Bai
- School of Life Science and Technology, Tongji University, Shanghai, China.,CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Lili Wang
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Joel Vega-Rodríguez
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Guandong Wang
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Sibao Wang
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
16
|
Zhu SJ, Hendry JA, Almagro-Garcia J, Pearson RD, Amato R, Miles A, Weiss DJ, Lucas TC, Nguyen M, Gething PW, Kwiatkowski D, McVean G. The origins and relatedness structure of mixed infections vary with local prevalence of P. falciparum malaria. eLife 2019; 8:e40845. [PMID: 31298657 PMCID: PMC6684230 DOI: 10.7554/elife.40845] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 07/10/2019] [Indexed: 02/07/2023] Open
Abstract
Individual malaria infections can carry multiple strains of Plasmodium falciparum with varying levels of relatedness. Yet, how local epidemiology affects the properties of such mixed infections remains unclear. Here, we develop an enhanced method for strain deconvolution from genome sequencing data, which estimates the number of strains, their proportions, identity-by-descent (IBD) profiles and individual haplotypes. Applying it to the Pf3k data set, we find that the rate of mixed infection varies from 29% to 63% across countries and that 51% of mixed infections involve more than two strains. Furthermore, we estimate that 47% of symptomatic dual infections contain sibling strains likely to have been co-transmitted from a single mosquito, and find evidence of mixed infections propagated over successive infection cycles. Finally, leveraging data from the Malaria Atlas Project, we find that prevalence correlates within Africa, but not Asia, with both the rate of mixed infection and the level of IBD.
Collapse
Affiliation(s)
- Sha Joe Zhu
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, United Kingdom
| | - Jason A Hendry
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, United Kingdom
| | - Jacob Almagro-Garcia
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, United Kingdom
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Medical Research Council Centre for Genomics and Global Health, University of Oxford, Oxford, United Kingdom
- Wellcome Sanger Institute, Hinxton, United Kingdom
| | - Richard D Pearson
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Medical Research Council Centre for Genomics and Global Health, University of Oxford, Oxford, United Kingdom
- Wellcome Sanger Institute, Hinxton, United Kingdom
| | - Roberto Amato
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Medical Research Council Centre for Genomics and Global Health, University of Oxford, Oxford, United Kingdom
- Wellcome Sanger Institute, Hinxton, United Kingdom
| | - Alistair Miles
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, United Kingdom
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Medical Research Council Centre for Genomics and Global Health, University of Oxford, Oxford, United Kingdom
- Wellcome Sanger Institute, Hinxton, United Kingdom
| | - Daniel J Weiss
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, United Kingdom
| | - Tim Cd Lucas
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, United Kingdom
| | - Michele Nguyen
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, United Kingdom
| | - Peter W Gething
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, United Kingdom
| | - Dominic Kwiatkowski
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, United Kingdom
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Medical Research Council Centre for Genomics and Global Health, University of Oxford, Oxford, United Kingdom
- Wellcome Sanger Institute, Hinxton, United Kingdom
| | - Gil McVean
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, United Kingdom
- Medical Research Council Centre for Genomics and Global Health, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
17
|
Baia-da-Silva DC, Orfanó AS, Nacif-Pimenta R, de Melo FF, Simões S, Cabral I, Lacerda MVG, Guerra MDGB, Monteiro WM, Secundino NFC, Pimenta PFP. The Midgut Muscle Network of Anopheles aquasalis (Culicidae, Anophelinae): Microanatomy and Structural Modification After Blood Meal and Plasmodium vivax (Haemosporida, Plasmodiidae) Infection. JOURNAL OF MEDICAL ENTOMOLOGY 2019; 56:421-431. [PMID: 30508123 DOI: 10.1093/jme/tjy199] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Indexed: 06/09/2023]
Abstract
The mosquito midgut is divided into two regions named anterior midgut (AMG) and posterior midgut (PMG). The midgut expands intensely after the blood ingestion to accommodate a large amount of ingested food. To efficiently support the bloodmeal-induced changes, the organization of the visceral muscle fibers has significant adjustments. This study describes the spatial organization of the Anopheles aquasalis (Culicidae, Anophelinae) midgut muscle network and morphological changes after bloodmeal ingestion and infection with Plasmodium vivax (Haemosporida, Plasmodiidae). The midgut muscle network is composed of two types of fibers: longitudinal and circular. The two types of muscle fibers are composed of thick and thin filaments, similar to myosin and actin, respectively. Invagination of sarcoplasm membrane forms the T-system tubules. Sarcoplasmic reticulum cisternae have been observed in association with these invaginations. At different times after the bloodmeal, the fibers in the AMG are not modified. A remarkable dilation characterizes the transitional area between the AMG and the PMG. In the PMG surface, after the completion of bloodmeal ingestion, the stretched muscle fibers became discontinued. At 72 h after bloodmeal digestion, it is possible to observe the presence of disorganized muscle fibers in the midgut regions. The Plasmodium oocyst development along the basal layer of the midgut does not have a significant role in the visceral musculature distribution. This study provides features of the visceral musculature at different blood feeding times of An. aquasalis and shows important changes in midgut topography including when the mosquitoes are infected with P. vivax.
Collapse
Affiliation(s)
- Djane C Baia-da-Silva
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, PMG, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, PMG, Brazil
| | - Alessandra S Orfanó
- Instituto de Pesquisas René Rachou, Fundação Oswaldo Cruz-Minas Gerais, Barro Preto, Belo Horizonte, MG, Brazil
| | - Rafael Nacif-Pimenta
- Instituto de Pesquisas René Rachou, Fundação Oswaldo Cruz-Minas Gerais, Barro Preto, Belo Horizonte, MG, Brazil
| | - Fabrício Freire de Melo
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, BA, Brazil
| | - Suzan Simões
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, PMG, Brazil
| | - Iria Cabral
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, PMG, Brazil
| | - Marcus Vinicíus Guimarães Lacerda
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, PMG, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, PMG, Brazil
- Instituto Leônidas & Maria Deane, Fundação Oswaldo Cruz - Manaus, Manaus, AM, Brazil
| | - Maria das Graças Barbosa Guerra
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, PMG, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, PMG, Brazil
| | - Wuelton M Monteiro
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, PMG, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, PMG, Brazil
| | - Nagila F C Secundino
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, PMG, Brazil
- Instituto de Pesquisas René Rachou, Fundação Oswaldo Cruz-Minas Gerais, Barro Preto, Belo Horizonte, MG, Brazil
| | - Paulo F P Pimenta
- Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, PMG, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, PMG, Brazil
- Instituto de Pesquisas René Rachou, Fundação Oswaldo Cruz-Minas Gerais, Barro Preto, Belo Horizonte, MG, Brazil
| |
Collapse
|
18
|
para-Aminobenzoate Synthesis versus Salvage in Malaria Parasites. Trends Parasitol 2019; 35:176-178. [PMID: 30709568 DOI: 10.1016/j.pt.2019.01.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 01/18/2019] [Indexed: 11/20/2022]
Abstract
Enzymes of the folate de novo synthesis pathway in malaria parasites are proven antimalarial drug targets. A key precursor for folate synthesis is para-aminobenzoate (pABA). In a recent study [1] (Cell Rep. 2019;26:356-363 e4), the contributions of pABA synthesis versus salvage were re-evaluated in a rodent malaria model with knockout parasites grown in mice fed with various diets. The results imply that malaria parasites can either synthesize or salvage pABA to meet the demand for folates.
Collapse
|
19
|
Dong Y, Simões ML, Marois E, Dimopoulos G. CRISPR/Cas9 -mediated gene knockout of Anopheles gambiae FREP1 suppresses malaria parasite infection. PLoS Pathog 2018. [PMID: 29518156 PMCID: PMC5843335 DOI: 10.1371/journal.ppat.1006898] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Plasmodium relies on numerous agonists during its journey through the mosquito vector, and these agonists represent potent targets for transmission-blocking by either inhibiting or interfering with them pre- or post-transcriptionally. The recently developed CRISPR/Cas9-based genome editing tools for Anopheles mosquitoes provide new and promising opportunities for the study of agonist function and for developing malaria control strategies through gene deletion to achieve complete agonist inactivation. Here we have established a modified CRISPR/Cas9 gene editing procedure for the malaria vector Anopheles gambiae, and studied the effect of inactivating the fibrinogen-related protein 1 (FREP1) gene on the mosquito’s susceptibility to Plasmodium and on mosquito fitness. FREP1 knockout mutants developed into adult mosquitoes that showed profound suppression of infection with both human and rodent malaria parasites at the oocyst and sporozoite stages. FREP1 inactivation, however, resulted in fitness costs including a significantly lower blood-feeding propensity, fecundity and egg hatching rate, a retarded pupation time, and reduced longevity after a blood meal. The causative agent of malaria, Plasmodium, has to complete a complex infection cycle in the Anopheles gambiae mosquito vector in order to reach the salivary gland from where it can be transmitted to a human host. The parasite’s development in the mosquito relies on numerous host factors (agonists), and their inhibition or inactivation can thereby result in suppression of infection and consequently malaria transmission. The recently developed CRISPR/Cas9-based genome editing tools for Anopheles mosquitoes provide new and promising opportunities to delete (inactivate) Plasmodium agonists to better understand their function and for blocking malaria transmission. Here we have established a modified CRISPR/Cas9 genome editing technique for malaria vector A. gambiae mosquitoes. Through this approach we have inactivated the fibrinogen-related protein 1 (FREP1) gene, via CRISPR/Cas9 genome editing, and the impact of this manipulation on the mosquito’s susceptibility to Plasmodium and on mosquito fitness. FREP1 knockout mutants showed a profound suppression of infection with both human and rodent malaria parasites, while it also resulted in fitness costs: a significantly lower blood-feeding propensity, fecundity and egg hatching rate, and a retarded larval development and pupation time, and reduced longevity after a blood meal.
