1
|
He B, Li M, Guo S, Zhu F, Jiao Z, Li J, Tan N, Jiao S, Liu T, Zhang J, Fan Y, Gao Y, Zhou T, Li J, Huang W, Jiang L, Lin Z, Wang S, Xu W. Host complement C3 promotes malaria transmission by killing symbiotic bacteria in the mosquito midgut. Proc Natl Acad Sci U S A 2025; 122:e2424570122. [PMID: 40434644 DOI: 10.1073/pnas.2424570122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 04/24/2025] [Indexed: 05/29/2025] Open
Abstract
Host-derived factors ingested during mosquito blood feeding are poorly understood modulators of malaria transmission. Here, we demonstrated that host complement C3, acquired by mosquitoes during Plasmodium infection, significantly enhanced rodent malaria infection in laboratory-reared mosquitoes. This effect was recapitulated in field-caught Anopheles sinensis mosquitoes, confirming its relevance to malaria transmission in a more natural setting. Moreover, host-derived C3 significantly reduced the efficacy of anti-Pfs25 antibodies in blocking malaria transmission. Mechanistically, host-derived C3 lyses the mosquito midgut symbiont Elizabethkingia anophelis (E. anophelis)-a bacterium that intrinsically suppresses parasite development by blocking the zygote-to-ookinete transition. Strikingly, host-derived C3 in mosquitoes appears to be activated by the alternative pathway, and inhibiting Factor B with Iptacopan (LNP023) reduced Plasmodium falciparum (P. falciparum) infection, while increased the efficacy of anti-Pfs25 antibodies to blocking P. falciparum transmission in the standard membrane-feeding assay. Therefore, this study describes a strategy of the malaria parasite to utilize host complement C3 to promote its transmission and provides us with an avenue to block malaria transmission and improve the blocking efficacy of anti-Pfs25 antibodies by the inhibition of C3 activation.
Collapse
Affiliation(s)
- Biao He
- Department of Pathogenic Biology, Army Medical University, Chongqing 400038, China
| | - Meilin Li
- Department of Pathogenic Biology, Army Medical University, Chongqing 400038, China
| | - Shuai Guo
- Department of Pathogenic Biology, Army Medical University, Chongqing 400038, China
| | - Feng Zhu
- Department of Pathogenic Biology, Army Medical University, Chongqing 400038, China
| | - Zhiwei Jiao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Jianyong Li
- Department of Pathogenic Biology, Army Medical University, Chongqing 400038, China
| | - Nie Tan
- Department of Pathogenic Biology, Army Medical University, Chongqing 400038, China
| | - Shiming Jiao
- Department of Pathogenic Biology, Army Medical University, Chongqing 400038, China
| | - Taiping Liu
- Department of Pathogenic Biology, Army Medical University, Chongqing 400038, China
| | - Jian Zhang
- Department of Pathogenic Biology, Army Medical University, Chongqing 400038, China
| | - Yongling Fan
- Department of Pathogenic Biology, Army Medical University, Chongqing 400038, China
| | - Yuanli Gao
- Department of Pathogenic Biology, Army Medical University, Chongqing 400038, China
| | - Taoli Zhou
- Department of Pathogenic Biology, Army Medical University, Chongqing 400038, China
| | - Jian Li
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Wei Huang
- Key Laboratory of Molecular Virology and Immunology, Shanghai institute of immunity and infection, Institut Pasteur of Shanghai, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200030, China
| | - Lubin Jiang
- Key Laboratory of Molecular Virology and Immunology, Shanghai institute of immunity and infection, Institut Pasteur of Shanghai, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200030, China
| | - Zurui Lin
- Dvision of Malaria Control, Yunnan Institute of Parasitic Diseases, Pu'er, Yunnan 655000, China
| | - Sibao Wang
- New Cornerstone Science Laboratory, Chinese Academy of Sciences Key Laboratory of Insect Developmental and Evolutionary Biology, Chinese Academy of Sciences Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai 200030, China
| | - Wenyue Xu
- Department of Pathogenic Biology, Army Medical University, Chongqing 400038, China
- Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, 400038, China
| |
Collapse
|
2
|
Volkan E, Karanis P. Current Risks and Prevention Strategies Against Vector-Borne Diseases in Cyprus. Microorganisms 2025; 13:726. [PMID: 40284563 PMCID: PMC12029801 DOI: 10.3390/microorganisms13040726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025] Open
Abstract
The island of Cyprus has historically been prone to vector-borne diseases due to its location at the crossroads of three continents. The introduction of novel vectors, microorganisms, or strains in Cyprus, coupled with the global climate change and antimicrobial resistance crisis, can lead to an altered infectious disease landscape and entomological status, causing a rise in vector-borne diseases on the island. The current review provides a broad snapshot of the status of vector-borne infectious diseases and associated risks in Cyprus. Our research has uncovered a pressing issue, the risk of the spread and emergence of various infectious diseases, including West Nile virus and malaria, respectively, due to the presence of Aedes and Anopheles spp. mosquitoes on the island, while underscoring the animal reservoirs of several pathogenic microorganisms. Our research emphasizes the importance of the One Health approach and the collaboration between communities for the improvement of vector control strategies to limit the spread of vector borne diseases.
Collapse
Affiliation(s)
- Ender Volkan
- Department of Basic and Clinical Sciences, University of Nicosia Medical School, 2414 Nicosia, Cyprus
| | - Panagiotis Karanis
- Department of Basic and Clinical Sciences, University of Nicosia Medical School, 2414 Nicosia, Cyprus
| |
Collapse
|
3
|
Sarkar A, Banerjee P, Kar S, Pal A, Mazumdar A. Investigating the influence of blood meal sources on the composition of culturable haemolytic gut bacteria of a wild-caught BTV vector Culicoides oxystoma Kieffer (Diptera: Ceratopogonidae). MEDICAL AND VETERINARY ENTOMOLOGY 2025. [PMID: 39873180 DOI: 10.1111/mve.12793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/15/2025] [Indexed: 01/30/2025]
Abstract
Culicoides oxystoma Kieffer (Diptera: Ceratopogonidae) transmits many pathogens, including seven viruses, four protozoa and one nematode. This species has a wide distribution range across northern Afro-tropical, Palearctic, Australian, Indo-Malayan realms with a broad host spectrum, including cattle, buffaloes, sheep, pigs, dogs, horses and even humans. The heterogeneous nature of Culicoides' blood-feeding patterns is well documented, but the influence of various host blood meal sources on gut bacterial composition remains scant. Adult midges were collected during April (2023) by operating UV light traps in cattle, buffalo sheds and poultry farm in Purulia (India). Besides C. oxystoma, eleven Culicoides species were collected across the sheds and farm, seven of which are vectors. Culicoides liui Wirth and Hubert and C. thurmanae Wirth and Hubert are reported from India for the first time. In all the sheds, engorged females of C. oxystoma were ubiquitous. Identification of culturable gut bacteria and the host blood meal of C. oxystoma were done through the polymerase chain reaction (PCR)-based method. Blood meal analysis confirmed the following hosts: cattle, buffaloes and humans. Identification of blood meal of engorged C. oxystoma caught from poultry farm showed positive results for humans but not for birds. Among bacteria, Bacillus cereus was abundant in all of the engorged females. Bacillus paramycoides and Enterococcus faecium were identified from females feeding on cattle and buffaloes' blood, while Alcaligenes faecalis was found in the gut contents of females that fed on cattle and human blood. The gut bacteria Alcaligenes faecalis exhibited alpha haemolytic activity. In contrast, Bacillus sp., B. cereus, B. flexus, B. licheniformis, B. thuringiensis, B. paramycoides, E. faecium, Paenibacillus sp. and Pseudomonas sp. exhibited beta haemolysis. This is the first report on the composition of gut bacteria, with particular emphasis on the haemolytic bacteria of C. oxystoma with different host blood meals. The pathogenic bacteria B. cereus, B. licheniformis and A. faecalis within the females could potentially impact pathogen acquisition and increase the probability of their zoonotic transmissibility.
Collapse
Affiliation(s)
- Ankita Sarkar
- Entomology Research Unit, Department of Zoology, The University of Burdwan, Burdwan, India
| | - Paramita Banerjee
- Entomology Research Unit, Department of Zoology, The University of Burdwan, Burdwan, India
| | - Surajit Kar
- Entomology Research Unit, Department of Zoology, The University of Burdwan, Burdwan, India
| | - Arjun Pal
- Entomology Research Unit, Department of Zoology, The University of Burdwan, Burdwan, India
| | - Abhijit Mazumdar
- Entomology Research Unit, Department of Zoology, The University of Burdwan, Burdwan, India
| |
Collapse
|
4
|
Rajendran D, Vinayagam S, Sekar K, Bhowmick IP, Sattu K. Symbiotic Bacteria: Wolbachia, Midgut Microbiota in Mosquitoes and Their Importance for Vector Prevention Strategies. MICROBIAL ECOLOGY 2024; 87:154. [PMID: 39681734 PMCID: PMC11649735 DOI: 10.1007/s00248-024-02444-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/02/2024] [Indexed: 12/18/2024]
Abstract
Mosquito-borne illnesses pose a significant threat to eradication under existing vector management measures. Chemo-based vector control strategies (use of insecticides) raise a complication of resistance and environmental pollution. Biological control methods are an alternative approach to overcoming this complication arising from insecticides. The mosquito gut microbiome is essential to supporting the factors that involve metabolic regulation and metamorphic development (from juvenile to adult), as well as the induction of an immune response. The induced immune response includes the JAK-STAT, IMD, and Toll pathways due to the microbial interaction with the midgut cells (MG cells) that prevent disease transmission to humans. The aforementioned sequel to the review provides information about endosymbiont Wolbachia, which contaminates insect cells, including germline and somatic cytoplasm, and inhibits disease-causing pathogen development and transmission by competing for resources within the cell. Moreover, it reduces the host population via cytoplasmic incompatibility (CI), feminization, male killing, and parthenogenesis. Furthermore, the Cif factor in Wolbachia is responsible for CI induction that produces inviable cells with the translocating systems and the embryonic defect-causing protein factor, WalE1 (WD0830), which manipulates the host actin. This potential of Wolbachia can be used to design a paratransgenic system to control vectors in the field. An extracellular symbiotic bacterium such as Asaia, which is grown in the growth medium, is used to transfer lethal genes within itself. Besides, the genetically transferred symbiotic bacteria infect the wild mosquito population and are easily manifold. So, it might be suitable for vector control strategies in the future.
Collapse
Affiliation(s)
- Devianjana Rajendran
- Department of Biotechnology, Periyar University, Centre for Postgraduate and Research Studies, Dharmapuri, Tamil Nadu, 635205, India
| | - Sathishkumar Vinayagam
- Department of Biotechnology, Periyar University, Centre for Postgraduate and Research Studies, Dharmapuri, Tamil Nadu, 635205, India
| | - Kathirvel Sekar
- Department of Biotechnology, Periyar University, Centre for Postgraduate and Research Studies, Dharmapuri, Tamil Nadu, 635205, India
| | - Ipsita Pal Bhowmick
- Department of Malariology, ICMR-RMRCNE Region, Dibrugarh, Assam, 786010, India
| | - Kamaraj Sattu
- Department of Biotechnology, Periyar University, Centre for Postgraduate and Research Studies, Dharmapuri, Tamil Nadu, 635205, India.
| |
Collapse
|
5
|
Rezende FO, da Silva DA, Comini S, de Mendonça S, Santos E, Baldon L, Marçal B, de Freitas AC, Moreira R, Sousa V, Lima M, Rocha M, Moreira LA, Ferreira A. Dietary Influences on the Longevity and Reproductive Success of Anopheles aquasalis in Laboratory Studies: Sucrose vs. Honey. INSECTS 2024; 15:978. [PMID: 39769580 PMCID: PMC11677520 DOI: 10.3390/insects15120978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/21/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025]
Abstract
Malaria continues to be a major public health challenge in tropical and subtropical regions. Anopheles aquasalis, a key laboratory model for malaria research, plays a critical role in the study of vector-parasite interactions. Although vector life traits and environmental factors such as age and resource availability can influence the transmission potential of mosquitoes for Plasmodium parasites, the impact of different adult diets on their survival and reproductive fitness remains underexplored. This study investigates the effects of sucrose and honey diets on the longevity, fertility, and fecundity of Anopheles aquasalis under controlled laboratory conditions. Our results demonstrate that the type of diet significantly affects mosquito survival and reproductive output. Specifically, mosquitoes consuming honey exhibited a substantially longer lifespan and higher fecundity compared to those fed on sucrose. Additionally, eggs laid by honey-fed females had notably higher hatching success rates than those from sucrose-fed females. These findings underscore the profound impact of dietary choices on the reproductive fitness of Anopheles aquasalis, with important implications for laboratory studies focusing on vector-parasite interactions. This study highlights the need for a careful consideration of diet in vector research to ensure accurate assessment of vector competence and disease transmission.
Collapse
Affiliation(s)
- Fernanda Oliveira Rezende
- Mosquitos Vetores: Endossimbiontes e Interação Patógeno-Vetor, Instituto René Rachou-Fiocruz, Belo Horizonte 30190-002, Brazil; (F.O.R.); (D.A.d.S.); (S.C.); (S.d.M.); (L.B.); (B.M.); (A.C.d.F.); (R.M.); (V.S.); (M.L.); (M.R.); (L.A.M.)
| | - Dimas Augusto da Silva
- Mosquitos Vetores: Endossimbiontes e Interação Patógeno-Vetor, Instituto René Rachou-Fiocruz, Belo Horizonte 30190-002, Brazil; (F.O.R.); (D.A.d.S.); (S.C.); (S.d.M.); (L.B.); (B.M.); (A.C.d.F.); (R.M.); (V.S.); (M.L.); (M.R.); (L.A.M.)
| | - Sara Comini
- Mosquitos Vetores: Endossimbiontes e Interação Patógeno-Vetor, Instituto René Rachou-Fiocruz, Belo Horizonte 30190-002, Brazil; (F.O.R.); (D.A.d.S.); (S.C.); (S.d.M.); (L.B.); (B.M.); (A.C.d.F.); (R.M.); (V.S.); (M.L.); (M.R.); (L.A.M.)
| | - Silvana de Mendonça
- Mosquitos Vetores: Endossimbiontes e Interação Patógeno-Vetor, Instituto René Rachou-Fiocruz, Belo Horizonte 30190-002, Brazil; (F.O.R.); (D.A.d.S.); (S.C.); (S.d.M.); (L.B.); (B.M.); (A.C.d.F.); (R.M.); (V.S.); (M.L.); (M.R.); (L.A.M.)
| | - Ellen Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 6627-Pampulha, Belo Horizonte 31270-901, Brazil;
| | - Lívia Baldon
- Mosquitos Vetores: Endossimbiontes e Interação Patógeno-Vetor, Instituto René Rachou-Fiocruz, Belo Horizonte 30190-002, Brazil; (F.O.R.); (D.A.d.S.); (S.C.); (S.d.M.); (L.B.); (B.M.); (A.C.d.F.); (R.M.); (V.S.); (M.L.); (M.R.); (L.A.M.)
| | - Bruno Marçal
- Mosquitos Vetores: Endossimbiontes e Interação Patógeno-Vetor, Instituto René Rachou-Fiocruz, Belo Horizonte 30190-002, Brazil; (F.O.R.); (D.A.d.S.); (S.C.); (S.d.M.); (L.B.); (B.M.); (A.C.d.F.); (R.M.); (V.S.); (M.L.); (M.R.); (L.A.M.)
| | - Amanda Cupertino de Freitas
- Mosquitos Vetores: Endossimbiontes e Interação Patógeno-Vetor, Instituto René Rachou-Fiocruz, Belo Horizonte 30190-002, Brazil; (F.O.R.); (D.A.d.S.); (S.C.); (S.d.M.); (L.B.); (B.M.); (A.C.d.F.); (R.M.); (V.S.); (M.L.); (M.R.); (L.A.M.)
| | - Rafaela Moreira
- Mosquitos Vetores: Endossimbiontes e Interação Patógeno-Vetor, Instituto René Rachou-Fiocruz, Belo Horizonte 30190-002, Brazil; (F.O.R.); (D.A.d.S.); (S.C.); (S.d.M.); (L.B.); (B.M.); (A.C.d.F.); (R.M.); (V.S.); (M.L.); (M.R.); (L.A.M.)