Collapse
Affiliation(s)
- Yuemei Dong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Malaria Research Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Maria L. Simões
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Malaria Research Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Eric Marois
- Inserm, CNRS, Université de Strasbourg, Strasbourg, France
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Malaria Research Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
20
|
Yamamoto DS, Sumitani M, Hatakeyama M, Matsuoka H. Malaria infectivity of xanthurenic acid-deficient anopheline mosquitoes produced by TALEN-mediated targeted mutagenesis. Transgenic Res 2018; 27:51-60. [DOI: 10.1007/s11248-018-0057-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 01/11/2018] [Indexed: 12/15/2022]
|
21
|
Sasaki H, Sekiguchi H, Sugiyama M, Ikadai H. Plasmodium berghei Cap93, a novel oocyst capsule-associated protein, plays a role in sporozoite development. Parasit Vectors 2017; 10:399. [PMID: 28841886 PMCID: PMC5574095 DOI: 10.1186/s13071-017-2337-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 08/16/2017] [Indexed: 11/20/2022] Open
Affiliation(s)
- Hanae Sasaki
- Hokusan Co. Ltd., 27-4, Kitanosato, Kitahiroshima, Hokkaido, 061-111, Japan
| | - Harumi Sekiguchi
- Laboratory of Veterinary Parasitology, School of Veterinary Medicine, Kitasato University, Towada, Aomori, 034-8628, Japan
| | - Makoto Sugiyama
- Laboratory of Veterinary Anatomy, School of Veterinary Medicine, Kitasato University, Towada, Aomori, 034-8628, Japan
| | - Hiromi Ikadai
- Laboratory of Veterinary Parasitology, School of Veterinary Medicine, Kitasato University, Towada, Aomori, 034-8628, Japan.
| |
Collapse
|
22
|
Domingos A, Pinheiro-Silva R, Couto J, do Rosário V, de la Fuente J. The Anopheles gambiae transcriptome - a turning point for malaria control. INSECT MOLECULAR BIOLOGY 2017; 26:140-151. [PMID: 28067439 DOI: 10.1111/imb.12289] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Mosquitoes are important vectors of several pathogens and thereby contribute to the spread of diseases, with social, economic and public health impacts. Amongst the approximately 450 species of Anopheles, about 60 are recognized as vectors of human malaria, the most important parasitic disease. In Africa, Anopheles gambiae is the main malaria vector mosquito. Current malaria control strategies are largely focused on drugs and vector control measures such as insecticides and bed-nets. Improvement of current, and the development of new, mosquito-targeted malaria control methods rely on a better understanding of mosquito vector biology. An organism's transcriptome is a reflection of its physiological state and transcriptomic analyses of different conditions that are relevant to mosquito vector competence can therefore yield important information. Transcriptomic analyses have contributed significant information on processes such as blood-feeding parasite-vector interaction, insecticide resistance, and tissue- and stage-specific gene regulation, thereby facilitating the path towards the development of new malaria control methods. Here, we discuss the main applications of transcriptomic analyses in An. gambiae that have led to a better understanding of mosquito vector competence.
Collapse
Affiliation(s)
- A Domingos
- Instituto de Higiene e Medicina Tropical (IHMT), Lisboa, Portugal
- Global Health and Tropical Medicine (GHMT), Instituto de Higiene e Medicina Tropical (IHMT), Lisboa, Portugal
| | - R Pinheiro-Silva
- Instituto de Higiene e Medicina Tropical (IHMT), Lisboa, Portugal
| | - J Couto
- Instituto de Higiene e Medicina Tropical (IHMT), Lisboa, Portugal
| | - V do Rosário
- Instituto de Higiene e Medicina Tropical (IHMT), Lisboa, Portugal
| | - J de la Fuente
- SaBio. Instituto de Investigación de Recursos Cinegéticos, IREC-CSIC-UCLM-JCCM, Ciudad Real, Spain
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, USA
| |
Collapse
|
23
|
Salivary Gland Proteome during Adult Development and after Blood Feeding of Female Anopheles dissidens Mosquitoes (Diptera: Culicidae). PLoS One 2016; 11:e0163810. [PMID: 27669021 PMCID: PMC5036837 DOI: 10.1371/journal.pone.0163810] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 09/14/2016] [Indexed: 12/28/2022] Open
Abstract
Understanding changes in mosquito salivary proteins during the time that sporozoite maturation occurs and after blood feeding may give information regarding the roles of salivary proteins during the malarial transmission. Anopheles dissidens (formerly Anopheles barbirostris species A1) is a potential vector of Plasmodium vivax in Thailand. In this study, analyses of the proteomic profiles of female An. dissidens salivary glands during adult development and after blood feeding were carried out using two-dimensional gel electrophoresis coupled with nano-liquid chromatography-mass spectrometry. Results showed at least 17 major salivary gland proteins present from day one to day 21 post emergence at 8 different time points sampled. Although there was variation observed, the patterns of protein expression could be placed into one of four groups. Fifteen protein spots showed significant depletion after blood feeding with the percentages of the amount of depletion ranging from 8.5% to 68.11%. The overall results identified various proteins, including a putative mucin-like protein, an anti-platelet protein, a long form D7 salivary protein, a putative gVAG protein precursor, a D7-related 3.2 protein, gSG7 salivary proteins, and a gSG6 protein. These results allow better understanding of the changes of the salivary proteins during the adult mosquito development. They also provide candidate proteins to investigate any possible link or not between sporozoite maturation, or survival of skin stage sporozoites, and salivary proteins.
Collapse
|
24
|
Yamamoto DS, Sumitani M, Kasashima K, Sezutsu H, Matsuoka H. Inhibition of Malaria Infection in Transgenic Anopheline Mosquitoes Lacking Salivary Gland Cells. PLoS Pathog 2016; 12:e1005872. [PMID: 27598328 PMCID: PMC5012584 DOI: 10.1371/journal.ppat.1005872] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 08/15/2016] [Indexed: 01/24/2023] Open
Abstract
Malaria is an important global public health challenge, and is transmitted by anopheline mosquitoes during blood feeding. Mosquito vector control is one of the most effective methods to control malaria, and population replacement with genetically engineered mosquitoes to block its transmission is expected to become a new vector control strategy. The salivary glands are an effective target tissue for the expression of molecules that kill or inactivate malaria parasites. Moreover, salivary gland cells express a large number of molecules that facilitate blood feeding and parasite transmission to hosts. In the present study, we adapted a functional deficiency system in specific tissues by inducing cell death using the mouse Bcl-2-associated X protein (Bax) to the Asian malaria vector mosquito, Anopheles stephensi. We applied this technique to salivary gland cells, and produced a transgenic strain containing extremely low amounts of saliva. Although probing times for feeding on mice were longer in transgenic mosquitoes than in wild-type mosquitoes, transgenic mosquitoes still successfully ingested blood. Transgenic mosquitoes also exhibited a significant reduction in oocyst formation in the midgut in a rodent malaria model. These results indicate that mosquito saliva plays an important role in malaria infection in the midgut of anopheline mosquitoes. The dysfunction in the salivary glands enabled the inhibition of malaria transmission from hosts to mosquito midguts. Therefore, salivary components have potential in the development of new drugs or genetically engineered mosquitoes for malaria control. Malaria, transmitted by anopheline mosquitoes, represents an important global public health challenge. The salivary glands of mosquitoes are an attractive target tissue for malaria and vector control. We produced a transgenic strain inducing cell death in the salivary glands with a cell death effector molecule in the Asian malaria vector mosquito, Anopheles stephensi. This transgenic strain contained extremely low amounts of saliva. An analysis of this strain revealed that saliva plays an important role in probing as well as malaria infection in the midgut in a rodent malaria model. The dysfunction in the salivary glands enabled the inhibition of malaria transmission to mosquito midguts. Therefore, salivary components are also important in malaria control.
Collapse
Affiliation(s)
- Daisuke S. Yamamoto
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Yakushiji, Shimotsuke, Tochigi, Japan
- * E-mail:
| | - Megumi Sumitani
- Transgenic Silkworm Research Unit, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Owashi, Tsukuba, Ibaraki, Japan
| | - Katsumi Kasashima
- Division of Functional Biochemistry, Department of Biochemistry, Jichi Medical University, Yakushiji, Shimotsuke, Tochigi, Japan
| | - Hideki Sezutsu
- Transgenic Silkworm Research Unit, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Owashi, Tsukuba, Ibaraki, Japan
| | - Hiroyuki Matsuoka
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Yakushiji, Shimotsuke, Tochigi, Japan
| |
Collapse
|
25
|
Kajla M, Choudhury TP, Kakani P, Gupta K, Dhawan R, Gupta L, Kumar S. Silencing of Anopheles stephensi Heme Peroxidase HPX15 Activates Diverse Immune Pathways to Regulate the Growth of Midgut Bacteria. Front Microbiol 2016; 7:1351. [PMID: 27630620 PMCID: PMC5006007 DOI: 10.3389/fmicb.2016.01351] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 08/16/2016] [Indexed: 11/20/2022] Open
Abstract
Anopheles mosquito midgut harbors a diverse group of endogenous bacteria that grow extensively after the blood feeding and help in food digestion and nutrition in many ways. Although, the growth of endogenous bacteria is regulated by various factors, however, the robust antibacterial immune reactions are generally suppressed in this body compartment by a heme peroxidase HPX15 crosslinked mucins barrier. This barrier is formed on the luminal side of the midgut and blocks the direct interactions and recognition of bacteria or their elicitors by the immune reactive midgut epithelium. We hypothesized that in the absence of HPX15, an increased load of exogenous bacteria will enormously induce the mosquito midgut immunity and this situation in turn, can easily regulate mosquito-pathogen interactions. In this study, we found that the blood feeding induced AsHPX15 gene in Anopheles stephensi midgut and promoted the growth of endogenous as well as exogenous fed bacteria. In addition, the mosquito midgut also efficiently regulated the number of these bacteria through the induction of classical Toll and Imd immune pathways. In case of AsHPX15 silenced midguts, the growth of midgut bacteria was largely reduced through the induction of nitric oxide synthase (NOS) gene, a downstream effector molecule of the JAK/STAT pathway. Interestingly, no significant induction of the classical immune pathways was observed in these midguts. Importantly, the NOS is a well known negative regulator of Plasmodium development, thus, we proposed that the induction of diverged immune pathways in the absence of HPX15 mediated midgut barrier might be one of the strategies to manipulate the vectorial capacity of Anopheles mosquito.
Collapse
Affiliation(s)
- Mithilesh Kajla
- Molecular Parasitology and Vector Biology Laboratory, Department of Biological Sciences, Birla Institute of Technology and Science Pilani, India
| | - Tania P Choudhury
- Molecular Parasitology and Vector Biology Laboratory, Department of Biological Sciences, Birla Institute of Technology and Science Pilani, India
| | - Parik Kakani
- Molecular Parasitology and Vector Biology Laboratory, Department of Biological Sciences, Birla Institute of Technology and Science Pilani, India
| | - Kuldeep Gupta
- Molecular Parasitology and Vector Biology Laboratory, Department of Biological Sciences, Birla Institute of Technology and Science Pilani, India
| | - Rini Dhawan
- Molecular Parasitology and Vector Biology Laboratory, Department of Biological Sciences, Birla Institute of Technology and Science Pilani, India
| | - Lalita Gupta
- Molecular Parasitology and Vector Biology Laboratory, Department of Biological Sciences, Birla Institute of Technology and SciencePilani, India; Department of Zoology, Chaudhary Bansi Lal UniversityBhiwani, India
| | - Sanjeev Kumar
- Molecular Parasitology and Vector Biology Laboratory, Department of Biological Sciences, Birla Institute of Technology and SciencePilani, India; Department of Biotechnology, Chaudhary Bansi Lal UniversityBhiwani, India
| |
Collapse
|
26
|
Species-specific escape of Plasmodium sporozoites from oocysts of avian, rodent, and human malarial parasites. Malar J 2016; 15:394. [PMID: 27480269 PMCID: PMC4969971 DOI: 10.1186/s12936-016-1451-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 07/21/2016] [Indexed: 12/24/2022] Open
Abstract
Background Malaria is transmitted when an infected mosquito delivers Plasmodium sporozoites into a vertebrate host. There are many species of Plasmodium and, in general, the infection is host-specific. For example, Plasmodium gallinaceum is an avian parasite, while Plasmodium berghei infects mice. These two parasites have been extensively used as experimental models of malaria transmission. Plasmodium falciparum and Plasmodium vivax are the most important agents of human malaria, a life-threatening disease of global importance. To complete their life cycle, Plasmodium parasites must traverse the mosquito midgut and form an oocyst that will divide continuously. Mature oocysts release thousands of sporozoites into the mosquito haemolymph that must reach the salivary gland to infect a new vertebrate host. The current understanding of the biology of oocyst formation and sporozoite release is mostly based on experimental infections with P.berghei, and the conclusions are generalized to other Plasmodium species that infect humans without further morphological analyses. Results Here, it is described the microanatomy of sporozoite escape from oocysts of four Plasmodium species: the two laboratory models, P. gallinaceum and P. berghei, and the two main species that cause malaria in humans, P.vivax and P. falciparum. It was found that sporozoites have species-specific mechanisms of escape from the oocyst. The two model species of Plasmodium had a common mechanism, in which the oocyst wall breaks down before sporozoites emerge. In contrast, P. vivax and P. falciparum sporozoites show a dynamic escape mechanism from the oocyst via polarized propulsion. Conclusions This study demonstrated that Plasmodium species do not share a common mechanism of sporozoite escape, as previously thought, but show complex and species-specific mechanisms. In addition, the knowledge of this phenomenon in human Plasmodium can facilitate transmission-blocking studies and not those ones only based on the murine and avian models.
Collapse
|
27
|
Lecona-Valera AN, Tao D, Rodríguez MH, López T, Dinglasan RR, Rodríguez MC. An antibody against an Anopheles albimanus midgut myosin reduces Plasmodium berghei oocyst development. Parasit Vectors 2016; 9:274. [PMID: 27165123 PMCID: PMC4863318 DOI: 10.1186/s13071-016-1548-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 04/27/2016] [Indexed: 11/13/2022] Open
Abstract
Background Malaria parasites are transmitted by Anopheles mosquitoes. Although several studies have identified mosquito midgut surface proteins that are putatively important for Plasmodium ookinete invasion, only a few have characterized these protein targets and demonstrated transmission-blocking activity. Molecular information about these proteins is essential for the development of transmission-blocking vaccines (TBV). The aim of the present study was to test three monoclonal antibodies (mAbs), A-140, A-78 and A-10, for their ability to recognize antigens and block oocyst infection of the midgut of Anopheles albimanus, a major malaria vector in Latin America. Method Western-blot of mAbs on antigens from midgut brush border membrane vesicles was used to select antibodies. Three mAbs were tested by membrane feeding assays to evaluate their potential transmission-blocking activity against Plasmodium berghei. The cognate antigens recognized by mAbs with oocyst-reducing activity were determined by immunoprecipitation followed by liquid chromatography tandem mass spectrometry. Results Only one mAb, A-140, significantly reduced oocyst infection intensity. Hence, its probable protein target in the Anopheles albimanus midgut was identified and characterized. It recognized three high-molecular mass proteins from a midgut brush border microvilli vesicle preparation. Chemical deglycosylation assays confirmed the peptide nature of the epitope recognized by mAb A-140. Immunoprecipitation followed by proteomic identification with tandem mass spectrometry revealed five proteins, presumably extracted together as a complex. Of these, AALB007909 had the highest mascot score and corresponds to a protein with a myosin head motor domain, indicating that the target of mAb A-140 is probably myosin located on the microvilli of the mosquito midgut. Conclusion These results provide support for the participation of myosin in mosquito midgut invasion by Plasmodium ookinetes. The potential inclusion of this protein in the design of new multivalent vaccine strategies for blocking Plasmodium transmission is discussed. Electronic supplementary material The online version of this article (doi:10.1186/s13071-016-1548-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alba N Lecona-Valera
- Center of Research on Infectious Diseases, National Institute of Public Health, Av. Universidad 655, Col. Santa María Ahuacatitlán, Cuernavaca, Morelos, C. P. 62508, Mexico
| | - Dingyin Tao
- W. Harry Feinstone Department of Molecular Microbiology & Immunology and the Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, Maryland, 21205, USA
| | - Mario H Rodríguez
- Center of Research on Infectious Diseases, National Institute of Public Health, Av. Universidad 655, Col. Santa María Ahuacatitlán, Cuernavaca, Morelos, C. P. 62508, Mexico
| | - Tomás López
- Instituto de Biotecnología, Universidad Nacional Autónoma de Méxic006F, Av. Universidad 2001, Colonia Chamilpa, Cuernavaca, Morelos, 62210, Mexico
| | - Rhoel R Dinglasan
- W. Harry Feinstone Department of Molecular Microbiology & Immunology and the Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, Maryland, 21205, USA
| | - María C Rodríguez
- Center of Research on Infectious Diseases, National Institute of Public Health, Av. Universidad 655, Col. Santa María Ahuacatitlán, Cuernavaca, Morelos, C. P. 62508, Mexico.