- Laboratório de Ecologia do Adoecimento & Florestas NUPEB/ICEB, Universidade Federal de Ouro Preto, Ouro Preto 35402-163, Brazil
| | - Viviane Sousa
- Mosquitos Vetores: Endossimbiontes e Interação Patógeno-Vetor, Instituto René Rachou-Fiocruz, Belo Horizonte 30190-002, Brazil; (F.O.R.); (D.A.d.S.); (S.C.); (S.d.M.); (L.B.); (B.M.); (A.C.d.F.); (R.M.); (V.S.); (M.L.); (M.R.); (L.A.M.)
| | - Mariana Lima
- Mosquitos Vetores: Endossimbiontes e Interação Patógeno-Vetor, Instituto René Rachou-Fiocruz, Belo Horizonte 30190-002, Brazil; (F.O.R.); (D.A.d.S.); (S.C.); (S.d.M.); (L.B.); (B.M.); (A.C.d.F.); (R.M.); (V.S.); (M.L.); (M.R.); (L.A.M.)
| | - Marcele Rocha
- Mosquitos Vetores: Endossimbiontes e Interação Patógeno-Vetor, Instituto René Rachou-Fiocruz, Belo Horizonte 30190-002, Brazil; (F.O.R.); (D.A.d.S.); (S.C.); (S.d.M.); (L.B.); (B.M.); (A.C.d.F.); (R.M.); (V.S.); (M.L.); (M.R.); (L.A.M.)
| | - Luciano A. Moreira
- Mosquitos Vetores: Endossimbiontes e Interação Patógeno-Vetor, Instituto René Rachou-Fiocruz, Belo Horizonte 30190-002, Brazil; (F.O.R.); (D.A.d.S.); (S.C.); (S.d.M.); (L.B.); (B.M.); (A.C.d.F.); (R.M.); (V.S.); (M.L.); (M.R.); (L.A.M.)
| | - Alvaro Ferreira
- Mosquitos Vetores: Endossimbiontes e Interação Patógeno-Vetor, Instituto René Rachou-Fiocruz, Belo Horizonte 30190-002, Brazil; (F.O.R.); (D.A.d.S.); (S.C.); (S.d.M.); (L.B.); (B.M.); (A.C.d.F.); (R.M.); (V.S.); (M.L.); (M.R.); (L.A.M.)
| |
Collapse
|
6
|
Vinayagam S, Sekar K, Rajendran D, Meenakshisundaram K, Panigrahi A, Arumugam DK, Bhowmick IP, Sattu K. The genetic composition of Anopheles mosquitoes and the diverse population of gut-microbiota within the Anopheles subpictus and Anopheles vagus mosquitoes in Tamil Nadu, India. Acta Trop 2024; 260:107439. [PMID: 39477048 DOI: 10.1016/j.actatropica.2024.107439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 10/21/2024] [Indexed: 11/11/2024]
Abstract
In recent days, in tropical and subtropical regions, secondary vectors of Anopheles mosquitoes are becoming more important in transmitting diseases to humans as primary vectors. Various molecular techniques have separated closely related Anopheles subpictus and Anopheles vagus mosquitoes based on their diversity with other mosquito species. Despite their widespread distribution, the An. subpictus and An. vagus mosquitoes, which carry Plasmodium in their salivary glands, were not considered primary malaria vectors in India. An. vagus mosquitoes are zoophilic and physically similar to An. subpictus. We intend to identify An. subpictus and An. vagus mosquito's sister species based on their Interspaced Transcribed Region-2 (ITS2). We isolated the midgut gDNA from each mosquito and used ITS2-PCR and Sanger sequencing to characterize the mosquito species. BioEdit software aligned the sequences, and MEGA7 built a phylogenetic tree from them. According to this study, the information gathered from these mosquito samples fits the An. subpictus species A form and the An. vagus Indian form. Furthermore, gut microbiome plays an important role in providing nutrients, immunity, and food processing, whereas mosquitoes' midgut microbiota changes their hosts and spreads illnesses. So, we used the Illumina sequencer to look at the gut microbiome diversity of An. subpictus and An. vagus mosquitoes using 16S rRNA-based metagenomic sequencing. Both mosquito species had an abundant phylum of Pseudomonadota (Proteobacteria), Bacillota, Bacteroidota, and Actinomycetota in their gut microbiomes. Notably, both mosquito species had the genus Serratia in their gut. In the subpictus midgut, the genus of Haematosprillum bacteria was dominant, whereas in the vagus mosquito, the genus of Salmonella was dominant. Notably, current research has observed the Sodalis spp. Bacterial genus for the first time.
Collapse
Affiliation(s)
- Sathishkumar Vinayagam
- Department of Biotechnology, Periyar University Centre for Postgraduate and Research Studies, Dharmapuri, Tamil Nadu 635 205, India
| | - Kathirvel Sekar
- Department of Biotechnology, Periyar University Centre for Postgraduate and Research Studies, Dharmapuri, Tamil Nadu 635 205, India
| | - Devianjana Rajendran
- Department of Biotechnology, Periyar University Centre for Postgraduate and Research Studies, Dharmapuri, Tamil Nadu 635 205, India
| | | | | | - Dhanush Kumar Arumugam
- Department of Biotechnology, Periyar University Centre for Postgraduate and Research Studies, Dharmapuri, Tamil Nadu 635 205, India
| | - Ipsita Pal Bhowmick
- ICMR-Regional Medical Research Centre North East Region, Dibrugarh, Assam 786010, India
| | - Kamaraj Sattu
- Department of Biotechnology, Periyar University Centre for Postgraduate and Research Studies, Dharmapuri, Tamil Nadu 635 205, India.
| |
Collapse
|
7
|
Kratou M, Maitre A, Abuin-Denis L, Piloto-Sardiñas E, Corona-Guerrero I, Cano-Argüelles AL, Wu-Chuang A, Bamgbose T, Almazan C, Mosqueda J, Obregón D, Mateos-Hernández L, Said MB, Cabezas-Cruz A. Disruption of bacterial interactions and community assembly in Babesia-infected Haemaphysalis longicornis following antibiotic treatment. BMC Microbiol 2024; 24:322. [PMID: 39237861 PMCID: PMC11378419 DOI: 10.1186/s12866-024-03468-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 08/19/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND A previous study highlighted the role of antibiotic-induced dysbiosis in the tick microbiota, facilitating the transstadial transmission of Babesia microti from nymph to adult in Haemaphysalis longicornis. This study builds on previous findings by analyzing sequence data from an earlier study to investigate bacterial interactions that could be linked to enhanced transstadial transmission of Babesia in ticks. The study employed antibiotic-treated (AT) and control-treated (CT) Haemaphysalis longicornis ticks to investigate shifts in microbial community assembly. Network analysis techniques were utilized to assess bacterial interactions, comparing network centrality measures between AT and CT groups, alongside studying network robustness and connectivity loss. Additionally, functional profiling was conducted to evaluate metabolic diversity in response to antibiotic treatment. RESULTS The analysis revealed notable changes in microbial community assembly in response to antibiotic treatment. Antibiotic-treated (AT) ticks displayed a greater number of connected nodes but fewer correlations compared to control-treated (CT) ticks, indicating a less interactive yet more connected microbial community. Network centrality measures such as degree, betweenness, closeness, and eigenvector centrality, differed significantly between AT and CT groups, suggesting alterations in local network dynamics due to antibiotic intervention. Coxiella and Acinetobacter exhibited disrupted connectivity and roles, with the former showing reduced interactions in AT group and the latter displaying a loss of connected nodes, emphasizing their crucial roles in microbial network stability. Robustness tests against node removal showed decreased stability in AT networks, particularly under directed attacks, confirming a susceptibility of the microbial community to disturbances. Functional profile analysis further indicated a higher diversity and richness in metabolic capabilities in the AT group, reflecting potential shifts in microbial metabolism as a consequence of antimicrobial treatment. CONCLUSIONS Our findings support that bacterial interaction traits boosting the transstadial transmission of Babesia could be associated with reduced colonization resistance. The disrupted microbial interactions and decreased network robustness in AT ticks suggest critical vulnerabilities that could be targeted for managing tick-borne diseases.
Collapse
Affiliation(s)
- Myriam Kratou
- Laboratory of Microbiology, National School of Veterinary Medicine of Sidi Thabet, University of Manouba, Manouba, 2010, Tunisia.
| | - Apolline Maitre
- UMR BIPAR, Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, 94700, France
- INRAE, UR 0045 Laboratoire de Recherches Sur Le Développement de L'Elevage (SELMET LRDE), Corte, France
- EA 7310, Laboratoire de Virologie, Université de Corse, Corte, France
| | - Lianet Abuin-Denis
- UMR BIPAR, Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, 94700, France
- Animal Biotechnology Department, Center for Genetic Engineering and Biotechnology, P.O. Box 6162, Avenue 31 Between 158 and 190, Havana, 10600, Cuba
| | - Elianne Piloto-Sardiñas
- UMR BIPAR, Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, 94700, France
- Direction of Animal Health, National Center for Animal and Plant Health, Carretera de Tapaste y Autopista Nacional, Apartado Postal 10, San José de Las Lajas, Mayabeque, 32700, Cuba
| | - Ivan Corona-Guerrero
- Immunology and Vaccines Laboratory, C. A. Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Queretaro, Mexico
- C.A. Salud Animal y Microbiologia Ambiental. Facultad de Ciencias Naturales, Universidad Autonoma de Queretaro, Queretaro, Mexico
| | - Ana Laura Cano-Argüelles
- Parasitology Laboratory, Institute of Natural Resources and Agrobiology (IRNASA, CSIC), Cordel de Merinas, 40-52, Salamanca, 37008, Spain
| | - Alejandra Wu-Chuang
- UMR BIPAR, Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, 94700, France
| | - Timothy Bamgbose
- Department of Biological Sciences, Microbiology Unit, Kings University, Odeomu, Osun State, Nigeria
- National Agency for Food and Drug Control and Administration (NAFDAC), Isolo, Lagos State, Nigeria
| | - Consuelo Almazan
- Immunology and Vaccines Laboratory, C. A. Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Queretaro, Mexico
- C.A. Salud Animal y Microbiologia Ambiental. Facultad de Ciencias Naturales, Universidad Autonoma de Queretaro, Queretaro, Mexico
| | - Juan Mosqueda
- Immunology and Vaccines Laboratory, C. A. Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Queretaro, Mexico
- C.A. Salud Animal y Microbiologia Ambiental. Facultad de Ciencias Naturales, Universidad Autonoma de Queretaro, Queretaro, Mexico
| | - Dasiel Obregón
- School of Environmental Sciences, University of Guelph, Guelph, ON, Canada
| | - Lourdes Mateos-Hernández
- UMR BIPAR, Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, 94700, France
| | - Mourad Ben Said
- Laboratory of Microbiology, National School of Veterinary Medicine of Sidi Thabet, University of Manouba, Manouba, 2010, Tunisia
- Department of Basic Sciences, Higher Institute of Biotechnology of Sidi Thabet, University of Manouba, Manouba, 2010, Tunisia
| | - Alejandro Cabezas-Cruz
- UMR BIPAR, Laboratoire de Santé Animale, ANSES, INRAE, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, 94700, France.
| |
Collapse
|
8
|
Kumar T, Maitra S, Rai R, Priyanka, Maitra S, Tirkey NN, Kumari R. The dichotomy between probiotic lactic acid bacteria and Plasmodium: A promising therapeutic avenue. Acta Trop 2024; 257:107284. [PMID: 38857820 DOI: 10.1016/j.actatropica.2024.107284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/02/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024]
Abstract
Our understanding of gut microbial populations and their immense influence on host immunity, health, and diseases has increased deeply in recent years. Numerous reports have identified the role of mosquito and mammalian gut microbiota in the modulation of host susceptibility to Plasmodium infection. Artemisinin resistance in malaria-endemic regions necessitates the development of new, safer, and more affordable treatments to supplement existing therapies. In this review, we compiled a colossal amount of data from numerous studies that have assessed the roles played by gut microbial communities in Plasmodium infection, progression, transmission, and severity. Most interestingly, our study points to the overwhelming evidence from experimental studies in mural malaria to human trials, suggesting that the presence of lactic acid bacteria in the gut microbiota of mammalian hosts provides a great degree of protection against malaria. Therefore, our study provides a compelling narrative for probiotic administration as an adjunct therapy for combatting malaria.
Collapse
Affiliation(s)
- Tarkeshwar Kumar
- Department of Zoology, Panch Pargana Kisan College, Ranchi University, Ranchi, Jharkhand, 835204, India.
| | - Satarupa Maitra
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Richa Rai
- Department of Zoology, Allahabad University, Prayagraj, Uttar Pradesh, India
| | - Priyanka
- Department of Zoology, Allahabad University, Prayagraj, Uttar Pradesh, India
| | - Satwat Maitra
- Noida International Institute of Medical Sciences, Greater Noida, Uttar Pradesh, India
| | | | - Rajesh Kumari
- Department of Zoology, Allahabad University, Prayagraj, Uttar Pradesh, India
| |
Collapse
|
9
|
Garcia-Van Smévoorde M, Calvez E, Quétel I, Dollin C, Breurec S, Vega-Rúa A. Ingestion of amoxicillin-clavulanic acid at therapeutic concentration during blood meal impacts Aedes aegypti microbiota and dengue virus transmission. Sci Rep 2024; 14:13701. [PMID: 38871831 DOI: 10.1038/s41598-024-64221-2] [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: 02/06/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024] Open
Abstract
Dengue virus (DENV), mainly transmitted by Aedes aegypti mosquitoes, is the most prevalent arbovirus worldwide, representing a public health problem in tropical and subtropical countries. In these areas, antibiotic consumption rises which may impact both mosquito microbiota and dengue transmission. Here, we assessed how the ingestion by Ae. aegypti of therapeutic concentrations of amoxicillin-clavulanic Acid association (Amox/Clav), a broad-spectrum antibiotic used to treat febrile symptoms worldwide, impacted its microbiota. We also evaluated whether simultaneous ingestion of antibiotic and DENV impacted Ae. aegypti ability to transmit this virus. We found that Amox/Clav ingestion impacted microbiota composition in Ae. aegypti and we confirmed such impact in field-collected mosquitoes. Furthermore, we observed that Amox/Clav ingestion enhanced DENV dissemination and transmission by this mosquito at 21 days post-DENV exposure. These findings increase our understanding of factors linked to human hosts that may influence dengue transmission dynamics in regions with mass-drug administration programs.
Collapse
Affiliation(s)
- Margot Garcia-Van Smévoorde
- Vector Control Research Laboratory, Transmission Reservoir and Pathogens Diversity Unit, Institut Pasteur de La Guadeloupe, 97139, Les Abymes, Guadeloupe, France
| | - Elodie Calvez
- Vector Control Research Laboratory, Transmission Reservoir and Pathogens Diversity Unit, Institut Pasteur de La Guadeloupe, 97139, Les Abymes, Guadeloupe, France
| | - Isaure Quétel
- Microbial Ecosystems Interaction Laboratory, Transmission Reservoir and Pathogens Diversity Unit, Institut Pasteur de La Guadeloupe, 97139, Les Abymes, Guadeloupe, France
| | - Christelle Dollin
- Vector Control Research Laboratory, Transmission Reservoir and Pathogens Diversity Unit, Institut Pasteur de La Guadeloupe, 97139, Les Abymes, Guadeloupe, France
| | - Sébastien Breurec
- Microbial Ecosystems Interaction Laboratory, Transmission Reservoir and Pathogens Diversity Unit, Institut Pasteur de La Guadeloupe, 97139, Les Abymes, Guadeloupe, France
- Department of Clinical Microbiology, University Hospitals of Guadeloupe, 97159, Pointe-À-Pitre/Les Abymes, Guadeloupe, France
- Faculty of Medecine Hyacinthe Bastaraud, University of the Antilles, 97110, Pointe-À-Pitre, Guadeloupe, France
- INSERM 1424, Center for Clinical Investigation, University Hospital Center of Guadeloupe, 97139, Les Abymes, Guadeloupe, France
- Pathogenesis and Control of Chronic and Emerging Infections, INSERM, Etablissement Français du Sang, University of Montpellier, 34394, Montpellier, France
| | - Anubis Vega-Rúa
- Vector Control Research Laboratory, Transmission Reservoir and Pathogens Diversity Unit, Institut Pasteur de La Guadeloupe, 97139, Les Abymes, Guadeloupe, France.
| |
Collapse
|
10
|
Cai JA, Christophides GK. Immune interactions between mosquitoes and microbes during midgut colonization. CURRENT OPINION IN INSECT SCIENCE 2024; 63:101195. [PMID: 38552792 DOI: 10.1016/j.cois.2024.101195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 03/14/2024] [Accepted: 03/21/2024] [Indexed: 04/07/2024]
Abstract
Mosquitoes encounter diverse microbes during their lifetime, including symbiotic bacteria, shaping their midgut ecosystem. The organization of the midgut supports microbiota persistence while defending against potential pathogens. The influx of nutrients during blood feeding triggers bacterial proliferation, challenging host homeostasis. Immune responses, aimed at controlling bacterial overgrowth, impact blood-borne pathogens such as malaria parasites. However, parasites deploy evasion strategies against mosquito immunity. Leveraging these mechanisms could help engineer malaria-resistant mosquitoes, offering a transformative tool for malaria elimination.
Collapse
Affiliation(s)
- Julia A Cai
- Department of Life Sciences, Imperial College London, Exhibition Road, SW7 2AZ London, United Kingdom
| | - George K Christophides
- Department of Life Sciences, Imperial College London, Exhibition Road, SW7 2AZ London, United Kingdom.
| |
Collapse
|
11
|
Polycarpo CR, Walter-Nuno AB, Azevedo-Reis L, Paiva-Silva GO. The vector-symbiont affair: a relationship as (im)perfect as it can be. CURRENT OPINION IN INSECT SCIENCE 2024; 63:101203. [PMID: 38705385 DOI: 10.1016/j.cois.2024.101203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/07/2024]
Abstract
Vector-borne diseases are globally prevalent and represent a major socioeconomic problem worldwide. Blood-sucking arthropods transmit most pathogenic agents that cause these human infections. The pathogens transmission to their vertebrate hosts depends on how efficiently they infect their vector, which is particularly impacted by the microbiota residing in the intestinal lumen, as well as its cells or internal organs such as ovaries. The balance between costs and benefits provided by these interactions ultimately determines the outcome of the relationship. Here, we will explore aspects concerning the nature of microbe-vector interactions, including the adaptive traits required for their establishment, the varied outcomes of symbiotic interactions, as well as the factors influencing the transition of these relationships across a continuum from parasitism to mutualism.
Collapse
Affiliation(s)
- Carla R Polycarpo
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro 21941-902, Brazil
| | - Ana B Walter-Nuno
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro 21941-902, Brazil
| | - Leonan Azevedo-Reis
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro 21941-902, Brazil
| | - Gabriela O Paiva-Silva
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro 21941-902, Brazil.
| |
Collapse
|
12
|
Garrigós M, Garrido M, Morales-Yuste M, Martínez-de la Puente J, Veiga J. Survival effects of antibiotic exposure during the larval and adult stages in the West Nile virus vector Culex pipiens. INSECT SCIENCE 2024; 31:542-550. [PMID: 37559499 DOI: 10.1111/1744-7917.13259] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/26/2023] [Accepted: 07/15/2023] [Indexed: 08/11/2023]
Abstract
The ability of mosquitoes to transmit a pathogen is affected, among other factors, by their survival rate, which is partly modulated by their microbiota. Mosquito microbiota is acquired during the larval phase and modified during their development and adult feeding behavior, being highly dependent on environmental factors. Pharmaceutical residues including antibiotics are widespread pollutants potentially being present in mosquito breeding waters likely affecting their microbiota. Here, we used Culex pipiens mosquitoes to assess the impact of antibiotic exposure during the larval and adult stages on the survival rate of adult mosquitoes. Wild-collected larvae were randomly assigned to two treatments: larvae maintained in water supplemented with antibiotics and control larvae. Emerged adults were subsequently assigned to each of two treatments, fed with sugar solution with antibiotics and fed only with sugar solution (controls). Larval exposure to antibiotics significantly increased the survival rate of adult females that received a control diet. In addition, the effect of adult exposure to antibiotics on the survival rate of both male and female mosquitoes depended on the number of days that larvae fed ad libitum in the laboratory before emergence. In particular, shorter larval ad libitum feeding periods reduced the survival rate of antibiotic-treated adult mosquitoes compared with those that emerged after a longer larval feeding period. These differences were not found in control adult mosquitoes. Our results extend the current understanding of the impact of antibiotic exposure of mosquitoes on a key component of vectorial capacity, that is the vector survival rate.
Collapse
Affiliation(s)
- Marta Garrigós
- Faculty of Pharmacy, Department of Parasitology, University of Granada, Granada, Spain
| | - Mario Garrido
- Faculty of Pharmacy, Department of Parasitology, University of Granada, Granada, Spain
| | - Manuel Morales-Yuste
- Faculty of Pharmacy, Department of Parasitology, University of Granada, Granada, Spain
| | - Josué Martínez-de la Puente
- Faculty of Pharmacy, Department of Parasitology, University of Granada, Granada, Spain
- CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Jesús Veiga
- Faculty of Pharmacy, Department of Parasitology, University of Granada, Granada, Spain
| |
Collapse
|
13
|
Du J, Yin H, Li J, Zhang W, Ding G, Zhou D, Sun Y, Shen B. Transcription factor B-H2 regulates CYP9J34 expression conveying deltamethrin resistance in Culex pipiens pallens. PEST MANAGEMENT SCIENCE 2024; 80:1991-2000. [PMID: 38092527 DOI: 10.1002/ps.7934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 12/08/2023] [Accepted: 12/14/2023] [Indexed: 01/19/2024]
Abstract
BACKGROUND Mosquitoes are vectors of various diseases, posing significant health threats worldwide. Chemical pesticides, particularly pyrethroids like deltamethrin, are commonly used for mosquito control, but the emergence of resistant mosquito populations has become a concern. In the deltamethrin-resistant (DR) strain of Culex pipiens pallens, the highly expressed cytochrome P450 9 J34 (CYP9J34) gene is believed to play a role in resistance, yet the underlying mechanism remains unclear. RESULTS Quantitative polymerase chain reaction with reverse transcription (qRT-PCR) analysis revealed that the expression of CYP9J34 was 14.6-fold higher in DR strains than in deltamethrin-susceptible (DS) strains. The recombinant production of CYP9J34 protein of Cx. pipiens pallens showed that the protein could directly metabolize deltamethrin, yielding the major metabolite 4'-OH deltamethrin. Through dual luciferase reporter assays and RNA interference, the transcription factor homeobox protein B-H2-like (B-H2) was identified to modulate the expression of the CYP9J34 gene, contributing to mosquito resistance to deltamethrin. CONCLUSIONS Our findings demonstrate that the CYP9J34 protein could directly degrade deltamethrin, and the transcription factor B-H2 could regulate CYP9J34 expression, influencing the resistance of mosquitoes to deltamethrin. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Jiajia Du
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Haitao Yin
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Jinze Li
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Wenxing Zhang
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Guangshuo Ding
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Dan Zhou
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Yan Sun
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, China
| | - Bo Shen
- Department of Pathogen Biology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
14
|
Zhong Z, Wang K, Wang J. Tick symbiosis. CURRENT OPINION IN INSECT SCIENCE 2024; 62:101163. [PMID: 38244689 DOI: 10.1016/j.cois.2024.101163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 12/29/2023] [Accepted: 01/12/2024] [Indexed: 01/22/2024]
Abstract
As obligate blood-feeders, ticks serve as vectors for a variety of pathogens that pose threats on both human and livestock health. The microbiota that ticks harbor play important roles in influencing tick nutrition, development, reproduction, and vector. These microbes also affect the capacity of ticks to transmit pathogens (vector competence). Therefore, comprehending the functions of tick microbiota will help in developing novel and effective tick control strategies. Here, we summarize the effects of main tick symbiotic bacteria on tick physiology and vector competency.
Collapse
Affiliation(s)
- Zhengwei Zhong
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, PR China; Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, PR China
| | - Kun Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, PR China; Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, PR China
| | - Jingwen Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, PR China; Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, PR China.
| |
Collapse
|
15
|
Romoli O, Henrion-Lacritick A, Blanc H, Frangeul L, Saleh MC. Limitations in harnessing oral RNA interference as an antiviral strategy in Aedes aegypti. iScience 2024; 27:109261. [PMID: 38433898 PMCID: PMC10907830 DOI: 10.1016/j.isci.2024.109261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/09/2024] [Accepted: 02/14/2024] [Indexed: 03/05/2024] Open
Abstract
Mosquitoes, particularly Aedes aegypti, are critical vectors for globally significant pathogenic viruses. This study examines the limitations of oral RNA interference (RNAi) as a strategy to disrupt viral transmission by Ae. aegypti. We hypothesized that double-stranded RNA (dsRNA) targeting the Zika virus (ZIKV) or chikungunya virus (CHIKV) genomes produced by engineered bacterial symbionts could trigger an antiviral response. Mosquitoes mono-colonized with Escherichia coli producing dsZIK or dsCHIK did not display reduced viral titers following exposure to virus-contaminated bloodmeals and failed to generate dsZIK- or dsCHIK-derived small interfering RNAs. To address potential limitations of bacterial dsRNA release, we explored dsRNA inoculation via feeding and injection. Although viral replication was impeded in mosquitoes injected with dsZIK or dsCHIK, no antiviral effect was observed in dsRNA-fed mosquitoes. These findings highlight complexities of implementing oral RNAi as an antiviral strategy in Ae. aegypti and warrant further exploration of local and systemic RNAi mechanisms.
Collapse
Affiliation(s)
- Ottavia Romoli
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Viruses and RNAi Unit, F-75015 Paris, France
| | | | - Hervé Blanc
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Viruses and RNAi Unit, F-75015 Paris, France
| | - Lionel Frangeul
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Viruses and RNAi Unit, F-75015 Paris, France
| | - Maria-Carla Saleh
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Viruses and RNAi Unit, F-75015 Paris, France
| |
Collapse
|
16
|
Llergo JL, Garuti H, Lopez C, Sanchez J, Calvo D. Artificial nighttime lighting impacts Plasmodium falciparum mature stage V gametocytes infectivity in Anopheles stephensi. Malar J 2024; 23:42. [PMID: 38326842 PMCID: PMC10851600 DOI: 10.1186/s12936-024-04866-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/30/2024] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND Malaria is one of the most important vector-borne diseases of humans with an estimated 241 million cases worldwide in 2020. As an urban and periurban mosquito species, Anopheles stephensi is exposed to artificial human stimuli like light that can alter many aspects of mosquito behaviour, physiology and metabolism. Therefore, fluctuations in the light environment may influence the host, parasite and/or mosquito biology and hence modulate risk for disease transmission. In this study, the effect of artifitial light at night on mosquito infectivity by Plasmodium falciparum during the first hours of blood digestion was tested. METHODS A total of three independent standard membrane feeding assays were performed to artificially fed septic and aseptic mosquitoes with P. falciparum infected blood. After blood feeding, females were transferred to incubators with different photoperiod cycles, so digestion occurred under day artificial light or dark. At 7 and 16 days post blood feeding, mosquitoes were dissected for midguts and salivary glands, respectively. Percentage of mosquitoes fed, percentage of prevalence and P. falciparum oocyst intensity between septic and aseptic mosquitoes in the two different photoperiod regimes, were compared using a Kruskal-Wallis test followed by a Dunn´s multiple comparison test . RESULTS The exposition of mosquitoes to light after they took an infected blood meal has a negative effect on the successful progression of P. falciparum in the mosquito midgut. Antibiotic treatment significantly incremented the number of oocysts per midgut. Photophase significantly reduced the median oocyst intensity in both septic and aseptic mosquitoes. The percentage of oocyst reduction, understood as the percentage of reduction in the mean oocyst intensity of the parasite in the mosquito midgut between photophase and scotophase, was 51% in the case of aseptic mosquitoes and 80% for septic mosquitoes, both in the photophase condition. CONCLUSION Although there are still many gaps in the understanding of parasite-mosquito interactions, these results support the idea that light can, not only, influence mosquito biting behaviour but also parasite success in the mosquito midgut. Hence, light can be considered an interesting additional mosquito-control strategy to reduce mosquito-borne diseases.
Collapse
Affiliation(s)
- Jose Luis Llergo
- Global Health Medicines R&D, GlaxoSmithKline, C/Severo Ochoa 2, Tres Cantos, 28760, Madrid, Spain
| | - Helena Garuti
- Global Health Medicines R&D, GlaxoSmithKline, C/Severo Ochoa 2, Tres Cantos, 28760, Madrid, Spain
| | - Celia Lopez
- Global Health Medicines R&D, GlaxoSmithKline, C/Severo Ochoa 2, Tres Cantos, 28760, Madrid, Spain
| | - Julia Sanchez
- In Vivo Science and Delivery (IVSD), GlaxoSmithKline, C/Severo Ochoa 2, Tres Cantos, 28760, Madrid, Spain
| | - David Calvo
- Global Health Medicines R&D, GlaxoSmithKline, C/Severo Ochoa 2, Tres Cantos, 28760, Madrid, Spain.
| |
Collapse
|
17
|
Yan J, Green K, Noel K, Kim CH, Stone CM. Effects of seasonality and developed land cover on Culex mosquito abundance and microbiome diversity. Front Microbiol 2024; 15:1332970. [PMID: 38404602 PMCID: PMC10885804 DOI: 10.3389/fmicb.2024.1332970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 01/30/2024] [Indexed: 02/27/2024] Open
Abstract
The vectorial capacity of mosquitoes, which influences the dynamics of vector-borne disease transmission, is intricately linked to mosquito abundance and the composition and diversity of their associated microbiomes. However, the influence of environmental factors on mosquito populations and microbiome diversity remains underexplored. Here we examined the effects of seasonality and developed land cover on Culex mosquito abundance and bacterial diversity. Biweekly field sampling of female Culex mosquitoes was conducted using gravid and CDC light traps, spanning summer to autumn across varying developed land cover levels in two urban areas in Central Illinois. Mosquito abundance was assessed by the number of mosquitoes captured per trap night and compared across seasons and developed levels. The mean mosquito abundance for gravid and light traps was 12.96 ± 2.15 and 7.67 ± 1.44, respectively. Notably, higher levels of developed land cover exhibited higher Culex abundance than the low level for light traps, but no significant difference was found between summer and early autumn. In gravid traps, no significant differences were detected across seasons or developed levels. Microbial analysis of the mosquito microbiome revealed that Proteobacteria and Wolbachia, with a mean relative abundance of 80.77 and 52.66% respectively, were identified as the most dominant bacterial phylum and genus. Their relative abundance remained consistent across seasons and developed land cover levels, with negligible variations. Alpha diversity, as measured by observed species, Chao1, Shannon, and Simpson, showed slightly higher values in early-autumn compared to late-summer. A notable pattern of bacterial diversity, as indicated by all four diversity indexes, is evident across varying levels of land development. Significantly, high or intermediate developed levels consistently showed reduced alpha diversity when compared to the lower level. This underscores the pronounced impact of anthropogenic ecological disturbances in shaping mosquito microbiomes. Beta diversity analysis revealed no significant dissimilarities in bacterial community composition across seasons and developed levels, although some separation was noted among different levels of developed land cover. These findings highlight the significant role of environmental factors in shaping mosquito abundance and their associated microbiomes, with potential implications for the vectorial capacity in the transmission of vector-borne diseases.
Collapse
Affiliation(s)
- Jiayue Yan
- Illinois Natural History Survey, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | | | | | | | - Chris M. Stone
- Illinois Natural History Survey, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| |
Collapse
|
18
|
Werling K, Itoe MA, Shaw WR, Hien RD, Bazié BJ, Aminata F, Adams KL, Ouattara BS, Sanou M, Peng D, Dabiré RK, Da DF, Yerbanga RS, Diabaté A, Lefèvre T, Catteruccia F. Development of circulating isolates of Plasmodium falciparum is accelerated in Anopheles vectors with reduced reproductive output. PLoS Negl Trop Dis 2024; 18:e0011890. [PMID: 38206958 PMCID: PMC10807765 DOI: 10.1371/journal.pntd.0011890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 01/24/2024] [Accepted: 12/28/2023] [Indexed: 01/13/2024] Open
Abstract
Anopheles gambiae and its sibling species Anopheles coluzzii are the most efficient vectors of the malaria parasite Plasmodium falciparum. When females of these species feed on an infected human host, oogenesis and parasite development proceed concurrently, but interactions between these processes are not fully understood. Using multiple natural P. falciparum isolates from Burkina Faso, we show that in both vectors, impairing steroid hormone signaling to disrupt oogenesis leads to accelerated oocyst growth and in a manner that appears to depend on both parasite and mosquito genotype. Consistently, we find that egg numbers are negatively linked to oocyst size, a metric for the rate of oocyst development. Oocyst growth rates are also strongly accelerated in females that are in a pre-gravid state, i.e. that fail to develop eggs after an initial blood meal. Overall, these findings advance our understanding of mosquito-parasite interactions that influence P. falciparum development in malaria-endemic regions.