| |
Collapse
|
28
|
Habtewold T, Groom Z, Duchateau L, Christophides GK. Detection of viable plasmodium ookinetes in the midguts of anopheles coluzzi using PMA-qrtPCR. Parasit Vectors 2015; 8:455. [PMID: 26373633 PMCID: PMC4572643 DOI: 10.1186/s13071-015-1087-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 09/11/2015] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Mosquito infection with malaria parasites depends on complex interactions between the mosquito immune response, the parasite developmental program and the midgut microbiota. Simultaneous monitoring of the parasite and bacterial dynamics is important when studying these interactions. PCR based methods of genomic DNA (gDNA) have been widely used, but their inability to discriminate between live and dead cells compromises their application. The alternative method of quantification of mRNA mainly reports on cell activity rather than density. METHOD Quantitative real-time (qrt) PCR in combination with Propidium Monoazide (PMA) treatment (PMA-qrtPCR) has been previously used for selectively enumerating viable microbial cells. PMA penetrates damaged cell membranes and intercalates in the DNA inhibiting its PCR amplification. Here, we tested the potential of PMA-qrtPCR to discriminate between and quantify live and dead Plasmodium berghei malarial parasites and commensal bacteria in the midgut of Anopheles coluzzii Coetzee & Wilkerson 2013 (formerly An. gambiae M-form). RESULTS By combining microscopic observations with reverse transcriptase PCR (RT-PCR) we reveal that, in addition to gDNA, mRNA from dead parasites also persists inside the mosquito midgut, therefore its quantification cannot accurately reflect live-only parasites at the time of monitoring. In contrast, pre-treating the samples with PMA selectively inhibited qrtPCR amplification of parasite gDNA, with about 15 cycles (Ct-value) difference between PMA-treated and control samples. The limit of detection corresponds to 10 Plasmodium ookinetes. Finally, we show that the PMA-qrtPCR method can be used to quantify bacteria that are present in the mosquito midgut. CONCLUSION The PMA-qrtPCR is a suitable method for quantification of viable parasites and bacteria in the midgut of Anopheles mosquitoes. The method will be valuable when studying the molecular interactions between the mosquito, the malaria parasite and midgut microbiota.
Collapse
Affiliation(s)
- Tibebu Habtewold
- Department of Life Sciences, Imperial College London, London, UK.
- Department of Comparative Physiology and Biometrics, University of Ghent, Ghent, Belgium.
| | - Zoe Groom
- Department of Life Sciences, Imperial College London, London, UK
- Costello Medical Consulting, Cambridge, UK
| | - Luc Duchateau
- Department of Comparative Physiology and Biometrics, University of Ghent, Ghent, Belgium
| | | |
Collapse
|
29
|
Valkiūnas G, Iezhova TA, Palinauskas V, Ilgūnas M, Bernotienė R. The evidence for rapid gametocyte viability changes in the course of parasitemia in Haemoproteus parasites. Parasitol Res 2015; 114:2903-9. [PMID: 25944737 DOI: 10.1007/s00436-015-4491-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 04/20/2015] [Indexed: 11/29/2022]
Abstract
Avian haemosporidian parasites of the genus Haemoproteus (Haemoproteidae, Haemosporida) are widespread, and some species cause diseases both in vertebrate hosts and blood-sucking insects. Parasitemia of Haemoproteus species usually is long-lasting, with gametocytes present in the circulation for several months. However, the viability of gametocytes and their ability to produce sexual cells have been insufficiently understood in the course of parasitemia. We initiated the sexual development in vitro conditions and calculated proportions of normal and anomalous ookinetes, which developed in two species of Haemoproteus. Mature gametocytes of the parasites were obtained from naturally infected avian hosts at different days of parasitemia. Haemoproteus (Parahaemoproteus) lanii (cytochrome b lineage hRB1) was isolated from one red-backed shrike Lanius collurio. Two isolates of Haemoproteus (Parahaemoproteus) tartakovskyi (cytochrome b lineage hSISKIN1) were used: one was obtained from a siskin Carduelis spinus and one from a common crossbill Loxia curvirostra. The wild-caught birds were kept indoors under controlled conditions, and blood was taken from them every 1 or 2 days during 10-14 days. After each blood sampling, the sexual process and ookinete development were initiated in vitro by exposure of infected blood containing mature gametocytes to air. Smears were prepared at intervals of 15 min, 3 h, and 12 h after the exposure; they were examined microscopically. In all, 25 experiments were performed; each experiment was repeated two times. The ratios of macro- and microgametocytes did not change in all experimental infections during this study. Sexual process occurred, and both normal and anomalous ookinetes developed in all parasites. The proportion of normal ookinetes did not change significantly in both isolates of H. tartakovskyi. Between 8 and 10 days of observation, the proportion of normal ookinetes of H. lanii decreased 6 times compared to the beginning of the experiment. That was accompanied with the rapid decrease of parasitemia and the inability of the majority of mature gametocytes to escape from erythrocytes and produce gametes, indicating disorder of the gametogenesis. There was clear difference in the gametogenesis between H. tartakovskyi and H. lanii from this point of view. This study shows that the viability of Haemoproteus gametocytes might change dramatically in the course of parasitemia within 1-2 days, and the presence of mature gametocytes in the circulation does not necessarily indicate their ability to exflagellate and produce ookinetes. We predict that this finding is important epidemiologically due to relationship with sporogony success.
Collapse
|
30
|
Pimenta PFP, Orfano AS, Bahia AC, Duarte APM, Ríos-Velásquez CM, Melo FF, Pessoa FAC, Oliveira GA, Campos KMM, Villegas LM, Rodrigues NB, Nacif-Pimenta R, Simões RC, Monteiro WM, Amino R, Traub-Cseko YM, Lima JBP, Barbosa MGV, Lacerda MVG, Tadei WP, Secundino NFC. An overview of malaria transmission from the perspective of Amazon Anopheles vectors. Mem Inst Oswaldo Cruz 2015; 110:23-47. [PMID: 25742262 PMCID: PMC4371216 DOI: 10.1590/0074-02760140266] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 12/18/2014] [Indexed: 02/07/2023] Open
Abstract
In the Americas, areas with a high risk of malaria transmission are mainly located in the Amazon Forest, which extends across nine countries. One keystone step to understanding the Plasmodium life cycle in Anopheles species from the Amazon Region is to obtain experimentally infected mosquito vectors. Several attempts to colonise Anopheles species have been conducted, but with only short-lived success or no success at all. In this review, we review the literature on malaria transmission from the perspective of its Amazon vectors. Currently, it is possible to develop experimental Plasmodium vivax infection of the colonised and field-captured vectors in laboratories located close to Amazonian endemic areas. We are also reviewing studies related to the immune response to P. vivax infection of Anopheles aquasalis, a coastal mosquito species. Finally, we discuss the importance of the modulation of Plasmodium infection by the vector microbiota and also consider the anopheline genomes. The establishment of experimental mosquito infections with Plasmodium falciparum, Plasmodium yoelii and Plasmodium berghei parasites that could provide interesting models for studying malaria in the Amazonian scenario is important. Understanding the molecular mechanisms involved in the development of the parasites in New World vectors is crucial in order to better determine the interaction process and vectorial competence.