Collapse
Affiliation(s)
- Kristine Werling
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Maurice A. Itoe
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - W. Robert Shaw
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| | | | - Bali Jean Bazié
- Institut de Recherche en Sciences de la Santé/Centre Muraz, Bobo-Dioulasso, Burkina Faso
| | - Fofana Aminata
- Institut de Recherche en Sciences de la Santé/Centre Muraz, Bobo-Dioulasso, Burkina Faso
| | - Kelsey L. Adams
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | | | - Mathias Sanou
- Institut de Recherche en Sciences de la Santé/Centre Muraz, Bobo-Dioulasso, Burkina Faso
| | - Duo Peng
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Roch K. Dabiré
- Institut de Recherche en Sciences de la Santé/Centre Muraz, Bobo-Dioulasso, Burkina Faso
| | - Dari F. Da
- Institut de Recherche en Sciences de la Santé/Centre Muraz, Bobo-Dioulasso, Burkina Faso
| | | | - Abdoulaye Diabaté
- Institut de Recherche en Sciences de la Santé/Centre Muraz, Bobo-Dioulasso, Burkina Faso
| | - Thierry Lefèvre
- Institut de Recherche en Sciences de la Santé/Centre Muraz, Bobo-Dioulasso, Burkina Faso
- MIVEGEC, IRD, CNRS, University of Montpellier, Montpellier, France
| | - Flaminia Catteruccia
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| |
Collapse
|
19
|
Yan J, Kim CH, Chesser L, Ramirez JL, Stone CM. Nutritional stress compromises mosquito fitness and antiviral immunity, while enhancing dengue virus infection susceptibility. Commun Biol 2023; 6:1123. [PMID: 37932414 PMCID: PMC10628303 DOI: 10.1038/s42003-023-05516-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 10/27/2023] [Indexed: 11/08/2023] Open
Abstract
Diet-induced nutritional stress can influence pathogen transmission potential in mosquitoes by impacting life history traits, infection susceptibility, and immunity. To investigate these effects, we manipulate mosquito diets at larval and adult stages, creating two nutritional levels (low and normal), and expose adults to dengue virus (DENV). We observe that egg number is reduced by nutritional stress at both stages and viral exposure separately and jointly, while the likelihood of laying eggs is exclusively influenced by adult nutritional stress. Adult nutritional stress alone shortens survival, while any pairwise combination between both-stage stress and viral exposure have a synergistic effect. Additionally, adult nutritional stress increases susceptibility to DENV infection, while larval nutritional stress likely has a similar effect operating via smaller body size. Furthermore, adult nutritional stress negatively impacts viral titers in infected mosquitoes; however, some survive and show increased titers over time. The immune response to DENV infection is overall suppressed by larval and adult nutritional stress, with specific genes related to Toll, JAK-STAT, and Imd immune signaling pathways, and antimicrobial peptides being downregulated. Our findings underscore the importance of nutritional stress in shaping mosquito traits, infection outcomes, and immune responses, all of which impact the vectorial capacity for DENV transmission.
Collapse
Affiliation(s)
- Jiayue Yan
- Illinois Natural History Survey, University of Illinois at Urbana-Champaign, Champaign, IL, USA.
| | - Chang-Hyun Kim
- Illinois Natural History Survey, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Leta Chesser
- Illinois Natural History Survey, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Jose L Ramirez
- USDA, Agricultural Research Service, National Center for Agricultural Utilization Research, Crop Bioprotection Research Unit, Peoria, IL, USA
| | - Chris M Stone
- Illinois Natural History Survey, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| |
Collapse
|
20
|
Garrigós M, Garrido M, Panisse G, Veiga J, Martínez-de la Puente J. Interactions between West Nile Virus and the Microbiota of Culex pipiens Vectors: A Literature Review. Pathogens 2023; 12:1287. [PMID: 38003752 PMCID: PMC10675824 DOI: 10.3390/pathogens12111287] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/21/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
The flavivirus West Nile virus (WNV) naturally circulates between mosquitoes and birds, potentially affecting humans and horses. Different species of mosquitoes play a role as vectors of WNV, with those of the Culex pipiens complex being particularly crucial for its circulation. Different biotic and abiotic factors determine the capacity of mosquitoes for pathogen transmission, with the mosquito gut microbiota being recognized as an important one. Here, we review the published studies on the interactions between the microbiota of the Culex pipiens complex and WNV infections in mosquitoes. Most articles published so far studied the interactions between bacteria of the genus Wolbachia and WNV infections, obtaining variable results regarding the directionality of this relationship. In contrast, only a few studies investigate the role of the whole microbiome or other bacterial taxa in WNV infections. These studies suggest that bacteria of the genera Serratia and Enterobacter may enhance WNV development. Thus, due to the relevance of WNV in human and animal health and the important role of mosquitoes of the Cx. pipiens complex in its transmission, more research is needed to unravel the role of mosquito microbiota and those factors affecting this microbiota on pathogen epidemiology. In this respect, we finally propose future lines of research lines on this topic.
Collapse
Affiliation(s)
- Marta Garrigós
- Department of Parasitology, University of Granada, 18071 Granada, Spain; (M.G.); (J.V.); (J.M.-d.l.P.)
| | - Mario Garrido
- Department of Parasitology, University of Granada, 18071 Granada, Spain; (M.G.); (J.V.); (J.M.-d.l.P.)
| | - Guillermo Panisse
- CEPAVE—Centro de Estudios Parasitológicos y de Vectores CONICET-UNLP, La Plata 1900, Argentina;
| | - Jesús Veiga
- Department of Parasitology, University of Granada, 18071 Granada, Spain; (M.G.); (J.V.); (J.M.-d.l.P.)
| | - Josué Martínez-de la Puente
- Department of Parasitology, University of Granada, 18071 Granada, Spain; (M.G.); (J.V.); (J.M.-d.l.P.)
- CIBER de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
| |
Collapse
|
21
|
Andrade AO, Santos NAC, Bastos AS, Pontual JDC, Araújo CS, Lima AS, Martinez LN, Ferreira AS, Aguiar ACC, Teles CBG, Guido RVC, Santana RA, Lopes SCP, Medeiros JF, Rizopoulos Z, Vinetz JM, Campo B, Lacerda MVG, Araújo MS. Optimization of Plasmodium vivax infection of colonized Amazonian Anopheles darlingi. Sci Rep 2023; 13:18207. [PMID: 37875508 PMCID: PMC10598059 DOI: 10.1038/s41598-023-44556-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/10/2023] [Indexed: 10/26/2023] Open
Abstract
Obtaining Plasmodium vivax sporozoites is essential for in vitro culture of liver stage parasites, not only to understand fundamental aspects of parasite biology, but also for drug and vaccine development. A major impediment to establish high-throughput in vitro P. vivax liver stage assays for drug development is obtaining sufficient numbers of sporozoites. To do so, female anopheline mosquitoes have to be fed on blood from P. vivax-infected patients through an artificial membrane-feeding system, which in turns requires a well-established Anopheles colony. In this study we established conditions to provide a robust supply of P. vivax sporozoites. Adding a combination of serum replacement and antibiotics to the membrane-feeding protocol was found to best improve sporozoite production. A simple centrifugation method appears to be a possible tool for rapidly obtaining purified sporozoites with a minimal loss of yield. However, this method needs to be better defined since sporozoite viability and hepatocyte infection were not evaluated.
Collapse
Affiliation(s)
- Alice O Andrade
- Plataforma de Produção e Infecção de Vetores da Malária (PIVEM)/Laboratório de Entomologia, Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
- Programa de Pós-Graduação em Saúde Publica, Faculdade de Saúde Pública, Universidade de São Paulo, São Paulo, Brazil
| | - Najara Akira C Santos
- Plataforma de Produção e Infecção de Vetores da Malária (PIVEM)/Laboratório de Entomologia, Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
- Programa de Pós-Graduação em Biologia Experimental, Fundação Universidade Federal de Rondônia, Porto Velho, Rondônia, Brazil
| | - Alessandra S Bastos
- Plataforma de Produção e Infecção de Vetores da Malária (PIVEM)/Laboratório de Entomologia, Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
- Programa de Pós-Graduação em Biologia Experimental, Fundação Universidade Federal de Rondônia, Porto Velho, Rondônia, Brazil
| | - José Daniel C Pontual
- Plataforma de Produção e Infecção de Vetores da Malária (PIVEM)/Laboratório de Entomologia, Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
| | - Cristiane S Araújo
- Plataforma de Produção e Infecção de Vetores da Malária (PIVEM)/Laboratório de Entomologia, Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
- Programa de Pós-Graduação em Conservação e uso de Recursos Naturais - PPGReN, Fundação Universidade Federal de Rondônia, Porto Velho, Rondônia, Brazil
| | - Analice S Lima
- Plataforma de Produção e Infecção de Vetores da Malária (PIVEM)/Laboratório de Entomologia, Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
- Faculdades Integradas Aparício Carvalho (FIMCA), Porto Velho, Rondônia, Brazil
| | - Leandro N Martinez
- Programa de Pós-Graduação em Saúde Publica, Faculdade de Saúde Pública, Universidade de São Paulo, São Paulo, Brazil
- Plataforma de Bioensaios de Malária e Leishmaniose da Fiocruz (PBML), Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
| | - Amália S Ferreira
- Plataforma de Bioensaios de Malária e Leishmaniose da Fiocruz (PBML), Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
| | - Anna Caroline C Aguiar
- Departamento de Biociência, Universidade Federal de São Paulo, Santos, São Paulo, Brazil
| | - Carolina B G Teles
- Programa de Pós-Graduação em Biologia Experimental, Fundação Universidade Federal de Rondônia, Porto Velho, Rondônia, Brazil
- Plataforma de Bioensaios de Malária e Leishmaniose da Fiocruz (PBML), Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
- Rede de Biodiversidade e Biotecnologia da Amazônia Legal - BIONORTE, Porto Velho, Rondônia, Brazil
| | - Rafael V C Guido
- São Carlos Institute of Physics, University of Sao Paulo, São Carlos, São Paulo, Brazil
| | - Rosa A Santana
- Instituto de Pesquisa Clínica Carlos Borborema, Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Stefanie C P Lopes
- Instituto de Pesquisa Clínica Carlos Borborema, Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, Brazil
- Instituto Leônidas & Maria Deane, FIOCRUZ, Manaus, Brazil
| | - Jansen F Medeiros
- Plataforma de Produção e Infecção de Vetores da Malária (PIVEM)/Laboratório de Entomologia, Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
- Programa de Pós-Graduação em Biologia Experimental, Fundação Universidade Federal de Rondônia, Porto Velho, Rondônia, Brazil
| | | | - Joseph M Vinetz
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Alexander von Humboldt Institute of Tropical Medicine and Faculty of Sciences, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Brice Campo
- Medicines for Malaria Venture, Geneva, Switzerland
| | - Marcus Vinicius G Lacerda
- Instituto de Pesquisa Clínica Carlos Borborema, Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas, Manaus, Brazil
- Instituto Leônidas & Maria Deane, FIOCRUZ, Manaus, Brazil
| | - Maisa S Araújo
- Plataforma de Produção e Infecção de Vetores da Malária (PIVEM)/Laboratório de Entomologia, Fiocruz Rondônia, Porto Velho, Rondônia, Brazil.
- Programa de Pós-Graduação em Conservação e uso de Recursos Naturais - PPGReN, Fundação Universidade Federal de Rondônia, Porto Velho, Rondônia, Brazil.
| |
Collapse
|
22
|
Nardini L, Brito-Fravallo E, Campagne P, Pain A, Genève C, Vernick KD, Mitri C. The voltage-gated sodium channel, para, limits Anopheles coluzzii vector competence in a microbiota dependent manner. Sci Rep 2023; 13:14572. [PMID: 37666840 PMCID: PMC10477260 DOI: 10.1038/s41598-023-40432-x] [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: 03/21/2023] [Accepted: 08/10/2023] [Indexed: 09/06/2023] Open
Abstract
The voltage-gated sodium channel, para, is a target of DDT and pyrethroid class insecticides. Single nucleotide mutations in para, called knockdown resistant or kdr, which contribute to resistance against DDT and pyrethroid insecticides, have been correlated with increased susceptibility of Anopheles to the human malaria parasite Plasmodium falciparum. However, a direct role of para activity on Plasmodium infection has not yet been established. Here, using RNA-mediated silencing, we provide in vivo direct evidence for the requirement of wild-type (wt) para function for insecticide activity of deltamethrin. Depletion of wt para, which is susceptible to insecticide, causes deltamethrin tolerance, indicating that insecticide-resistant kdr alleles are likely phenocopies of loss of para function. We then show that normal para activity in An. coluzzii limits Plasmodium infection prevalence for both P. falciparum and P. berghei. A transcriptomic analysis revealed that para activity does not modulate the expression of immune genes. However, loss of para function led to enteric dysbiosis with a significant increase in the total bacterial abundance, and we show that para function limiting Plasmodium infection is microbiota dependent. In the context of the bidirectional "enteric microbiota-brain" axis studied in mammals, these results pave the way for studying whether the activity of the nervous system could control Anopheles vector competence.
Collapse
Affiliation(s)
- Luisa Nardini
- Genetics and Genomics of Insect Vectors Unit, Department of Parasites and Insect Vectors, CNRS, Institut Pasteur, UMR2000, Université de Paris, 75015, Paris, France
| | - Emma Brito-Fravallo
- Genetics and Genomics of Insect Vectors Unit, Department of Parasites and Insect Vectors, CNRS, Institut Pasteur, UMR2000, Université de Paris, 75015, Paris, France
| | - Pascal Campagne
- Center of Bioinformatics, Biostatistics and Integrative Biology, Institut Pasteur, Université de Paris, 75015, Paris, France
| | - Adrien Pain
- Genetics and Genomics of Insect Vectors Unit, Department of Parasites and Insect Vectors, CNRS, Institut Pasteur, UMR2000, Université de Paris, 75015, Paris, France
| | - Corinne Genève
- Genetics and Genomics of Insect Vectors Unit, Department of Parasites and Insect Vectors, CNRS, Institut Pasteur, UMR2000, Université de Paris, 75015, Paris, France
| | - Kenneth D Vernick
- Genetics and Genomics of Insect Vectors Unit, Department of Parasites and Insect Vectors, CNRS, Institut Pasteur, UMR2000, Université de Paris, 75015, Paris, France
| | - Christian Mitri
- Genetics and Genomics of Insect Vectors Unit, Department of Parasites and Insect Vectors, CNRS, Institut Pasteur, UMR2000, Université de Paris, 75015, Paris, France.
| |
Collapse
|
23
|
Su X, Stadler RV, Xu F, Wu J. Malaria Genomics, Vaccine Development, and Microbiome. Pathogens 2023; 12:1061. [PMID: 37624021 PMCID: PMC10459703 DOI: 10.3390/pathogens12081061] [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: 07/21/2023] [Revised: 08/14/2023] [Accepted: 08/15/2023] [Indexed: 08/26/2023] Open
Abstract
Recent advances in malaria genetics and genomics have transformed many aspects of malaria research in areas of molecular evolution, epidemiology, transmission, host-parasite interaction, drug resistance, pathogenicity, and vaccine development. Here, in addition to introducing some background information on malaria parasite biology, parasite genetics/genomics, and genotyping methods, we discuss some applications of genetic and genomic approaches in vaccine development and in studying interactions with microbiota. Genetic and genomic data can be used to search for novel vaccine targets, design an effective vaccine strategy, identify protective antigens in a whole-organism vaccine, and evaluate the efficacy of a vaccine. Microbiota has been shown to influence disease outcomes and vaccine efficacy; studying the effects of microbiota in pathogenicity and immunity may provide information for disease control. Malaria genetics and genomics will continue to contribute greatly to many fields of malaria research.
Collapse
Affiliation(s)
- Xinzhuan Su
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA; (R.V.S.); (F.X.); (J.W.)
| | | | | | | |
Collapse
|
24
|
Duval P, Antonelli P, Aschan-Leygonie C, Valiente Moro C. Impact of Human Activities on Disease-Spreading Mosquitoes in Urban Areas. J Urban Health 2023; 100:591-611. [PMID: 37277669 PMCID: PMC10322816 DOI: 10.1007/s11524-023-00732-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/25/2023] [Indexed: 06/07/2023]
Abstract
Urbanization is one of the leading global trends of the twenty-first century that has a significant impact on health. Among health challenges caused by urbanization, the relationship of urbanization between emergence and the spread of mosquito-borne infectious diseases (MBIDs) is a great public health concern. Urbanization processes encompass social, economic, and environmental changes that directly impact the biology of mosquito species. In particular, urbanized areas experience higher temperatures and pollution levels than outlying areas but also favor the development of infrastructures and objects that are favorable to mosquito development. All these modifications may influence mosquito life history traits and their ability to transmit diseases. This review aimed to summarize the impact of urbanization on mosquito spreading in urban areas and the risk associated with the emergence of MBIDs. Moreover, mosquitoes are considered as holobionts, as evidenced by numerous studies highlighting the role of mosquito-microbiota interactions in mosquito biology. Taking into account this new paradigm, this review also represents an initial synthesis on how human-driven transformations impact microbial communities in larval habitats and further interfere with mosquito behavior and life cycle in urban areas.
Collapse
Affiliation(s)
- Pénélope Duval
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, INRAE, VetAgro Sup, UMR Ecologie Microbienne, Bât. André Lwoff, 10 rue Raphaël Dubois, F-69622, Villeurbanne, France
| | - Pierre Antonelli
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, INRAE, VetAgro Sup, UMR Ecologie Microbienne, Bât. André Lwoff, 10 rue Raphaël Dubois, F-69622, Villeurbanne, France
| | - Christina Aschan-Leygonie
- University of Lyon, Université Lumière Lyon 2, UMR 5600 CNRS Environnement Ville Société, F-69007, Lyon, France
| | - Claire Valiente Moro
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, INRAE, VetAgro Sup, UMR Ecologie Microbienne, Bât. André Lwoff, 10 rue Raphaël Dubois, F-69622, Villeurbanne, France.
| |
Collapse
|
25
|
Cottis S, Blisnick AA, Failloux AB, Vernick KD. Determinants of Chikungunya and O'nyong-Nyong Virus Specificity for Infection of Aedes and Anopheles Mosquito Vectors. Viruses 2023; 15:589. [PMID: 36992298 PMCID: PMC10051923 DOI: 10.3390/v15030589] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/02/2023] [Accepted: 02/14/2023] [Indexed: 02/23/2023] Open
Abstract
Mosquito-borne diseases caused by viruses and parasites are responsible for more than 700 million infections each year. Anopheles and Aedes are the two major vectors for, respectively, malaria and arboviruses. Anopheles mosquitoes are the primary vector of just one known arbovirus, the alphavirus o'nyong-nyong virus (ONNV), which is closely related to the chikungunya virus (CHIKV), vectored by Aedes mosquitoes. However, Anopheles harbor a complex natural virome of RNA viruses, and a number of pathogenic arboviruses have been isolated from Anopheles mosquitoes in nature. CHIKV and ONNV are in the same antigenic group, the Semliki Forest virus complex, are difficult to distinguish via immunodiagnostic assay, and symptomatically cause essentially the same human disease. The major difference between the arboviruses appears to be their differential use of mosquito vectors. The mechanisms governing this vector specificity are poorly understood. Here, we summarize intrinsic and extrinsic factors that could be associated with vector specificity by these viruses. We highlight the complexity and multifactorial aspect of vectorial specificity of the two alphaviruses, and evaluate the level of risk of vector shift by ONNV or CHIKV.
Collapse
Affiliation(s)
- Solène Cottis
- Genetics and Genomics of Insect Vectors Unit, Department of Parasites and Insect Vectors, Institut Pasteur, Université de Paris Cité, CNRS UMR2000, F-75015 Paris, France
- Graduate School of Life Sciences ED515, Sorbonne Université UPMC Paris VI, 75252 Paris, France
| | - Adrien A. Blisnick
- Arboviruses and Insect Vectors Unit, Department of Virology, Institut Pasteur, Université de Paris Cité, F-75015 Paris, France
| | - Anna-Bella Failloux
- Arboviruses and Insect Vectors Unit, Department of Virology, Institut Pasteur, Université de Paris Cité, F-75015 Paris, France
| | - Kenneth D. Vernick
- Genetics and Genomics of Insect Vectors Unit, Department of Parasites and Insect Vectors, Institut Pasteur, Université de Paris Cité, CNRS UMR2000, F-75015 Paris, France
- Graduate School of Life Sciences ED515, Sorbonne Université UPMC Paris VI, 75252 Paris, France
| |
Collapse
|
26
|
Vinayagam S, Rajendran D, Sekar K, Renu K, Sattu K. The microbiota, the malarial parasite, and the mosquito [MMM] - A three-sided relationship. Mol Biochem Parasitol 2023; 253:111543. [PMID: 36642385 DOI: 10.1016/j.molbiopara.2023.111543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 11/23/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023]
Abstract
The mosquito gut microbiota is vital to the proper functioning of the host organism. Mosquitoes may benefit from this microbiota in their guts because it promotes factors including blood digestion, fecundity, metamorphosis, and living habitat and inhibits malarial parasites (Plasmodium) growth or transmission. In this overview, we analyzed how mosquitoes acquire their gut microbiota, characterized those bacteria, and discussed the functions they provide. We also investigated the effects of microbiota on malaria vectors, with a focus on the mosquito species Anopheles, as well as the relationship between microbiota and Plasmodium, the aspects in which microbiota influences Plasmodium via immune response, metabolism, and redox mechanisms, and the strategies in which gut bacteria affect the life cycle of malaria vectors and provide the ability to resist insecticides. This article explores the difficulties in studying triadic interactions, such as the interplay between Mosquitoes, Malarial parasite, and the Microbiota that dwell in the mosquitoes' guts, and need additional research for a better understanding of these multiple connections to implement an exact vector control strategies using Gut microbiota in malaria control.
Collapse
Affiliation(s)
- Sathishkumar Vinayagam
- Department of Biotechnology, Periyar University, Centre for Postgraduate and Research Studies, Dharmapuri, Tamil Nadu 635205, India
| | - Devianjana Rajendran
- Department of Biotechnology, Periyar University, Centre for Postgraduate and Research Studies, Dharmapuri, Tamil Nadu 635205, India
| | - Kathirvel Sekar
- Department of Biotechnology, Periyar University, Centre for Postgraduate and Research Studies, Dharmapuri, Tamil Nadu 635205, India
| | - Kaviyarasi Renu
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu 600077, India
| | - Kamaraj Sattu
- Department of Biotechnology, Periyar University, Centre for Postgraduate and Research Studies, Dharmapuri, Tamil Nadu 635205, India.
| |
Collapse
|
27
|
Trzebny A, Slodkowicz-Kowalska A, Björkroth J, Dabert M. Microsporidian Infection in Mosquitoes (Culicidae) Is Associated with Gut Microbiome Composition and Predicted Gut Microbiome Functional Content. MICROBIAL ECOLOGY 2023; 85:247-263. [PMID: 34939130 PMCID: PMC9849180 DOI: 10.1007/s00248-021-01944-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 12/10/2021] [Indexed: 06/14/2023]
Abstract
The animal gut microbiota consist of many different microorganisms, mainly bacteria, but archaea, fungi, protozoans, and viruses may also be present. This complex and dynamic community of microorganisms may change during parasitic infection. In the present study, we investigated the effect of the presence of microsporidians on the composition of the mosquito gut microbiota and linked some microbiome taxa and functionalities to infections caused by these parasites. We characterised bacterial communities of 188 mosquito females, of which 108 were positive for microsporidian DNA. To assess how bacterial communities change during microsporidian infection, microbiome structures were identified using 16S rRNA microbial profiling. In total, we identified 46 families and four higher taxa, of which Comamonadaceae, Enterobacteriaceae, Flavobacteriaceae and Pseudomonadaceae were the most abundant mosquito-associated bacterial families. Our data suggest that the mosquito gut microbial composition varies among host species. In addition, we found a correlation between the microbiome composition and the presence of microsporidians. The prediction of metagenome functional content from the 16S rRNA gene sequencing suggests that microsporidian infection is characterised by some bacterial species capable of specific metabolic functions, especially the biosynthesis of ansamycins and vancomycin antibiotics and the pentose phosphate pathway. Moreover, we detected a positive correlation between the presence of microsporidian DNA and bacteria belonging to Spiroplasmataceae and Leuconostocaceae, each represented by a single species, Spiroplasma sp. PL03 and Weissella cf. viridescens, respectively. Additionally, W. cf. viridescens was observed only in microsporidian-infected mosquitoes. More extensive research, including intensive and varied host sampling, as well as determination of metabolic activities based on quantitative methods, should be carried out to confirm our results.
Collapse
Affiliation(s)
- Artur Trzebny
- Molecular Biology Techniques Laboratory, Faculty of Biology, Adam Mickiewicz University, Poznan, Poland.
| | - Anna Slodkowicz-Kowalska
- Department of Biology and Medical Parasitology, Poznan University of Medical Sciences, Poznan, Poland
| | - Johanna Björkroth
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Miroslawa Dabert
- Molecular Biology Techniques Laboratory, Faculty of Biology, Adam Mickiewicz University, Poznan, Poland
| |
Collapse
|
28
|
Habtewold T, Tapanelli S, Masters EKG, Windbichler N, Christophides GK. The circadian clock modulates Anopheles gambiae infection with Plasmodium falciparum. PLoS One 2022; 17:e0278484. [PMID: 36454885 PMCID: PMC9714873 DOI: 10.1371/journal.pone.0278484] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 11/16/2022] [Indexed: 12/03/2022] Open
Abstract
Key behaviours, physiologies and gene expressions in Anopheles mosquitoes impact the transmission of Plasmodium. Such mosquito factors are rhythmic to closely follow diel rhythms. Here, we set to explore the impact of the mosquito circadian rhythm on the tripartite interaction between the vector, the parasite and the midgut microbiota, and investigate how this may affect the parasite infection outcomes. We assess Plasmodium falciparum infection prevalence and intensity, as a proxy for gametocyte infectivity, in Anopheles gambiae mosquitoes that received a gametocyte-containing bloodfeed and measure the abundance of the midgut microbiota at different times of the mosquito rearing light-dark cycle. Gametocyte infectivity is also compared in mosquitoes reared and maintained under a reversed light-dark regime. The effect of the circadian clock on the infection outcome is also investigated through silencing of the CLOCK gene that is central in the regulation of animal circadian rhythms. The results reveal that the A. gambiae circadian cycle plays a key role in the intensity of infection of P. falciparum gametocytes. We show that parasite gametocytes are more infectious during the night-time, where standard membrane feeding assays (SMFAs) at different time points in the mosquito natural circadian rhythm demonstrate that gametocytes are more infectious when ingested at midnight than midday. When mosquitoes were cultured under a reversed light/dark regime, disrupting their natural physiological homeostasis, and infected with P. falciparum at evening hours, the infection intensity and prevalence were significantly decreased. Similar results were obtained in mosquitoes reared under the standard light/dark regime upon silencing of CLOCK, a key regulator of the circadian rhythm, highlighting the importance of the circadian rhythm for the mosquito vectorial capacity. At that time, the mosquito midgut microbiota load is significantly reduced, while the expression of lysozyme C-1 (LYSC-1) is elevated, which is involved in both the immune response and microbiota digestion. We conclude that the tripartite interactions between the mosquito vector, the malaria parasite and the mosquito gut microbiota are finely tuned to support and maintain malaria transmission. Our data add to the knowledge framework required for designing appropriate and biologically relevant SMFA protocols.
Collapse
Affiliation(s)
- Tibebu Habtewold
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Sofia Tapanelli
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Ellen K. G. Masters
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Nikolai Windbichler
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | | |
Collapse
|
29
|
An Overview on the Impact of Microbiota on Malaria Transmission and Severity: Plasmodium-Vector-Host Axis. Acta Parasitol 2022; 67:1471-1486. [PMID: 36264525 DOI: 10.1007/s11686-022-00631-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 10/10/2022] [Indexed: 11/01/2022]
Abstract
PURPOSE Malaria, which is a vector-borne disease caused by Plasmodium sp., continue to become a serious threat, causing more than 600,000 deaths annually, especially in developing countries. Due to the lack of a long-term, and effective vaccine, and an increasing resistance to antimalarials, new strategies are needed for prevention and treatment of malaria. Recently, the impact of microbiota on development and transmission of Plasmodium, and the severity of malaria has only begun to emerge, although its contribution to homeostasis and a wide variety of disorders is well-understood. Further evidence has shown that microbiota of both mosquito and human host play important roles in transmission, progression, and clearance of Plasmodium infection. Furthermore, Plasmodium can cause significant alterations in the host and mosquito gut microbiota, affecting the clinical outcome of malaria. METHODOLOGY In this review, we attempt to summarize results from published studies on the influence of the host microbiota on the outcome of Plasmodium infections in both arthropods and mammalian hosts. CONCLUSION Modifications of microbiota may be an important potential strategy in blocking Plasmodium transmission in vectors and in the diagnosis, treatment, and prevention of malaria in humans in the future.
Collapse
|
30
|
dos Santos NAC, Magi FN, Andrade AO, Bastos ADS, Pereira SDS, Medeiros JF, Araujo MDS. Assessment of antibiotic treatment on Anopheles darlingi survival and susceptibility to Plasmodium vivax. Front Microbiol 2022; 13:971083. [PMID: 36274692 PMCID: PMC9583876 DOI: 10.3389/fmicb.2022.971083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 09/14/2022] [Indexed: 11/13/2022] Open
Abstract
Antibiotic treatment has been used to enhance anopheline susceptibility to Plasmodium infection, because bacterial microbiota play a fundamental role in modulating the vector competence of mosquitoes that transmit Plasmodium parasites. However, few studies have examined the impact of antibiotic treatments on Plasmodium vivax sporogonic development in neotropical anopheline mosquitoes. Herein, we assessed the impact of antibiotic treatment on P. vivax development and survival in Anopheles darlingi, the main vector of malaria in the Amazon region. Female mosquitoes were treated continuously with antibiotics to impact the gut bacterial load and then tested for prevalence, infection intensity, and survival in comparison with untreated mosquitoes. Antibiotic-fed mosquitoes had not dramatic impact on P. vivax development previously observed in P. falciparum. However, antibiotic treatment increases mosquito survival, which is known to increase vectorial capacity. These findings raise questions about the effect of antibiotics on P. vivax development and survival in An. darlingi.
Collapse
Affiliation(s)
- Najara Akira Costa dos Santos
- Postgraduate Program in Experimental Biology, Federal University of Rondonia/Fiocruz Rondonia, Porto Velho, Brazil
- Platform of Production and Infection of Malaria Vectors (PIVEM), Laboratory of Entomology, Fiocruz Rondonia, Porto Velho, Brazil
| | - Felipe Neves Magi
- Platform of Production and Infection of Malaria Vectors (PIVEM), Laboratory of Entomology, Fiocruz Rondonia, Porto Velho, Brazil
| | - Alice Oliveira Andrade
- Platform of Production and Infection of Malaria Vectors (PIVEM), Laboratory of Entomology, Fiocruz Rondonia, Porto Velho, Brazil
| | - Alessandra da Silva Bastos
- Postgraduate Program in Experimental Biology, Federal University of Rondonia/Fiocruz Rondonia, Porto Velho, Brazil
- Platform of Production and Infection of Malaria Vectors (PIVEM), Laboratory of Entomology, Fiocruz Rondonia, Porto Velho, Brazil
| | | | - Jansen Fernandes Medeiros
- Postgraduate Program in Experimental Biology, Federal University of Rondonia/Fiocruz Rondonia, Porto Velho, Brazil
- Platform of Production and Infection of Malaria Vectors (PIVEM), Laboratory of Entomology, Fiocruz Rondonia, Porto Velho, Brazil
| | - Maisa da Silva Araujo
- Platform of Production and Infection of Malaria Vectors (PIVEM), Laboratory of Entomology, Fiocruz Rondonia, Porto Velho, Brazil
- *Correspondence: Maisa da Silva Araujo,
| |
Collapse
|
31
|
Barillas-Mury C, Ribeiro JMC, Valenzuela JG. Understanding pathogen survival and transmission by arthropod vectors to prevent human disease. Science 2022; 377:eabc2757. [PMID: 36173836 DOI: 10.1126/science.abc2757] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Many endemic poverty-associated diseases, such as malaria and leishmaniasis, are transmitted by arthropod vectors. Pathogens must interact with specific molecules in the vector gut, the microbiota, and the vector immune system to survive and be transmitted. The vertebrate host, in turn, is infected when the pathogen and vector-derived factors, such as salivary proteins, are delivered into the skin by a vector bite. Here, we review recent progress in our understanding of the biology of pathogen transmission from the human to the vector and back, from the vector to the host. We also highlight recent advances in the biology of vector-borne disease transmission, which have translated into additional strategies to prevent human disease by either reducing vector populations or by disrupting their ability to transmit pathogens.
Collapse
Affiliation(s)
- Carolina Barillas-Mury
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, MD 20852, USA
| | - José M C Ribeiro
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, MD 20852, USA
| | - Jesus G Valenzuela
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, MD 20852, USA
| |
Collapse
|
32
|
Antonelli P, Duval P, Luis P, Minard G, Valiente Moro C. Reciprocal interactions between anthropogenic stressors and insect microbiota. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:64469-64488. [PMID: 35864395 DOI: 10.1007/s11356-022-21857-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/01/2022] [Indexed: 06/15/2023]
Abstract
Insects play many important roles in nature due to their diversity, ecological role, and impact on agriculture or human health. They are directly influenced by environmental changes and in particular anthropic activities that constitute an important driver of change in the environmental characteristics. Insects face numerous anthropogenic stressors and have evolved various detoxication mechanisms to survive and/or resist to these compounds. Recent studies highligted the pressure exerted by xenobiotics on insect life-cycle and the important role of insect-associated bacterial microbiota in the insect responses to environmental changes. Stressor exposure can have various impacts on the composition and structure of insect microbiota that in turn may influence insect biology. Moreover, bacterial communities associated with insects can be directly or indirectly involved in detoxification processes with the selection of certain microorganisms capable of degrading xenobiotics. Further studies are needed to assess the role of insect-associated microbiota as key contributor to the xenobiotic metabolism and thus as a driver for insect adaptation to polluted habitats.