Collapse
Affiliation(s)
- Paulo FP Pimenta
- Centro de Pesquisas René Rachou-Fiocruz, Belo Horizonte, MG,
Brasil
- Fundação de Medicina Tropical Dr Heitor Vieira Dourado, Manaus, AM,
Brasil
| | | | - Ana C Bahia
- Instituto Oswaldo Cruz-Fiocruz, Rio de Janeiro, RJ, Brasil
| | - Ana PM Duarte
- Centro de Pesquisas René Rachou-Fiocruz, Belo Horizonte, MG,
Brasil
| | | | - Fabrício F Melo
- Centro de Pesquisas René Rachou-Fiocruz, Belo Horizonte, MG,
Brasil
| | | | | | - Keillen MM Campos
- Fundação de Medicina Tropical Dr Heitor Vieira Dourado, Manaus, AM,
Brasil
| | | | | | | | - Rejane C Simões
- Instituto Nacional de Pesquisas da Amazônia, Manaus, AM, Brasil
| | - Wuelton M Monteiro
- Fundação de Medicina Tropical Dr Heitor Vieira Dourado, Manaus, AM,
Brasil
| | - Rogerio Amino
- Unité de Biologie et Génétique du Paludisme, Institut Pasteur, Paris,
France
| | | | - José BP Lima
- Fundação de Medicina Tropical Dr Heitor Vieira Dourado, Manaus, AM,
Brasil
- Instituto Oswaldo Cruz-Fiocruz, Rio de Janeiro, RJ, Brasil
| | - Maria GV Barbosa
- Fundação de Medicina Tropical Dr Heitor Vieira Dourado, Manaus, AM,
Brasil
| | - Marcus VG Lacerda
- Fundação de Medicina Tropical Dr Heitor Vieira Dourado, Manaus, AM,
Brasil
- Instituto Leônidas e Maria Deane-Fiocruz, Manaus, AM, Brasil
| | | | | |
Collapse
|
31
|
Chaitanya RK, Sridevi P, Kumar KS, Mastan BS, Kumar KA, Dutta-Gupta A. Expression analysis of reactive oxygen species detoxifying enzyme genes in Anopheles stephensi during Plasmodium berghei midgut invasion. ASIAN PAC J TROP MED 2014. [DOI: 10.1016/s1995-7645(14)60116-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
32
|
Garcia-Longoria L, Hellgren O, Bensch S. Molecular identification of the chitinase genes in Plasmodium relictum. Malar J 2014; 13:239. [PMID: 24943514 PMCID: PMC4072489 DOI: 10.1186/1475-2875-13-239] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 06/07/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Malaria parasites need to synthesize chitinase in order to go through the peritrophic membrane, which is created around the mosquito midgut, to complete its life cycle. In mammalian malaria species, the chitinase gene comprises either a large or a short copy. In the avian malaria parasites Plasmodium gallinaceum both copies are present, suggesting that a gene duplication in the ancestor to these extant species preceded the loss of either the long or the short copy in Plasmodium parasites of mammals. Plasmodium gallinaceum is not the most widespread and harmful parasite of birds. This study is the first to search for and identify the chitinase gene in one of the most prevalent avian malaria parasites, Plasmodium relictum. METHODS Both copies of P. gallinaceum chitinase were used as reference sequences for primer design. Different sequences of Plasmodium spp. were used to build the phylogenetic tree of chitinase gene. RESULTS The gene encoding for chitinase was identified in isolates of two mitochondrial lineages of P. relictum (SGS1 and GRW4). The chitinase found in these two lineages consists both of the long (PrCHT1) and the short (PrCHT2) copy. The genetic differences found in the long copy of the chitinase gene between SGS1 and GRW4 were higher than the difference observed for the cytochrome b gene. CONCLUSION The identification of both copies in P. relictum sheds light on the phylogenetic relationship of the chitinase gene in the genus Plasmodium. Due to its high variability, the chitinase gene could be used to study the genetic population structure in isolates from different host species and geographic regions.
Collapse
Affiliation(s)
- Luz Garcia-Longoria
- Departamento de Biología Animal, Universidad de Extremadura, E-06071 Badajoz, Spain.
| | | | | |
Collapse
|
33
|
Levi T, Ben-Dov E, Shahi P, Borovsky D, Zaritsky A. Growth and development of Aedes aegypti larvae at limiting food concentrations. Acta Trop 2014; 133:42-4. [PMID: 24524949 DOI: 10.1016/j.actatropica.2014.02.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Revised: 01/26/2014] [Accepted: 02/01/2014] [Indexed: 11/15/2022]
Abstract
Mosquitoes have a complex life-cycle with dramatic changes in shape, function, and habitat. Aedes aegypti was studied by growing individual larvae at different concentrations of a defined rich food source. At higher food concentrations, rate of larval growth was faster, but the time required for 4th instar larvae to molt into the pupal stage was unexpectedly extended. These opposite tendencies resulted in constant times from hatching to pupation and up to adult eclosion at permissive food concentrations. The results demonstrate that nutritional conditions of 4th instar larvae impact initiation of the first metamorphic molt.
Collapse
Affiliation(s)
- Tal Levi
- Department of Life Sciences, Ben-Gurion University of the Negev, POB 653, Be'er-Sheva 84105, Israel
| | | | - Preeti Shahi
- Faculty of Natural Sciences, Ben-Gurion University of the Negev, POB 653, Kiryat Bergman, Be'er-Sheva 84105, Israel
| | - Dov Borovsky
- Borovsky Consulting, 135 36th CT, Vero Beach, FL 32968, USA
| | - Arieh Zaritsky
- Faculty of Natural Sciences, Ben-Gurion University of the Negev, POB 653, Kiryat Bergman, Be'er-Sheva 84105, Israel.
| |
Collapse
|
34
|
Bahia AC, Dong Y, Blumberg BJ, Mlambo G, Tripathi A, BenMarzouk-Hidalgo OJ, Chandra R, Dimopoulos G. Exploring Anopheles gut bacteria for Plasmodium blocking activity. Environ Microbiol 2014; 16:2980-94. [PMID: 24428613 DOI: 10.1111/1462-2920.12381] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 12/31/2013] [Indexed: 12/30/2022]
Abstract
Malaria parasite transmission requires the successful development of Plasmodium gametocytes into flagellated microgametes upon mosquito blood ingestion, and the subsequent fertilization of microgametes and macrogametes for the development of motile zygotes, called ookinetes, which invade and transverse the Anopheles vector mosquito midgut at around 18-36 h after blood ingestion. Within the mosquito midgut, the malaria parasite has to withstand the mosquito's innate immune response and the detrimental effect of its commensal bacterial flora. We have assessed the midgut colonization capacity of five gut bacterial isolates from field-derived, and two from laboratory colony, mosquitoes and their effect on Plasmodium development in vivo and in vitro, along with their impact on mosquito survival. Some bacterial isolates activated the mosquito's immune system, affected the mosquito's lifespan, and were capable of blocking Plasmodium development. We have also shown that the ability of these bacteria to inhibit the parasites is likely to involve different mechanisms and factors. A Serratia marcescens isolate was particularly efficient in colonizing the mosquitoes' gut, compromising mosquito survival and inhibiting both Plasmodium sexual- and asexual-stage through secreted factors, thereby rendering it a potential candidate for the development of a malaria transmission intervention strategy.
Collapse
Affiliation(s)
- Ana C Bahia
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
In vitro development of Haemoproteus parasites: the efficiency of reproductive cells increase during simultaneous sexual process of different lineages. Parasitol Res 2014; 113:1417-23. [DOI: 10.1007/s00436-014-3782-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 01/10/2014] [Indexed: 11/26/2022]
|
36
|
Fidelis QC, Ribeiro TA, Araújo MF, de Carvalho MG. Ouratea genus: chemical and pharmacological aspects. REVISTA BRASILEIRA DE FARMACOGNOSIA-BRAZILIAN JOURNAL OF PHARMACOGNOSY 2014. [DOI: 10.1590/0102-695x20142413361] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
37
|
Tchioffo MT, Boissière A, Churcher TS, Abate L, Gimonneau G, Nsango SE, Awono-Ambéné PH, Christen R, Berry A, Morlais I. Modulation of malaria infection in Anopheles gambiae mosquitoes exposed to natural midgut bacteria. PLoS One 2013; 8:e81663. [PMID: 24324714 PMCID: PMC3855763 DOI: 10.1371/journal.pone.0081663] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Accepted: 10/15/2013] [Indexed: 12/15/2022] Open
Abstract
The development of Plasmodium falciparum within the Anopheles gambiae mosquito relies on complex vector-parasite interactions, however the resident midgut microbiota also plays an important role in mediating parasite infection. In natural conditions, the mosquito microbial flora is diverse, composed of commensal and symbiotic bacteria. We report here the isolation of culturable midgut bacteria from mosquitoes collected in the field in Cameroon and their identification based on the 16S rRNA gene sequencing. We next measured the effect of selected natural bacterial isolates on Plasmodium falciparum infection prevalence and intensity over multiple infectious feedings and found that the bacteria significantly reduced the prevalence and intensity of infection. These results contrast with our previous study where the abundance of Enterobacteriaceae positively correlated with P. falciparum infection (Boissière et al. 2012). The oral infection of bacteria probably led to the disruption of the gut homeostasis and activated immune responses, and this pinpoints the importance of studying microbe-parasite interactions in natural conditions. Our results indicate that the effect of bacterial exposure on P. falciparum infection varies with factors from the parasite and the human host and calls for deeper dissection of these parameters for accurate interpretation of bacterial exposure results in laboratory settings.