Collapse
Affiliation(s)
- Pierre Antonelli
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, INRAE, VetAgro Sup, UMR Ecologie Microbienne, 69622, Villeurbanne, France
| | - Pénélope Duval
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, INRAE, VetAgro Sup, UMR Ecologie Microbienne, 69622, Villeurbanne, France
| | - Patricia Luis
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, INRAE, VetAgro Sup, UMR Ecologie Microbienne, 69622, Villeurbanne, France
| | - Guillaume Minard
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, INRAE, VetAgro Sup, UMR Ecologie Microbienne, 69622, Villeurbanne, France
| | - Claire Valiente Moro
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, INRAE, VetAgro Sup, UMR Ecologie Microbienne, 69622, Villeurbanne, France.
| |
Collapse
|
33
|
da Silva H, Oliveira TMP, Sabino EC, Alonso DP, Sallum MAM. Bacterial diversity in Haemagogus leucocelaenus (Diptera: Culicidae) from Vale do Ribeira, São Paulo, Brazil. BMC Microbiol 2022; 22:161. [PMID: 35733096 PMCID: PMC9215073 DOI: 10.1186/s12866-022-02571-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/06/2022] [Indexed: 01/29/2023] Open
Abstract
INTRODUCTION Mosquitoes (Diptera: Culicidae) are vectors that transmit numerous pathogens to humans and other vertebrates. Haemagogus leucocelaenus is a mosquito associated with transmission of yellow fever virus. The insect gut harbors a variety of microorganisms that can live and multiply within it, thus contributing to digestion, nutrition, and development of its host. The composition of bacterial communities in mosquitoes can be influenced by both biotic and abiotic factors. The goal of this study was to investigate the bacterial diversity of Hg. leucocelaenus and verify the differences between the bacterial communities in Hg. leucocelaenus from three different locations in the Atlantic tropical rain forest and southeastern state of São Paulo State, Brazil. RESULTS The phylum Proteobacteria was found in mosquitoes collected from the three selected study sites. More than 50% of the contigs belong to Wolbachia, followed by 5% Swaminathania, and 3% Acinetobacter. The genus Serratia was found in samples from two locations. CONCLUSIONS Wolbachia was reported for the first time in this species and may indicates that the vector competence of the populations of the species can vary along its geographical distribution area. The presence of Serratia might facilitate viral invasion caused by the disruption of the midgut barrier via action of the SmEnhancin protein, which digests the mucins present in the intestinal epithelium.
Collapse
Affiliation(s)
- Herculano da Silva
- Departamento de Epidemiologia, Faculdade de Saúde Pública, Universidade de São Paulo, Av. Dr. Arnaldo 715, São Paulo, SP 01246-904 Brazil
| | - Tatiane M. P. Oliveira
- Departamento de Epidemiologia, Faculdade de Saúde Pública, Universidade de São Paulo, Av. Dr. Arnaldo 715, São Paulo, SP 01246-904 Brazil
| | - Ester C. Sabino
- Departamento de Moléstias Infecciosas e Parasitarias, Instituto de Medicina Tropical da Faculdade de Medicina da Universidade de São Paulo, Av. Dr. Eneas de Carvalho 470, 1º andar, São Paulo, 05403-000 Brazil
| | - Diego Peres Alonso
- Departamento de Epidemiologia, Faculdade de Saúde Pública, Universidade de São Paulo, Av. Dr. Arnaldo 715, São Paulo, SP 01246-904 Brazil
- Biotechnology Institute and Bioscience Institute, Sao Paulo State University (UNESP), Botucatu, 18618-689 Brazil
| | - Maria Anice M. Sallum
- Departamento de Epidemiologia, Faculdade de Saúde Pública, Universidade de São Paulo, Av. Dr. Arnaldo 715, São Paulo, SP 01246-904 Brazil
| |
Collapse
|
34
|
Santos NAC, Andrade AO, Santos TC, Martinez LN, Ferreira AS, Bastos AS, Martins MM, Pontual JDC, Teles CBG, Medeiros JF, Araújo MS. Evaluation of sustainable susceptibility to Plasmodium vivax infection among colonized Anopheles darlingi and Anopheles deaneorum. Malar J 2022; 21:163. [PMID: 35658964 PMCID: PMC9164182 DOI: 10.1186/s12936-022-04204-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 05/25/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The colonization of mosquitoes susceptible to Plasmodium vivax via direct membrane feeding assay (DMFA) has the potential to significantly advance our knowledge of P. vivax biology, vector-parasite interaction and transmission-blocking vaccine research. Anopheles darlingi and Anopheles deaneorum are important vectors of malaria in the Western Brazilian Amazon. Since 2018, well-established colonies of these species have been maintained in order to mass produce mosquitoes destined for P. vivax infection. Plasmodium susceptibility was confirmed when the colonies were established, but susceptibility needs to be maintained for these colonies to remain good models for pathogen transmission. Thus, the susceptibility was assessed of colonized mosquitoes to P. vivax isolates circulating in the Western Amazon. METHODS Laboratory-reared mosquitoes from F10-F25 generations were fed on P. vivax blood isolates via DMFA. Susceptibility was determined by prevalence and intensity of infection as represented by oocyst load seven days after blood feeding, and sporozoite load 14 days after blood feeding. The effect of infection on mosquito survival was evaluated from initial blood feeding until sporogonic development and survival rates were compared between mosquitoes fed on infected and uninfected blood. Correlation was calculated between gametocytaemia and prevalence/intensity of infection, and between oocyst and sporozoite load. RESULTS Significant differences were found in prevalence and intensity of infection between species. Anopheles darlingi showed a higher proportion of infected mosquitoes and higher oocyst and sporozoite intensity than An. deaneorum. Survival analysis showed that An. deaneorum survival decreased drastically until 14 days post infection (dpi). Plasmodium vivax infection decreased survival in both species relative to uninfected mosquitoes. No correlation was observed between gametocytaemia and prevalence/intensity of infection, but oocyst and sporozoite load had a moderate to strong correlation. CONCLUSIONS Colonized An. darlingi make excellent subjects for modelling pathogen transmission. On the other hand, An. deaneorum could serve as a model for immunity studies due the low susceptibility under current colonized conditions. In the application of DMFA, gametocyte density is not a reliable parameter for predicting mosquito infection by P. vivax, but oocyst intensity should be used to schedule sporozoite experiments.
Collapse
Affiliation(s)
- Najara A C Santos
- Plataforma de Produção e Infecção de Vetores da Malária (PIVEM), Laboratório de Entomologia, Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
- Programa de Pós-Graduação em Biologia Experimental, Fundação Universidade Federal de Rondônia, FIOCRUZ Rondônia, Porto Velho, Rondônia, Brazil
| | - Alice O Andrade
- Plataforma de Produção e Infecção de Vetores da Malária (PIVEM), Laboratório de Entomologia, Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
| | - Thais C Santos
- Plataforma de Produção e Infecção de Vetores da Malária (PIVEM), Laboratório de Entomologia, Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
| | - Leandro N Martinez
- Programa de Pós-Graduação em Biologia Experimental, Fundação Universidade Federal de Rondônia, FIOCRUZ Rondônia, Porto Velho, Rondônia, Brazil
- Plataforma de Bioensaios de Malária e Leishmaniose da Fiocruz (PBML), Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
- Instituto Nacional de Epidemiologia na Amazônia Ocidental, INCT-EpiAmO, Porto Velho, Rondônia, Brazil
| | - Amália S Ferreira
- Plataforma de Bioensaios de Malária e Leishmaniose da Fiocruz (PBML), Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
- Instituto Nacional de Epidemiologia na Amazônia Ocidental, INCT-EpiAmO, Porto Velho, Rondônia, Brazil
| | - Alessandra S Bastos
- Plataforma de Produção e Infecção de Vetores da Malária (PIVEM), Laboratório de Entomologia, Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
- Programa de Pós-Graduação em Biologia Experimental, Fundação Universidade Federal de Rondônia, FIOCRUZ Rondônia, Porto Velho, Rondônia, Brazil
| | - Mirilene M Martins
- Plataforma de Produção e Infecção de Vetores da Malária (PIVEM), Laboratório de Entomologia, Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
| | - José D C Pontual
- Plataforma de Produção e Infecção de Vetores da Malária (PIVEM), Laboratório de Entomologia, Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
| | - Carolina B G Teles
- Programa de Pós-Graduação em Biologia Experimental, Fundação Universidade Federal de Rondônia, FIOCRUZ Rondônia, Porto Velho, Rondônia, Brazil
- Plataforma de Bioensaios de Malária e Leishmaniose da Fiocruz (PBML), Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
- Instituto Nacional de Epidemiologia na Amazônia Ocidental, INCT-EpiAmO, Porto Velho, Rondônia, Brazil
| | - Jansen F Medeiros
- Plataforma de Produção e Infecção de Vetores da Malária (PIVEM), Laboratório de Entomologia, Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
- Programa de Pós-Graduação em Biologia Experimental, Fundação Universidade Federal de Rondônia, FIOCRUZ Rondônia, Porto Velho, Rondônia, Brazil
- Instituto Nacional de Epidemiologia na Amazônia Ocidental, INCT-EpiAmO, Porto Velho, Rondônia, Brazil
| | - Maisa S Araújo
- Plataforma de Produção e Infecção de Vetores da Malária (PIVEM), Laboratório de Entomologia, Fiocruz Rondônia, Porto Velho, Rondônia, Brazil.
| |
Collapse
|
35
|
Abstract
Some antimalarial drugs that have lost clinical usefulness have been repurposed for experimental applications. One example is sulfadiazine, an analog of p-aminobenzoic acid (pABA), which inhibits the parasite's folate synthesis pathway to block DNA synthesis. Sulfadiazine treatment of mice infected with Plasmodium yoelii and P. berghei is routinely used to enrich for gametocytes by killing asexual blood-stage parasites, but it is not well known if there are downstream effects on transmission. To determine if there was a significant effect of sulfadiazine exposure upon transmission, we transmitted Plasmodium yoelii (17XNL strain) parasites to Anopheles stephensi mosquitoes and evaluated the prevalence and intensity of infection under different sulfadiazine treatment conditions. We observed that there was a reduction in both the number of mosquitoes that became infected and in the intensity of infection if parasites were exposed to sulfadiazine in the mouse host or mosquito vector. Sulfadiazine treatment could be marginally overcome if mosquitoes were provided fresh pABA. In contrast, we determined that gametocytes exposed to sulfadiazine could develop into morphologically mature ookinetes in vitro, thus sulfadiazine exposure in the host may be reversible if the drug is washed out and the parasites are supplemented with pABA in the culture media. Overall, this indicates that sulfadiazine dampens host-to-vector transmission and that this inhibition can only be partially overcome by exposure to fresh pABA in vivo and in vitro. Because gametocytes are of great interest for developing transmission-blocking interventions, we recommend the use of less disruptive approaches for gametocyte enrichment. IMPORTANCE In this work, we have uncovered a substantial problem with how many studies of the sexual stages of rodent malaria parasites are conducted. Briefly, the isolation of sexual blood-stage Plasmodium parasites, or gametocytes, is essential to study pretransmission and transmission-stage biology of malaria. A routine method for the isolation of this specific stage in rodent-infectious malaria models is drug treatment with sulfadiazine, an antifolate that selectively kills actively replicating asexual blood-stage parasites but not gametocytes. Thus, researchers use this as a convenient way to produce highly enriched gametocyte samples. However, in this work, we describe how this standard drug selection with sulfadiazine not only kills asexual blood-stage parasites but also substantially impacts host-to-vector transmission.
Collapse
|
36
|
Aželytė J, Wu-Chuang A, Žiegytė R, Platonova E, Mateos-Hernandez L, Maye J, Obregon D, Palinauskas V, Cabezas-Cruz A. Anti-Microbiota Vaccine Reduces Avian Malaria Infection Within Mosquito Vectors. Front Immunol 2022; 13:841835. [PMID: 35309317 PMCID: PMC8928750 DOI: 10.3389/fimmu.2022.841835] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/11/2022] [Indexed: 01/03/2023] Open
Abstract
Animal and human pathogens that are transmitted by arthropods are a global concern, particularly those vectored by mosquitoes (e.g., Plasmodium spp. and dengue virus). Vector microbiota may hold the key to vector-borne pathogen control, as mounting evidence suggests that the contributions of the vector microbiota to vector physiology and pathogen life cycle are so relevant that vectorial capacity cannot be understood without considering microbial communities within the vectors. Anti-tick microbiota vaccines targeting commensal bacteria of the vector microbiota alter vector feeding and modulate the taxonomic and functional profiles of vector microbiome, but their impact on vector-borne pathogen development within the vector has not been tested. In this study, we tested whether anti-microbiota vaccination in birds targeting Enterobacteriaceae within mosquito midguts modulates the mosquito microbiota and disrupt Plasmodium relictum development in its natural vector Culex quinquefasciatus. Domestic canaries (Serinus canaria domestica) were experimentally infected with P. relictum and/or immunized with live vaccines containing different strains of Escherichia coli. Immunization of birds induced E. coli-specific antibodies. The midgut microbial communities of mosquitoes fed on Plasmodium-infected and/or E. coli-immunized birds were different from those of mosquitoes fed on control birds. Notably, mosquito midgut microbiota modulation was associated with a significant decrease in the occurrence of P. relictum oocysts and sporozoites in the midguts and salivary glands of C. quinquefasciatus, respectively. A significant reduction in the number of oocysts was also observed. These findings suggest that anti-microbiota vaccines can be used as a novel tool to control malaria transmission and potentially other vector-borne pathogens.
Collapse
Affiliation(s)
- Justė Aželytė
- Nature Research Centre, Akademijos 2, Vilnius, Lithuania
| | - Alejandra Wu-Chuang
- ANSES, INRAE, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratoire de Santé Animale, Maisons-Alfort, France
| | - Rita Žiegytė
- Nature Research Centre, Akademijos 2, Vilnius, Lithuania
| | | | - Lourdes Mateos-Hernandez
- ANSES, INRAE, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratoire de Santé Animale, Maisons-Alfort, France
| | - Jennifer Maye
- SEPPIC Paris La Défense, La Garenne Colombes, France
| | - Dasiel Obregon
- School of Environmental Sciences, University of Guelph, Guelph, ON, Canada
| | | | - Alejandro Cabezas-Cruz
- ANSES, INRAE, Ecole Nationale Vétérinaire d'Alfort, UMR BIPAR, Laboratoire de Santé Animale, Maisons-Alfort, France
| |
Collapse
|
37
|
Coon KL, Hegde S, Hughes GL. Interspecies microbiome transplantation recapitulates microbial acquisition in mosquitoes. MICROBIOME 2022; 10:58. [PMID: 35410630 PMCID: PMC8996512 DOI: 10.1186/s40168-022-01256-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 03/07/2022] [Indexed: 05/13/2023]
Abstract
BACKGROUND Mosquitoes harbor microbial communities that play important roles in their growth, survival, reproduction, and ability to transmit human pathogens. Microbiome transplantation approaches are often used to study host-microbe interactions and identify microbial taxa and assemblages associated with health or disease. However, no such approaches have been developed to manipulate the microbiota of mosquitoes. RESULTS Here, we developed an approach to transfer entire microbial communities between mosquito cohorts. We undertook transfers between (Culex quinquefasciatus to Aedes aegypti) and within (Ae. aegypti to Ae. aegypti) species to validate the approach and determine the number of mosquitoes required to prepare donor microbiota. After the transfer, we monitored mosquito development and microbiota dynamics throughout the life cycle. Typical holometabolous lifestyle-related microbiota structures were observed, with higher dynamics of microbial structures in larval stages, including the larval water, and less diversity in adults. Microbiota diversity in recipient adults was also more similar to the microbiota diversity in donor adults. CONCLUSIONS This study provides the first evidence for successful microbiome transplantation in mosquitoes. Our results highlight the value of such methods for studying mosquito-microbe interactions and lay the foundation for future studies to elucidate the factors underlying microbiota acquisition, assembly, and function in mosquitoes under controlled conditions. Video Abstract.