Collapse
Affiliation(s)
- Majoline T. Tchioffo
- UMR MIVEGEC (IRD 224- CNRS 5290- UM1- UM2), Institut de Recherche pour le Développement, Montpellier, France
- Laboratoire d'entomologie médicale, Organisation de Coordination pour la lutte contre les Endémies en Afrique Centrale, Yaoundé, Cameroon
| | - Anne Boissière
- UMR MIVEGEC (IRD 224- CNRS 5290- UM1- UM2), Institut de Recherche pour le Développement, Montpellier, France
| | - Thomas S. Churcher
- Department of Infectious Disease Epidemiology, Imperial College London, London, United Kingdom
| | - Luc Abate
- UMR MIVEGEC (IRD 224- CNRS 5290- UM1- UM2), Institut de Recherche pour le Développement, Montpellier, France
| | - Geoffrey Gimonneau
- UMR MIVEGEC (IRD 224- CNRS 5290- UM1- UM2), Institut de Recherche pour le Développement, Montpellier, France
- Laboratoire d'entomologie médicale, Organisation de Coordination pour la lutte contre les Endémies en Afrique Centrale, Yaoundé, Cameroon
| | - Sandrine E. Nsango
- Laboratoire d'entomologie médicale, Organisation de Coordination pour la lutte contre les Endémies en Afrique Centrale, Yaoundé, Cameroon
- Université de Douala, Faculté de Médecine et des Sciences Pharmaceutiques, Douala, Cameroon
| | - Parfait H. Awono-Ambéné
- Laboratoire d'entomologie médicale, Organisation de Coordination pour la lutte contre les Endémies en Afrique Centrale, Yaoundé, Cameroon
| | - Richard Christen
- CNRS UMR 7138, Université de Nice, Faculté des Sciences, Nice, France
- Laboratoire de Biologie Virtuelle, UMR 713, Université de Nice, Faculté des Sciences, Nice, France
| | - Antoine Berry
- Service de Parasitologie-Mycologie, Centre Hospitalier Universitaire de Toulouse, Hôpital Rangueil, Toulouse, France
| | - Isabelle Morlais
- UMR MIVEGEC (IRD 224- CNRS 5290- UM1- UM2), Institut de Recherche pour le Développement, Montpellier, France
- Laboratoire d'entomologie médicale, Organisation de Coordination pour la lutte contre les Endémies en Afrique Centrale, Yaoundé, Cameroon
- * E-mail:
| |
Collapse
|
38
|
Walker DM, Oghumu S, Gupta G, McGwire BS, Drew ME, Satoskar AR. Mechanisms of cellular invasion by intracellular parasites. Cell Mol Life Sci 2013; 71:1245-63. [PMID: 24221133 DOI: 10.1007/s00018-013-1491-1] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 10/04/2013] [Accepted: 10/07/2013] [Indexed: 12/22/2022]
Abstract
Numerous disease-causing parasites must invade host cells in order to prosper. Collectively, such pathogens are responsible for a staggering amount of human sickness and death throughout the world. Leishmaniasis, Chagas disease, toxoplasmosis, and malaria are neglected diseases and therefore are linked to socio-economical and geographical factors, affecting well-over half the world's population. Such obligate intracellular parasites have co-evolved with humans to establish a complexity of specific molecular parasite-host cell interactions, forming the basis of the parasite's cellular tropism. They make use of such interactions to invade host cells as a means to migrate through various tissues, to evade the host immune system, and to undergo intracellular replication. These cellular migration and invasion events are absolutely essential for the completion of the lifecycles of these parasites and lead to their for disease pathogenesis. This review is an overview of the molecular mechanisms of protozoan parasite invasion of host cells and discussion of therapeutic strategies, which could be developed by targeting these invasion pathways. Specifically, we focus on four species of protozoan parasites Leishmania, Trypanosoma cruzi, Plasmodium, and Toxoplasma, which are responsible for significant morbidity and mortality.
Collapse
Affiliation(s)
- Dawn M Walker
- Department of Microbial Infection and Immunity, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA
| | | | | | | | | | | |
Collapse
|
39
|
Reduced glycerol incorporation into phospholipids contributes to impaired intra-erythrocytic growth of glycerol kinase knockout Plasmodium falciparum parasites. Biochim Biophys Acta Gen Subj 2013; 1830:5326-34. [PMID: 23954205 DOI: 10.1016/j.bbagen.2013.08.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 07/12/2013] [Accepted: 08/09/2013] [Indexed: 11/22/2022]
Abstract
BACKGROUND Malaria is a devastating disease and Plasmodium falciparum is the most lethal parasite infecting humans. Understanding the biology of this parasite is vital in identifying potential novel drug targets. During every 48-hour intra-erythrocytic asexual replication cycle, a single parasite can produce up to 32 progeny. This extensive proliferation implies that parasites require substantial amounts of lipid precursors for membrane biogenesis. Glycerol kinase is a highly conserved enzyme that functions at the interface of lipid synthesis and carbohydrate metabolism. P. falciparum glycerol kinase catalyzes the ATP-dependent phosphorylation of glycerol to glycerol-3-phosphate, a major phospholipid precursor. METHODS The P. falciparum glycerol kinase gene was disrupted using double crossover homologous DNA recombination to generate a knockout parasite line. Southern hybridization and mRNA analysis were used to verify gene disruption. Parasite growth rates were monitored by flow cytometry. Radiolabelling studies were used to assess incorporation of glycerol into parasite phospholipids. RESULTS Disruption of the P. falciparum glycerol kinase gene produced viable parasites, but their growth was significantly reduced to 56.5±1.8% when compared to wild type parasites. (14)C-glycerol incorporation into the major phospholipids of the parasite membrane, phosphatidylcholine and phosphatidylethanolamine, was 48.4±10.8% and 53.1±5.7% relative to an equivalent number of wild type parasites. CONCLUSIONS P. falciparum glycerol kinase is required for optimal intra-erythrocytic asexual parasite development. Exogenous glycerol may be used as an alternative carbon source for P. falciparum phospholipid biogenesis, despite the lack of glycerol kinase to generate glycerol-3-phosphate. GENERAL SIGNIFICANCE These studies provide new insight into glycerolipid metabolism in P. falciparum.
Collapse
|
40
|
Mitraka E, Stathopoulos S, Siden-Kiamos I, Christophides GK, Louis C. Asaia accelerates larval development of Anopheles gambiae. Pathog Glob Health 2013; 107:305-11. [PMID: 24091152 DOI: 10.1179/2047773213y.0000000106] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Arthropod borne diseases cause significant human morbidity and mortality and, therefore, efficient measures to control transmission of the disease agents would have great impact on human health. One strategy to achieve this goal is based on the manipulation of bacterial symbionts of vectors. Bacteria of the Gram-negative, acetic acid bacterium genus Asaia have been found to be stably associated with larvae and adults of the Southeast Asian malaria vector Anopheles stephensi, dominating the microbiota of the mosquito. We show here that after the infection of Anopheles gambiae larvae with Asaia the bacteria were stably associated with the mosquitoes, becoming part of the microflora of the midgut and remaining there for the duration of the life cycle. Moreover they were passed on to the next generation through vertical transmission. Additionally, we show that there is an increase in the developmental rate when additional bacteria are introduced into the organism which leads us to the conclusion that Asaia plays a yet undetermined crucial role during the larval stages. Our microarray analysis showed that the larval genes that are mostly affected are involved in cuticle formation, and include mainly members of the CPR gene family.
Collapse
Affiliation(s)
- Elvira Mitraka
- Institute of Molecular Biology and Biotechnology, FORTH, Heraklion, Greece
| | | | | | | | | |
Collapse
|
41
|
Wang J, Zhang Y, Zhao YO, Li MWM, Zhang L, Dragovic S, Abraham NM, Fikrig E. Anopheles gambiae circumsporozoite protein-binding protein facilitates plasmodium infection of mosquito salivary glands. J Infect Dis 2013; 208:1161-9. [PMID: 23801601 DOI: 10.1093/infdis/jit284] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Malaria, a mosquito-borne disease caused by Plasmodium species, causes substantial morbidity and mortality throughout the world. Plasmodium sporozoites mature in oocysts formed in the mosquito gut wall and then invade the salivary glands, where they remain until transmitted to the vertebrate host during a mosquito bite. The Plasmodium circumsporozoite protein (CSP) binds to salivary glands and plays a role in the invasion of this organ by sporozoites. We identified an Anopheles salivary gland protein, named CSP-binding protein (CSPBP), that interacts with CSP. Downregulation of CSPBP in mosquito salivary glands inhibited invasion by Plasmodium organisms. In vivo bioassays showed that mosquitoes that were fed blood with CSPBP antibody displayed a 25% and 90% reduction in the parasite load in infected salivary glands 14 and 18 days after the blood meal, respectively. These results suggest that CSPBP is important for the infection of the mosquito salivary gland by Plasmodium organisms and that blocking CSPBP can interfere with the Plasmodium life cycle.
Collapse
Affiliation(s)
- Jiuling Wang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06520-8022, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Ability of TEP1 in intestinal flora to modulate natural resistance of Anopheles dirus. Exp Parasitol 2013; 134:460-5. [PMID: 23648664 DOI: 10.1016/j.exppara.2013.04.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 04/12/2013] [Accepted: 04/13/2013] [Indexed: 11/20/2022]
Abstract
Blocking transmission of malaria is a reliable way to control and eliminate infection. However, in-depth knowledge of the interaction between Plasmodium and mosquito is needed. Studies suggest that innate immunity is the main mechanism inhibiting development of malaria parasites in the mosquito. Recent studies have found that use of antibiotics that inhibit the mosquito gut flora can reduce the immune response of Anopheles gambiae, thereby contributing to the development of malaria parasites. In our study, we used the non susceptible model of Anopheles dirus-Plasmodium yoelii to explore the effect of Anopheles intestinal flora on the natural resistance of A. dirus to P. yoelii. We found that in mosquitoes infected with Plasmodium, the intestinal flora can regulate expression of thioester-containing protein (TEP1) via an RNAi gene-silencing approach. Our results suggest that in the absence of TEP1, the natural microbiota cannot suppress the development of P. yoelii in A. dirus. This suggests that AdTEP1 plays an important role in the resistance of A. dirus to P. yoelii. The intestinal flora may modulate the development of P. yoelii in A. dirus by regulating TEP1 expression.