Collapse
Affiliation(s)
- Kerri L Coon
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, 53706, USA.
| | - Shivanand Hegde
- Departments of Vector Biology and Tropical Disease Biology, Centre for Neglected Topical Disease, Liverpool School of Tropical Medicine, Liverpool, L3 5QA, UK
| | - Grant L Hughes
- Departments of Vector Biology and Tropical Disease Biology, Centre for Neglected Topical Disease, Liverpool School of Tropical Medicine, Liverpool, L3 5QA, UK.
| |
Collapse
|
38
|
Caragata EP, Short SM. Vector microbiota and immunity: modulating arthropod susceptibility to vertebrate pathogens. CURRENT OPINION IN INSECT SCIENCE 2022; 50:100875. [PMID: 35065286 DOI: 10.1016/j.cois.2022.100875] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 01/03/2022] [Accepted: 01/13/2022] [Indexed: 06/14/2023]
Abstract
Arthropods, including mosquitoes, sand flies, tsetse flies, and ticks are vectors of many bacterial, parasitic, and viral pathogens that cause serious disease in humans and animals. Their microbiota, that is, all microorganisms that dwell within their tissues, can impact vector immunity and susceptibility to pathogen infection. Historically, host-pathogen-microbiota interactions have not been well described, with little known about mechanism. In this review, we highlight recent advances in understanding how individual microorganisms and microbial communities interact with vectors and human pathogens, the mechanisms they utilize to achieve these effects, and the potential for exploiting these interactions to control pathogen transmission. These studies fill important knowledge gaps and further our understanding of the roles that the vector microbiota plays in pathogen transmission.
Collapse
Affiliation(s)
- Eric P Caragata
- Florida Medical Entomology Laboratory, Department of Entomology and Nematology, Institute of Food and Agricultural Sciences, University of Florida, Vero Beach, FL 32962, USA
| | - Sarah M Short
- Department of Entomology, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
39
|
Sarma DK, Kumar M, Dhurve J, Pal N, Sharma P, James MM, Das D, Mishra S, Shubham S, Kumawat M, Verma V, Tiwari RR, Nagpal R, Marotta F. Influence of Host Blood Meal Source on Gut Microbiota of Wild Caught Aedes aegypti, a Dominant Arboviral Disease Vector. Microorganisms 2022; 10:332. [PMID: 35208787 PMCID: PMC8880539 DOI: 10.3390/microorganisms10020332] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 02/07/2023] Open
Abstract
Blood feeding is an important behavior of Aedes aegypti, a dominant arboviral disease vector, as it can establish and transmit viruses to humans. Bacteria associated with the mosquito gut can modulate the biological characteristics and behavior of disease vectors. In this study, we characterized the gut microbiota composition of human-blood-fed (HF), non-human-blood-fed (NHF) and non-fed (NF) field-collected Ae. aegypti mosquitoes, using a 16S metagenomic approach, to assess any association of bacterial taxa with the blood-feeding behavior of Ae. aegypti. A significant difference in the microbiota composition between the HF and NF mosquito group was observed. A significant association was observed in the relative abundance of families Rhodobacteraceae, Neisseriaceae and Dermacoccaceae in the HF group in contrast to NF and NHF Ae. aegypti mosquitoes, respectively. At the class level, two classes (Rhodobacterales and Neisseriales) were found to be in higher abundance in the HF mosquitoes compared to a single class of bacteria (Caulobacterales) in the NF mosquitoes. These results show that human-blood feeding may change the gut microbiota in wild Ae. aegypti populations. More research is needed to determine how changes in the midgut bacterial communities in response to human-blood-feeding affect the vectorial capacity of Ae. aegypti.
Collapse
Affiliation(s)
- Devojit Kumar Sarma
- ICMR—National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhouri, Bhopal 462030, Madhya Pradesh, India; (M.K.); (J.D.); (N.P.); (P.S.); (M.M.J.); (D.D.); (S.M.); (S.S.); (M.K.); (R.R.T.)
| | - Manoj Kumar
- ICMR—National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhouri, Bhopal 462030, Madhya Pradesh, India; (M.K.); (J.D.); (N.P.); (P.S.); (M.M.J.); (D.D.); (S.M.); (S.S.); (M.K.); (R.R.T.)
| | - Jigyasa Dhurve
- ICMR—National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhouri, Bhopal 462030, Madhya Pradesh, India; (M.K.); (J.D.); (N.P.); (P.S.); (M.M.J.); (D.D.); (S.M.); (S.S.); (M.K.); (R.R.T.)
| | - Namrata Pal
- ICMR—National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhouri, Bhopal 462030, Madhya Pradesh, India; (M.K.); (J.D.); (N.P.); (P.S.); (M.M.J.); (D.D.); (S.M.); (S.S.); (M.K.); (R.R.T.)
| | - Poonam Sharma
- ICMR—National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhouri, Bhopal 462030, Madhya Pradesh, India; (M.K.); (J.D.); (N.P.); (P.S.); (M.M.J.); (D.D.); (S.M.); (S.S.); (M.K.); (R.R.T.)
| | - Meenu Mariya James
- ICMR—National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhouri, Bhopal 462030, Madhya Pradesh, India; (M.K.); (J.D.); (N.P.); (P.S.); (M.M.J.); (D.D.); (S.M.); (S.S.); (M.K.); (R.R.T.)
| | - Deepanker Das
- ICMR—National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhouri, Bhopal 462030, Madhya Pradesh, India; (M.K.); (J.D.); (N.P.); (P.S.); (M.M.J.); (D.D.); (S.M.); (S.S.); (M.K.); (R.R.T.)
| | - Sweta Mishra
- ICMR—National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhouri, Bhopal 462030, Madhya Pradesh, India; (M.K.); (J.D.); (N.P.); (P.S.); (M.M.J.); (D.D.); (S.M.); (S.S.); (M.K.); (R.R.T.)
| | - Swasti Shubham
- ICMR—National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhouri, Bhopal 462030, Madhya Pradesh, India; (M.K.); (J.D.); (N.P.); (P.S.); (M.M.J.); (D.D.); (S.M.); (S.S.); (M.K.); (R.R.T.)
| | - Manoj Kumawat
- ICMR—National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhouri, Bhopal 462030, Madhya Pradesh, India; (M.K.); (J.D.); (N.P.); (P.S.); (M.M.J.); (D.D.); (S.M.); (S.S.); (M.K.); (R.R.T.)
| | - Vinod Verma
- Stem Cell Research Centre, Department of Hematology, Sanjay Gandhi Post-Graduate Institute of Medical Sciences, Lucknow 226014, Uttar Pradesh, India;
| | - Rajnarayan R. Tiwari
- ICMR—National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhouri, Bhopal 462030, Madhya Pradesh, India; (M.K.); (J.D.); (N.P.); (P.S.); (M.M.J.); (D.D.); (S.M.); (S.S.); (M.K.); (R.R.T.)
| | - Ravinder Nagpal
- Department of Nutrition & Integrative Physiology, College of Health & Human Sciences, Florida State University, Tallahassee, FL 32306, USA;
| | - Francesco Marotta
- ReGenera R&D International for Aging Intervention, 20144 Milano, Lombardia, Italy
| |
Collapse
|
40
|
Maitre A, Wu-Chuang A, Aželytė J, Palinauskas V, Mateos-Hernández L, Obregon D, Hodžić A, Valiente Moro C, Estrada-Peña A, Paoli JC, Falchi A, Cabezas-Cruz A. Vector microbiota manipulation by host antibodies: the forgotten strategy to develop transmission-blocking vaccines. Parasit Vectors 2022; 15:4. [PMID: 34983601 PMCID: PMC8725291 DOI: 10.1186/s13071-021-05122-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 12/11/2021] [Indexed: 01/04/2023] Open
Abstract
Human and animal pathogens that are transmitted by arthropods are a global concern, particularly those vectored by ticks (e.g. Borrelia burgdorferi and tick-borne encephalitis virus) and mosquitoes (e.g. malaria and dengue virus). Breaking the circulation of pathogens in permanent foci by controlling vectors using acaricide-based approaches is threatened by the selection of acaricide resistance in vector populations, poor management practices and relaxing of control measures. Alternative strategies that can reduce vector populations and/or vector-mediated transmission are encouraged worldwide. In recent years, it has become clear that arthropod-associated microbiota are involved in many aspects of host physiology and vector competence, prompting research into vector microbiota manipulation. Here, we review how increased knowledge of microbial ecology and vector-host interactions is driving the emergence of new concepts and tools for vector and pathogen control. We focus on the immune functions of host antibodies taken in the blood meal as they can target pathogens and microbiota bacteria within hematophagous arthropods. Anti-microbiota vaccines are presented as a tool to manipulate the vector microbiota and interfere with the development of pathogens within their vectors. Since the importance of some bacterial taxa for colonization of vector-borne pathogens is well known, the disruption of the vector microbiota by host antibodies opens the possibility to develop novel transmission-blocking vaccines.
Collapse
Affiliation(s)
- Apolline Maitre
- UMR BIPAR, Laboratoire de Santé Animale, Anses, INRAE, Ecole Nationale Vétérinaire d'Alfort, 94700, Maisons-Alfort, France
- INRAE, UR 0045 Laboratoire de Recherches Sur Le Développement de L'Elevage (SELMET-LRDE), 20250, Corte, France
- EA 7310, Laboratoire de Virologie, Université de Corse, Corte, France
| | - Alejandra Wu-Chuang
- UMR BIPAR, Laboratoire de Santé Animale, Anses, INRAE, Ecole Nationale Vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Justė Aželytė
- Nature Research Centre, Akademijos 2, 09412, Vilnius, Lithuania
| | | | - Lourdes Mateos-Hernández
- UMR BIPAR, Laboratoire de Santé Animale, Anses, INRAE, Ecole Nationale Vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Dasiel Obregon
- School of Environmental Sciences, University of Guelph, Guelph, ON, Canada
| | - Adnan Hodžić
- Institute of Parasitology, Department of Pathobiology, University of Veterinary Medicine Vienna, Veterinaerplatz 1, 1210, Vienna, Austria
| | - Claire Valiente Moro
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, INRAE, VetAgro Sup, UMR Ecologie Microbienne, 69622, Villeurbanne, France
| | | | - Jean-Christophe Paoli
- INRAE, UR 0045 Laboratoire de Recherches Sur Le Développement de L'Elevage (SELMET-LRDE), 20250, Corte, France
| | - Alessandra Falchi
- EA 7310, Laboratoire de Virologie, Université de Corse, Corte, France
| | - Alejandro Cabezas-Cruz
- UMR BIPAR, Laboratoire de Santé Animale, Anses, INRAE, Ecole Nationale Vétérinaire d'Alfort, 94700, Maisons-Alfort, France.
| |
Collapse
|
41
|
Fofana A, Gendrin M, Romoli O, Yarbanga GAB, Ouédraogo GA, Yerbanga RS, Ouédraogo JB. Analyzing gut microbiota composition in individual Anopheles mosquitoes after experimental treatment. iScience 2021; 24:103416. [PMID: 34901787 PMCID: PMC8637483 DOI: 10.1016/j.isci.2021.103416] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 08/27/2021] [Accepted: 11/05/2021] [Indexed: 01/04/2023] Open
Abstract
The microbiota of Anopheles mosquitoes influences malaria transmission. Antibiotics ingested during a blood meal impact the mosquito microbiome and malaria transmission, with substantial differences between drugs. Here, we assessed if amoxicillin affects the gut mosquito microbiota. We collected Anopheles larvae in Burkina Faso, kept them in semi-field conditions, and offered a blood meal to adult females. We tested the impact of blood supplementation with two alternative amoxicillin preparations on microbiota composition, determined by high-throughput sequencing in individual gut samples. Our analysis detected four major genera, Elizabethkingia, Wigglesworthia, Asaia, and Serratia. The antibiotic treatment significantly affected overall microbiota composition, with a specific decrease in the relative abundance of Elizabethkingia and Asaia during blood digestion. Besides its interest on the influence of amoxicillin on the mosquito microbiota, our study proposes a thorough approach to report negative-control data of high-throughput sequencing studies on samples with a reduced microbial load.
Collapse
Affiliation(s)
- Aminata Fofana
- Institut de Recherche en Sciences de la Santé, Bobo Dioulasso, Burkina Faso.,Université Nazi Boni, Bobo-Dioulasso 1091, Burkina Faso
| | - Mathilde Gendrin
- Microbiota of Insect Vectors Group, Institut Pasteur de la Guyane, 97306 Cayenne, French Guiana.,Institut Pasteur, Université de Paris, Department of Insect Vectors, 75015 Paris, France
| | - Ottavia Romoli
- Microbiota of Insect Vectors Group, Institut Pasteur de la Guyane, 97306 Cayenne, French Guiana
| | | | | | - Rakiswende Serge Yerbanga
- Institut de Recherche en Sciences de la Santé, Bobo Dioulasso, Burkina Faso.,Institut des Sciences et Techniques, 2779 Bobo Dioulasso, Burkina Faso
| | - Jean-Bosco Ouédraogo
- Institut de Recherche en Sciences de la Santé, Bobo Dioulasso, Burkina Faso.,Institut des Sciences et Techniques, 2779 Bobo Dioulasso, Burkina Faso
| |
Collapse
|
42
|
Tapanelli S, Inghilterra MG, Cai J, Philpott J, Capriotti P, Windbichler N, Christophides GK. Assessment of Plasmodium falciparum Infection and Fitness of Genetically Modified Anopheles gambiae Aimed at Mosquito Population Replacement. FRONTIERS IN TROPICAL DISEASES 2021. [DOI: 10.3389/fitd.2021.806880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Genetically modified (GM) mosquitoes expressing anti-plasmodial effectors propagating through wild mosquito populations by means of gene drive is a promising tool to support current malaria control strategies. The process of generating GM mosquitoes involves genetic transformation of mosquitoes from a laboratory colony and, often, interbreeding with other GM lines to cross in auxiliary traits. These mosquito colonies and GM lines thus often have different genetic backgrounds and GM lines are invariably highly inbred, which in conjunction with their independent rearing in the laboratory may translate to differences in their susceptibility to malaria parasite infection and life history traits. Here, we show that laboratory Anopheles gambiae colonies and GM lines expressing Cas9 and Cre recombinase vary greatly in their susceptibility to Plasmodium falciparum NF54 infection. Therefore, the choice of mosquitoes to be used as a reference when conducting infection or life history trait assays requires careful consideration. To address these issues, we established an experimental pipeline involving genetic crosses and genotyping of mosquitoes reared in shared containers throughout their lifecycle. We used this protocol to examine whether GM lines expressing the antimicrobial peptide (AMP) Scorpine in the mosquito midgut interfere with parasite infection and mosquito survival. We demonstrate that Scorpine expression in the Peritrophin 1 (Aper1) genomic locus reduces both P. falciparum sporozoite prevalence and mosquito lifespan; both these phenotypes are likely to be associated with the disturbance of the midgut microbiota homeostasis. These data lead us to conclude that the Aper1-Sco GM line could be used in proof-of-concept experiments aimed at mosquito population replacement, although the impact of its reduced fitness on the spread of the transgene through wild populations requires further investigation.
Collapse
|
43
|
Knockout of Anopheles stephensi immune gene LRIM1 by CRISPR-Cas9 reveals its unexpected role in reproduction and vector competence. PLoS Pathog 2021; 17:e1009770. [PMID: 34784388 PMCID: PMC8631644 DOI: 10.1371/journal.ppat.1009770] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 11/30/2021] [Accepted: 11/01/2021] [Indexed: 12/27/2022] Open
Abstract
PfSPZ Vaccine against malaria is composed of Plasmodium falciparum (Pf) sporozoites (SPZ) manufactured using aseptically reared Anopheles stephensi mosquitoes. Immune response genes of Anopheles mosquitoes such as Leucin-Rich protein (LRIM1), inhibit Plasmodium SPZ development (sporogony) in mosquitoes by supporting melanization and phagocytosis of ookinetes. With the aim of increasing PfSPZ infection intensities, we generated an A. stephensi LRIM1 knockout line, Δaslrim1, by embryonic genome editing using CRISPR-Cas9. Δaslrim1 mosquitoes had a significantly increased midgut bacterial load and an altered microbiome composition, including elimination of commensal acetic acid bacteria. The alterations in the microbiome caused increased mosquito mortality and unexpectedly, significantly reduced sporogony. The survival rate of Δaslrim1 mosquitoes and their ability to support PfSPZ development, were partially restored by antibiotic treatment of the mosquitoes, and fully restored to baseline when Δaslrim1 mosquitoes were produced aseptically. Deletion of LRIM1 also affected reproductive capacity: oviposition, fecundity and male fertility were significantly compromised. Attenuation in fecundity was not associated with the altered microbiome. This work demonstrates that LRIM1's regulation of the microbiome has a major impact on vector competence and longevity of A. stephensi. Additionally, LRIM1 deletion identified an unexpected role for this gene in fecundity and reduction of sperm transfer by males.