Collapse
|
43
|
Eappen AG, Smith RC, Jacobs-Lorena M. Enterobacter-activated mosquito immune responses to Plasmodium involve activation of SRPN6 in Anopheles stephensi. PLoS One 2013; 8:e62937. [PMID: 23658788 PMCID: PMC3643921 DOI: 10.1371/journal.pone.0062937] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 03/27/2013] [Indexed: 12/20/2022] Open
Abstract
Successful development of Plasmodium in the mosquito is essential for the transmission of malaria. A major bottleneck in parasite numbers occurs during midgut invasion, partly as a consequence of the complex interactions between the endogenous microbiota and the mosquito immune response. We previously identified SRPN6 as an immune component which restricts Plasmodium berghei development in the mosquito. Here we demonstrate that SRPN6 is differentially activated by bacteria in Anopheles stephensi, but only when bacteria exposure occurs on the lumenal surface of the midgut epithelium. Our data indicate that AsSRPN6 is strongly induced following exposure to Enterobacter cloacae, a common component of the mosquito midgut microbiota. We conclude that AsSRPN6 is a vital component of the E. cloacae-mediated immune response that restricts Plasmodium development in the mosquito An. stephensi.
Collapse
Affiliation(s)
- Abraham G. Eappen
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Ryan C. Smith
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Marcelo Jacobs-Lorena
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| |
Collapse
|
44
|
Kangwanrangsan N, Tachibana M, Jenwithisuk R, Tsuboi T, Riengrojpitak S, Torii M, Ishino T. A member of the CPW-WPC protein family is expressed in and localized to the surface of developing ookinetes. Malar J 2013; 12:129. [PMID: 23587146 PMCID: PMC3637178 DOI: 10.1186/1475-2875-12-129] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Accepted: 04/08/2013] [Indexed: 01/11/2023] Open
Abstract
Background Despite the development of malaria control programs, billions of people are still at risk for this infectious disease. Recently, the idea of the transmission-blocking vaccine, which works by interrupting the infection of mosquitoes by parasites, has gained attention as a promising strategy for malaria control and eradication. To date, a limited number of surface proteins have been identified in mosquito-stage parasites and investigated as potential targets for transmission-blocking vaccines. Therefore, for the development of effective transmission-blocking strategies in epidemic areas, it is necessary to identify novel zygote/ookinete surface proteins as candidate antigens. Methods Since the expression of many zygote/ookinete proteins is regulated post-transcriptionally, proteins that are regulated by well-known translational mediators were focused. Through in silico screening, CPW-WPC family proteins were selected as potential zygote/ookinete surface proteins. All experiments were performed in the rodent malaria parasite, Plasmodium yoelii XNL. mRNA and protein expression profiles were examined by RT-PCR and western blotting, respectively, over the course of the life cycle of the malaria parasite. Protein function was also investigated by the generation of gene-disrupted transgenic parasites. Results The CPW-WPC protein family, named after the unique WxC repeat domains, is highly conserved among Plasmodium species. It is revealed that CPW-WPC mRNA transcripts are transcribed in gametocytes, while CPW-WPC proteins are expressed in zygote/ookinete-stage parasites. Localization analysis reveals that one of the CPW-WPC family members, designated as PyCPW-WPC-1, is a novel zygote/ookinete stage-specific surface protein. Targeted disruption of the pycpw-wpc-1 gene caused no obvious defects during ookinete and oocyst formation, suggesting that PyCPW-WPC-1 is not essential for mosquito-stage parasite development. Conclusions It is demonstrated that PyCPW-WPC-1 can be classified as a novel, post-transcriptionally regulated zygote/ookinete surface protein. Additional studies are required to determine whether all CPW-WPC family members are also present on the ookinete surface and share similar biological roles during mosquito-stage parasite development. Further investigations of CPW-WPC family proteins may facilitate understanding of parasite biology in the mosquito stage and development of transmission-blocking vaccines.
Collapse
Affiliation(s)
- Niwat Kangwanrangsan
- Department of Molecular Parasitology, Graduate School of Medicine, Ehime University, Shitsukawa, Toon, Matsuyama, Ehime 791-0295, Japan
| | | | | | | | | | | | | |
Collapse
|
45
|
Sor-Suwan S, Jariyapan N, Roytrakul S, Paemanee A, Saeung A, Thongsahuan S, Phattanawiboon B, Bates PA, Poovorawan Y, Choochote W. Salivary gland proteome of the human malaria vector, Anopheles campestris-like (Diptera: Culicidae). Parasitol Res 2013; 112:1065-1075. [PMID: 23263250 DOI: 10.1007/s00436-012-3233-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Accepted: 11/30/2012] [Indexed: 12/20/2022]
Abstract
Anopheles campestris-like is proven to be a high-potential vector of Plasmodium vivax in Thailand. In this study, A. campestris-like salivary gland proteins were determined and analyzed by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE), two-dimensional gel electrophoresis, and nano-liquid chromatography-mass spectrometry. The total amount of salivary gland proteins in the mosquitoes aged 3-5 days was approximately 0.1 ± 0.05 μg/male and 1.38 ± 0.01 μg/female. SDS-PAGE analysis revealed at least 12 major proteins found in the female salivary glands and each morphological region of the female glands contained different major proteins. Two-dimensional gel electrophoresis showed approximately 20 major and several minor protein spots displaying relative molecular masses from 10 to 72 kDa with electric points ranging from 3.9 to 10. At least 15 glycoproteins were detected in the female glands. Similar electrophoretic protein profiles were detected comparing the male and proximal-lateral lobes of the female glands, suggesting that these lobes are responsible for sugar feeding. Blood-feeding proteins, i.e., putative 5'-nucleotidase/apyrase, anti-platelet protein, long-form D7 salivary protein, D7-related 1 protein, and gSG6, were detected in the distal-lateral lobes (DL) and/or medial lobes (ML) of the female glands. The major spots related to housekeeping proteins from other arthropod species including Culex quinquefasciatus serine/threonine-protein kinase rio3 expressed in both male and female glands, Ixodes scapularis putative sil1 expressed in DL and ML, and I. scapularis putative cyclophilin A expressed in DL. These results provide information for further study on the salivary gland proteins of A. campestris-like that are involved in hematophagy and disease transmission.
Collapse
Affiliation(s)
- Sriwatapron Sor-Suwan
- Department of Parasitology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Wang S, Jacobs-Lorena M. Genetic approaches to interfere with malaria transmission by vector mosquitoes. Trends Biotechnol 2013; 31:185-93. [PMID: 23395485 PMCID: PMC3593784 DOI: 10.1016/j.tibtech.2013.01.001] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 01/03/2013] [Accepted: 01/03/2013] [Indexed: 11/20/2022]
Abstract
Malaria remains one of the most devastating diseases worldwide, causing over 1 million deaths every year. The most vulnerable stages of Plasmodium development in the vector mosquito occur in the midgut lumen, making the midgut a prime target for intervention. Mosquito transgenesis and paratransgenesis are two novel strategies that aim at rendering the vector incapable of sustaining Plasmodium development. Mosquito transgenesis involves direct genetic engineering of the mosquito itself for delivery of anti-Plasmodium effector molecules. Conversely, paratransgenesis involves the genetic modification of mosquito symbionts for expression of anti-pathogen effector molecules. Here we consider both genetic manipulation strategies for rendering mosquitoes refractory to Plasmodium infection, and discuss challenges for the translation of laboratory findings to field applications.
Collapse
Affiliation(s)
- Sibao Wang
- Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, China.
| | | |
Collapse
|
47
|
Saboia-Vahia L, Borges-Veloso A, Mesquita-Rodrigues C, Cuervo P, Dias-Lopes G, Britto C, Silva APDB, De Jesus JB. Trypsin-like serine peptidase profiles in the egg, larval, and pupal stages of Aedes albopictus. Parasit Vectors 2013; 6:50. [PMID: 23445661 PMCID: PMC3606343 DOI: 10.1186/1756-3305-6-50] [Citation(s) in RCA: 11] [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] [Received: 10/10/2012] [Accepted: 02/18/2013] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Aedes albopictus, a ubiquitous mosquito, is one of the main vectors of dengue and yellow fever, representing an important threat to public health worldwide. Peptidases play key roles in processes such as digestion, oogenesis, and metamorphosis of insects. However, most of the information on the proteolytic enzymes of mosquitoes is derived from insects in the adult stages and is often directed towards the understanding of blood digestion. The aim of this study was to investigate the expression of active peptidases from the preimaginal stages of Ae. albopictus. METHODS Ae. albopictus eggs, larvae, and pupae were analyzed using zymography with susbtrate-SDS-PAGE. The pH, temperature and peptidase inhibitor sensitivity was evaluated. In addition, the proteolytic activities of larval instars were assayed using the fluorogenic substrate Z-Phe-Arg-AMC. RESULTS The proteolytic profile of the larval stage was composed of 8 bands ranging from 17 to 130 kDa. These enzymes displayed activity in a broad range of pH values, from 5.5 to 10.0. The enzymatic profile of the eggs was similar to that of the larvae, although the proteolytic bands of the eggs showed lower intensities. The pupal stage showed a complex proteolytic pattern, with at least 6 bands with apparent molecular masses ranging from 30 to 150 kDa and optimal activity at pH 7.5. Peptidases from larval instars were active from 10°C to 60°C, with optimal activity at temperatures between 37°C and 50°C. The proteolytic profile of both the larval and pupal stages was inhibited by phenyl-methyl sulfonyl-fluoride (PMSF) and Nα-Tosyl L-lysine chloromethyl ketone hydrochloride (TLCK), indicating that the main peptidases expressed during these developmental stages are trypsin-like serine peptidases. CONCLUSION The preimaginal stages of Ae. albopictus exhibited a complex profile of trypsin-like serine peptidase activities. A comparative analysis of the active peptidase profiles revealed differential expression of trypsin-like isoforms among the preimaginal stages, suggesting that some of these enzymes are stage specific. Additionally, a comparison of the peptidase expression between larvae from eggs collected in the natural environment and larvae obtained from the eggs of female mosquitoes maintained in colonies for a long period of time demonstrated that the proteolytic profile is invariable under such conditions.
Collapse
Affiliation(s)
- Leonardo Saboia-Vahia
- Laboratório de Biologia Molecular e Doenças Endêmicas, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - André Borges-Veloso
- Laboratório de Biologia Molecular e Doenças Endêmicas, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Camila Mesquita-Rodrigues
- Laboratório de Biologia Molecular e Doenças Endêmicas, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Patricia Cuervo
- Laboratório de Pesquisa em Leishmaniose, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Geovane Dias-Lopes
- Laboratório de Biologia Molecular e Doenças Endêmicas, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Constança Britto
- Laboratório de Biologia Molecular e Doenças Endêmicas, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Ana Paula de Barros Silva
- Laboratório de Biologia Molecular e Doenças Endêmicas, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Jose B De Jesus
- Laboratório de Biologia Molecular e Doenças Endêmicas, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
- Departamento de Engenharia de Biossistemas, Universidade Federal de São João Del Rey, Minas Gerais, Brazil
| |
Collapse
|
48
|
Smith RC, Eappen AG, Radtke AJ, Jacobs-Lorena M. Regulation of anti-Plasmodium immunity by a LITAF-like transcription factor in the malaria vector Anopheles gambiae. PLoS Pathog 2012; 8:e1002965. [PMID: 23093936 PMCID: PMC3475675 DOI: 10.1371/journal.ppat.1002965] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2012] [Accepted: 08/27/2012] [Indexed: 11/18/2022] Open
Abstract
The mosquito is the obligate vector for malaria transmission. To complete its development within the mosquito, the malaria parasite Plasmodium must overcome the protective action of the mosquito innate immune system. Here we report on the involvement of the Anopheles gambiae orthologue of a conserved component of the vertebrate immune system, LPS-induced TNFα transcription factor (LITAF), and its role in mosquito anti-Plasmodium immunity. An. gambiae LITAF-like 3 (LL3) expression is up-regulated in response to midgut invasion by both rodent and human malaria parasites. Silencing of LL3 expression greatly increases parasite survival, indicating that LL3 is part of an anti-Plasmodium defense mechanism. Electrophoretic mobility shift assays identified specific LL3 DNA-binding motifs within the promoter of SRPN6, a gene that also mediates mosquito defense against Plasmodium. Further experiments indicated that these motifs play a direct role in LL3 regulation of SRPN6 expression. We conclude that LL3 is a transcription factor capable of modulating SRPN6 expression as part of the mosquito anti-Plasmodium immune response. The mosquito innate immune system serves as the primary defense response against invading pathogens, including that of the malaria parasite Plasmodium. The mosquito immune response is remarkably efficient in eliminating the parasite as indicated by the low prevalence of Plasmodium oocysts in wild caught mosquitoes. In an effort to understand the mechanisms of immune response, we report the first evidence of a LPS-induced TNF-α factor (LITAF)-like gene family in insects and describe the role of one member, LITAF-like 3 (LL3), in anti-Plasmodium immunity in the mosquito Anopheles gambiae. Silencing of LL3 greatly increases parasite survival. The gene appears to function as a transcription factor that binds to specific regions of the SRPN6 promoter, a known anti-Plasmodium gene, and modulates its transcript abundance. In summary, LL3 appears to be a novel component of the mosquito innate immune response.
Collapse
Affiliation(s)
| | | | | | - Marcelo Jacobs-Lorena
- Department of Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
49
|
Tchouassi DP, Quakyi IA, Addison EA, Bosompem KM, Wilson MD, Appawu MA, Brown CA, Boakye DA. Characterization of malaria transmission by vector populations for improved interventions during the dry season in the Kpone-on-Sea area of coastal Ghana. Parasit Vectors 2012; 5:212. [PMID: 23013551 PMCID: PMC3495633 DOI: 10.1186/1756-3305-5-212] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 09/13/2012] [Indexed: 11/17/2022] Open
Abstract
Background Malaria is a major public health problem in Ghana. We present a site-specific entomological study of malaria vectors and transmission indices as part of an effort to develop a site for the testing of improved control strategies including possible vaccine trials. Methods Pyrethrum spray catches (PSC), and indoor and outdoor human landing collections of adult female anopheline mosquitoes were carried out over a six-month period (November 2005 - April 2006) at Kpone-on-Sea, a fishing village in southern Ghana. These were morphologically identified to species level and sibling species of the Anopheles gambiae complex further characterized by the polymerase chain reaction (PCR). Enzyme-linked immunosorbent assay was used to detect Plasmodium falciparum mosquito infectivity and host blood meal sources. Parity rate was examined based on dilatation of ovarian tracheoles following dissection. Results Of the 1233 Anopheles mosquitoes collected, An. gambiae s.l. was predominant (99.5%), followed by An. funestus (0.4%) and An. pharoensis (0.1%). All An. gambiae s.l. examined (480) were identified as An. gambiae s.s. with a majority of M molecular form (98.2%) and only 1.8% S form with no record of M/S hybrid. A significantly higher proportion of anophelines were observed outdoors relative to indoors (χ2 = 159.34, df = 1, p < 0.0000). Only An. gambiae M molecular form contributed to transmission with a high degree of anthropophily, parity rate and an estimated entomological inoculation rate (EIR) of 62.1 infective bites/person/year. The Majority of the infective bites occurred outdoors after 09.00 pm reaching peaks between 12.00-01.00 am and 03.00-04.00 am. Conclusion Anopheles gambiae M molecular form is responsible for maintaining the status quo of malaria in the surveyed site during the study period. The findings provide a baseline for evidence-based planning and implementation of improved malaria interventions. The plasticity observed in biting patterns especially the combined outdoor and early biting behavior of the vector may undermine the success of insecticide-based strategies using insecticide treated nets (ITN) and indoor residual spray (IRS). As such, novel or improved vector interventions should be informed by the local malaria epidemiology data as it relates to vector behavior.
Collapse
|
50
|
Jariyapan N, Roytrakul S, Paemanee A, Junkum A, Saeung A, Thongsahuan S, Sor-suwan S, Phattanawiboon B, Poovorawan Y, Choochote W. Proteomic analysis of salivary glands of female Anopheles barbirostris species A2 (Diptera: Culicidae) by two-dimensional gel electrophoresis and mass spectrometry. Parasitol Res 2012; 111:1239-1249. [PMID: 22584379 DOI: 10.1007/s00436-012-2958-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Accepted: 05/03/2012] [Indexed: 12/21/2022]
Abstract
Salivary gland proteins of adult female Anopheles barbirostris species A2, a potential vector of Plasmodium vivax in Thailand, were analyzed using a proteomic approach (two-dimensional gel electrophoresis followed by nanoLC-MS). Two-dimensional gel electrophoresis revealed approximately 75 well-resolved spots on the reference gel. Most of the protein spots displayed relative molecular masses from 14 to 85 kDa and isoelectric points ranging from 3.9 to 10. The proteome profiles of A. barbirostris species A2 female salivary glands were affected by aging. The typical electrophoretic pattern of the female salivary glands was reached in 48 h post emergence, suggesting the maturation of salivary glands and saliva contents for blood feeding. Proteins involved in blood feeding, i.e., putative 5' nucleotidase/apyrase, anti-platelet protein, long form D7 salivary protein, D7-related 1 protein, and gSG6 salivary protein, start to accumulate from emergence and gradually increase becoming predominant within 48 h. There are different salivary components expressed within each region of the female glands. The blood-feeding proteins were detected in the distal-lateral lobes and/or medial lobes. Proteins detected and/or identified by this approach could be tested in strategies developed to control pathogen and disease transmission. Moreover, the information of a 2D map of the female salivary gland could be used for comparison with other related species in the A. barbirostris complex to distinguish species members in the complex.
Collapse
Affiliation(s)
- Narissara Jariyapan
- Department of Parasitology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
| | | | | | | | | | | | | | | | | | | |
Collapse
|