Collapse
|
44
|
Overabundance of Asaia and Serratia Bacteria Is Associated with Deltamethrin Insecticide Susceptibility in Anopheles coluzzii from Agboville, Côte d'Ivoire. Microbiol Spectr 2021; 9:e0015721. [PMID: 34668745 PMCID: PMC8528120 DOI: 10.1128/spectrum.00157-21] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Insecticide resistance among mosquito species is now a pervasive phenomenon that threatens to jeopardize global malaria vector control efforts. Evidence of links between the mosquito microbiota and insecticide resistance is emerging, with significant enrichment of insecticide degrading bacteria and enzymes in resistant populations. Using 16S rRNA amplicon sequencing, we characterized and compared the microbiota of Anopheles coluzzii in relation to their deltamethrin resistance and exposure profiles. Comparisons between 2- and 3-day-old deltamethrin-resistant and -susceptible mosquitoes demonstrated significant differences in microbiota diversity. Ochrobactrum, Lysinibacillus, and Stenotrophomonas genera, each of which comprised insecticide-degrading species, were significantly enriched in resistant mosquitoes. Susceptible mosquitoes had a significant reduction in alpha diversity compared to resistant individuals, with Asaia and Serratia dominating microbial profiles. There was no significant difference in deltamethrin-exposed and -unexposed 5- to 6-day-old individuals, suggesting that insecticide exposure had minimal impact on microbial composition. Serratia and Asaia were also dominant in 5- to 6-day-old mosquitoes, which had reduced microbial diversity compared to 2- to 3-day-old mosquitoes. Our findings revealed significant alterations of Anopheles coluzzii microbiota associated with deltamethrin resistance, highlighting the potential for identification of novel microbial markers for insecticide resistance surveillance. qPCR detection of Serratia and Asaia was consistent with 16S rRNA sequencing, suggesting that population-level field screening of bacterial microbiota may be feasibly integrated into wider resistance monitoring, if reliable and reproducible markers associated with phenotype can be identified. IMPORTANCE Control of insecticide-resistant vector populations remains a significant challenge to global malaria control and while substantial progress has been made elucidating key target site mutations, overexpressed detoxification enzymes and alternate gene families, the contribution of the mosquito microbiota to phenotypic insecticide resistance has been largely overlooked. We focused on determining the effects of deltamethrin resistance intensity on Anopheles coluzzii microbiota and identifying any microbial taxa associated with phenotype. We demonstrated a significant reduction in microbial diversity between deltamethrin-resistant and -susceptible mosquitoes. Insecticide degrading bacterial species belonging to Ochrobactrum, Lysinibacillus, and Stenotrophomonas genera were significantly enriched in resistant mosquitoes, while Asaia and Serratia dominated microbial profiles of susceptible individuals. Our results revealed significant alterations of Anopheles coluzzii microbiota associated with deltamethrin resistance, highlighting the potential for identification of novel microbial markers for surveillance and opportunities for designing innovative control techniques to prevent the further evolution and spread of insecticide resistance.
Collapse
|
45
|
Ernest M, Alves E Silva TL, Vega-Rodríguez J. The best sugar in town for malaria transmission. Trends Parasitol 2021; 37:775-776. [PMID: 34275728 PMCID: PMC11022271 DOI: 10.1016/j.pt.2021.07.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 07/01/2021] [Indexed: 11/18/2022]
Abstract
Anopheles mosquitoes feed on plant nectars as their main source of sugar. Wang et al. show that Asaia bacteria proliferate in the midgut of mosquitoes that feed on glucose or trehalose. Asaia increases the lumenal pH by downregulating mosquito vacuolar ATPase expression, therefore increasing Plasmodium gametogenesis and vector competence.
Collapse
Affiliation(s)
- Medard Ernest
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Thiago Luiz Alves E Silva
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Joel Vega-Rodríguez
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA.
| |
Collapse
|
46
|
Yaméogo KB, Yerbanga RS, Ouattara SB, Yao FA, Lefèvre T, Zongo I, Nikièma F, Compaoré YD, Tinto H, Chandramohan D, Greenwood B, Belem AMG, Cohuet A, Ouédraogo JB. Effect of seasonal malaria chemoprevention plus azithromycin on Plasmodium falciparum transmission: gametocyte infectivity and mosquito fitness. Malar J 2021; 20:326. [PMID: 34315475 PMCID: PMC8314489 DOI: 10.1186/s12936-021-03855-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 07/16/2021] [Indexed: 11/24/2022] Open
Abstract
Background Seasonal malaria chemoprevention (SMC) consists of administration of sulfadoxine-pyrimethamine (SP) + amodiaquine (AQ) at monthly intervals to children during the malaria transmission period. Whether the addition of azithromycin (AZ) to SMC could potentiate the benefit of the intervention was tested through a double-blind, randomized, placebo-controlled trial. The effect of SMC and the addition of AZ, on malaria transmission and on the life history traits of Anopheles gambiae mosquitoes have been investigated. Methods The study included 438 children randomly selected from among participants in the SMC + AZ trial and 198 children from the same area who did not receive chemoprevention. For each participant in the SMC + AZ trial, blood was collected 14 to 21 days post treatment, examined for the presence of malaria sexual and asexual stages and provided as a blood meal to An. gambiae females using a direct membrane-feeding assay. Results The SMC treatment, with or without AZ, significantly reduced the prevalence of asexual Plasmodium falciparum (LRT X22 = 69, P < 0.0001) and the gametocyte prevalence (LRT X22 = 54, P < 0.0001). In addition, the proportion of infectious feeds (LRT X22 = 61, P < 0.0001) and the prevalence of oocysts among exposed mosquitoes (LRT X22 = 22.8, P < 0.001) was reduced when mosquitoes were fed on blood from treated children compared to untreated controls. The addition of AZ to SPAQ was associated with an increased proportion of infectious feeds (LRT X21 = 5.2, P = 0.02), suggesting a significant effect of AZ on gametocyte infectivity. There was a slight negative effect of SPAQ and SPAQ + AZ on mosquito survival compared to mosquitoes fed with blood from control children (LRTX22 = 330, P < 0.0001). Conclusion This study demonstrates that SMC may contribute to a reduction in human to mosquito transmission of P. falciparum, and the reduced mosquito longevity observed for females fed on treated blood may increase the benefit of this intervention in control of malaria. The addition of AZ to SPAQ in SMC appeared to enhance the infectivity of gametocytes providing further evidence that this combination is not an appropriate intervention.
Collapse
Affiliation(s)
- Koudraogo Bienvenue Yaméogo
- Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso. .,Université Nazi Boni, Bobo-Dioulasso, Burkina Faso.
| | - Rakiswendé Serge Yerbanga
- Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso.,Institut des Sciences et Techniques (INSTech Bobo), BP2779, Bobo-Dioulasso, Burkina Faso
| | | | - Franck A Yao
- Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
| | - Thierry Lefèvre
- Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso.,MIVEGEC, University of Montpellier, IRD, CNRS, Montpellier, France.,Laboratoire Mixte International Sur Les Vecteurs (LAMIVECT), Bobo Dioulasso, Burkina Faso.,Centre de Recherche en Écologie et Évolution de la Santé (CREES), Montpellier, France
| | - Issaka Zongo
- Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
| | - Frederic Nikièma
- Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
| | | | - Halidou Tinto
- Institut de Recherche en Sciences de la Santé, Nanoro, Burkina Faso
| | | | | | | | - Anna Cohuet
- MIVEGEC, University of Montpellier, IRD, CNRS, Montpellier, France.,Laboratoire Mixte International Sur Les Vecteurs (LAMIVECT), Bobo Dioulasso, Burkina Faso
| | - Jean Bosco Ouédraogo
- Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso.,Institut des Sciences et Techniques (INSTech Bobo), BP2779, Bobo-Dioulasso, Burkina Faso
| |
Collapse
|
47
|
Lundberg DS, Pramoj Na Ayutthaya P, Strauß A, Shirsekar G, Lo WS, Lahaye T, Weigel D. Host-associated microbe PCR (hamPCR) enables convenient measurement of both microbial load and community composition. eLife 2021; 10:e66186. [PMID: 34292157 PMCID: PMC8387020 DOI: 10.7554/elife.66186] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 07/19/2021] [Indexed: 12/26/2022] Open
Abstract
The ratio of microbial population size relative to the amount of host tissue, or 'microbial load', is a fundamental metric of colonization and infection, but it cannot be directly deduced from microbial amplicon data such as 16S rRNA gene counts. Because existing methods to determine load, such as serial dilution plating, quantitative PCR, and whole metagenome sequencing add substantial cost and/or experimental burden, they are only rarely paired with amplicon sequencing. We introduce host-associated microbe PCR (hamPCR), a robust strategy to both quantify microbial load and describe interkingdom microbial community composition in a single amplicon library. We demonstrate its accuracy across multiple study systems, including nematodes and major crops, and further present a cost-saving technique to reduce host overrepresentation in the library prior to sequencing. Because hamPCR provides an accessible experimental solution to the well-known limitations and statistical challenges of compositional data, it has far-reaching potential in culture-independent microbiology.
Collapse
Affiliation(s)
- Derek S Lundberg
- Department of Molecular Biology, Max Planck Institute for Developmental BiologyTübingenGermany
| | | | - Annett Strauß
- Department of Evolutionary Biology, Max Planck Institute for Developmental BiologyTübingenGermany
| | - Gautam Shirsekar
- Department of Molecular Biology, Max Planck Institute for Developmental BiologyTübingenGermany
| | - Wen-Sui Lo
- ZMBP-General Genetics, University of TübingenTübingenGermany
| | - Thomas Lahaye
- ZMBP-General Genetics, University of TübingenTübingenGermany
| | - Detlef Weigel
- Department of Molecular Biology, Max Planck Institute for Developmental BiologyTübingenGermany
| |
Collapse
|
48
|
The Possible Role of Microorganisms in Mosquito Mass Rearing. INSECTS 2021; 12:insects12070645. [PMID: 34357305 PMCID: PMC8305455 DOI: 10.3390/insects12070645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/07/2021] [Accepted: 07/11/2021] [Indexed: 01/12/2023]
Abstract
Simple Summary One of the most promising control methods against Aedes albopictus is the sterile insect technique, which consists of mass rearing the target species, separation of males from females, and male exposure to sterilizing ionizing radiation. Once released in the environment, the sterile males are expected to search for wild females to mate with. The quality of sterile males is a crucial aspect in SIT programs in order to optimize effectiveness and limit production costs. The integration of probiotic microorganisms in larval and adult mosquito diets could enhance the quality parameters of the released sterile males. Abstract In Europe, one of the most significant mosquitoes of public health importance is Aedes albopictus (Skuse), an allochthonous species of Asian origin. One of the most promising control methods against Aedes albopictus is the sterile insect technique (SIT), which consists of mass rearing the target species, separation of males from females, and male exposure to sterilizing ionizing radiation. Once released in the environment, the sterile males are expected to search for wild females to mate with. If mating occurs, no offspring is produced. The quality of sterile males is a crucial aspect in SIT programs in order to optimize effectiveness and limit production costs. The integration of probiotic microorganisms in larval and adult mosquito diets could enhance the quality parameters of the released sterile males. In this review, we attempt to give the most representative picture of the present knowledge on the relationships between gut microbiota of mosquitoes and the natural or artificial larval diet. Furthermore, the possible use of probiotic microorganisms for mosquito larvae rearing is explored. Based on the limited amount of data found in the literature, we hypothesize that a better understanding of the interaction between mosquitoes and their microbiota may bring significant improvements in mosquito mass rearing for SIT purposes.
Collapse
|
49
|
Medjigbodo AA, Sonounameto EG, Djihinto OY, Abbey E, Salavi EB, Djossou L, Badolo A, Djogbénou LS. Interplay Between Oxytetracycline and the Homozygote kdr (L1014F) Resistance Genotype on Fecundity in Anopheles gambiae (Diptera: Culicidae) Mosquitoes. JOURNAL OF INSECT SCIENCE (ONLINE) 2021; 21:13. [PMID: 34379759 PMCID: PMC8356962 DOI: 10.1093/jisesa/ieab056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Indexed: 06/13/2023]
Abstract
The insecticide resistance in Anopheles gambiae mosquitoes has remained the major threat for vector control programs but the fitness effects conferred by these mechanisms are poorly understood. To fill this knowledge gap, the present study aimed at testing the hypothesis that antibiotic oxytetracycline could have an interaction with insecticide resistance genotypes and consequently inhibit the fecundity in An. gambiae. Four strains of An. gambiae: Kisumu (susceptible), KisKdr (kdr (L1014F) resistant), AcerKis (ace-1 (G119S) resistant) and AcerKdrKis (both kdr (L1014F) and ace-1 (G119S) resistant) were used in this study. The different strains were allowed to bloodfeed on a rabbit previously treated with antibiotic oxytetracycline at a concentration of 39·10-5 M. Three days later, ovarian follicles were dissected from individual mosquito ovaries into physiological saline solution (0.9% NaCl) under a stereomicroscope and the eggs were counted. Fecundity was substantially lower in oxytetracycline-exposed KisKdr females when compared to that of the untreated individuals and oxytetracycline-exposed Kisumu females. The exposed AcerKis females displayed an increased fecundity compared to their nontreated counterparts whereas they had reduced fecundity compared to that of oxytetracycline-exposed Kisumu females. There was no substantial difference between the fecundity in the treated and untreated AcerKdrKis females. The oxytetracycline-exposed AcerKdrKis mosquitoes had an increased fecundity compared to that of the exposed Kisumu females. Our data indicate an indirect effect of oxytetracycline in reducing fecundity of An. gambiae mosquitoes carrying kdrR (L1014F) genotype. These findings could be useful for designing new integrated approaches for malaria vector control in endemic countries.
Collapse
Affiliation(s)
- Adandé A Medjigbodo
- Tropical Infectious Diseases Research Centre (TIDRC), University of Abomey-Calavi, 01BP 526, Cotonou, Benin
- Laboratory of Fundamental and Applied Entomology, University Joseph KI-ZERBO, BP 7021, Ouagadougou 03, Burkina Faso, West Africa
| | - Eric G Sonounameto
- Tropical Infectious Diseases Research Centre (TIDRC), University of Abomey-Calavi, 01BP 526, Cotonou, Benin
| | - Oswald Y Djihinto
- Tropical Infectious Diseases Research Centre (TIDRC), University of Abomey-Calavi, 01BP 526, Cotonou, Benin
| | - Emmanuella Abbey
- Tropical Infectious Diseases Research Centre (TIDRC), University of Abomey-Calavi, 01BP 526, Cotonou, Benin
| | - Esther B Salavi
- Tropical Infectious Diseases Research Centre (TIDRC), University of Abomey-Calavi, 01BP 526, Cotonou, Benin
| | - Laurette Djossou
- Regional Institute of Public Health, University of Abomey-Calavi, BP 384, Ouidah, Benin
| | - Athanase Badolo
- Laboratory of Fundamental and Applied Entomology, University Joseph KI-ZERBO, BP 7021, Ouagadougou 03, Burkina Faso, West Africa
| | - Luc S Djogbénou
- Tropical Infectious Diseases Research Centre (TIDRC), University of Abomey-Calavi, 01BP 526, Cotonou, Benin
- Regional Institute of Public Health, University of Abomey-Calavi, BP 384, Ouidah, Benin
- Department of Vector Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
| |
Collapse
|
50
|
MacLeod HJ, Dimopoulos G, Short SM. Larval Diet Abundance Influences Size and Composition of the Midgut Microbiota of Aedes aegypti Mosquitoes. Front Microbiol 2021; 12:645362. [PMID: 34220739 PMCID: PMC8249813 DOI: 10.3389/fmicb.2021.645362] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 04/19/2021] [Indexed: 11/13/2022] Open
Abstract
The midgut microbiota of the yellow fever mosquito Aedes aegypti impacts pathogen susceptibility and transmission by this important vector species. However, factors influencing the composition and size of the microbiome in mosquitoes are poorly understood. We investigated the impact of larval diet abundance during development on the composition and size of the larval and adult microbiota by rearing Aedes aegypti under four larval food regimens, ranging from nutrient deprivation to nutrient excess. We assessed the persistent impacts of larval diet availability on the microbiota of the larval breeding water, larval mosquitoes, and adult mosquitoes under sugar and blood fed conditions using qPCR and high-throughput 16S amplicon sequencing to determine bacterial load and microbiota composition. Bacterial loads in breeding water increased with increasing larval diet. Larvae reared with the lowest diet abundance had significantly fewer bacteria than larvae from two higher diet treatments, but not from the highest diet abundance. Adults from the lowest diet abundance treatment had significantly fewer bacteria in their midguts compared to all higher diet abundance treatments. Larval diet amount also had a significant impact on microbiota composition, primarily within larval breeding water and larvae. Increasing diet correlated with increased relative levels of Enterobacteriaceae and Flavobacteriaceae and decreased relative levels of Sphingomonadaceae. Multiple individual OTUs were significantly impacted by diet including one mapping to the genus Cedecea, which increased with higher diet amounts. This was consistent across all sample types, including sugar fed and blood fed adults. Taken together, these data suggest that availability of diet during development can cause lasting shifts in the size and composition of the microbiota in the disease vector Aedes aegypti.
Collapse
Affiliation(s)
- Hannah J MacLeod
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Sarah M Short
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|