1
|
Li H, Zhao Q, Xu J, Li X, Chen X, Zhang Y, Li H, Zhu Y, Liu M, Zhao L, Hua D, Zhang X, Chen K. From Biomphalaria glabrata to Drosophila melanogaster and Anopheles gambiae: the diversity and role of FREPs and Dscams in immune response. Front Immunol 2025; 16:1579905. [PMID: 40370466 PMCID: PMC12074976 DOI: 10.3389/fimmu.2025.1579905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 04/10/2025] [Indexed: 05/16/2025] Open
Abstract
Fibrinogen-related proteins (FREPs) and Down syndrome cell adhesion molecules (Dscams) are important immune-related molecules in invertebrates. Although they are found in different taxonomic groups and possess unique functions, both exhibit high diversity and adaptability. FREPs are characterized by their fibrinogen-related domains and have been primarily studied in mollusks, such as Biomphalaria glabrata. Through mechanisms of diversity generation, such as gene conversion and point mutations, BgFREP plays a critical role in the host's defense against parasites. Dscams are immunoglobulin-like transmembrane proteins, mainly studied in arthropods, such as Drosophila melanogaster and Anopheles gambiae. Through alternative splicing, Dscams generate multiple isoforms that participate in pathogen recognition and the precise wiring of neural circuits. In D. melanogaster, DmDscam plays a role not only in neuronal self-recognition but also in pathogen recognition. In A. gambiae, AgDscam defends against parasite infections, by binding to pathogens and mediating phagocytosis. This paper highlights the key roles of FREPs and Dscams in the immunity of two major invertebrate groups-mollusks and arthropods-and summarizes the main advancements in current research. These studies not only deepen the understanding of invertebrate immune mechanisms but also lay a solid foundation for future exploration of their potential applications in the biomedical field.
Collapse
Affiliation(s)
- Hongyu Li
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
- Ocean College, Beibu Gulf University, Qinzhou, China
| | - Qingzhi Zhao
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Jialu Xu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Xianwei Li
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Xintong Chen
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Yijie Zhang
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Hairun Li
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Yunhuan Zhu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Mingcheng Liu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Ling Zhao
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Dingji Hua
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Xiaofen Zhang
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Keda Chen
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| |
Collapse
|
2
|
Onyango SA, Machani MG, Ochwedo KO, Oriango RM, Lee MC, Kokwaro E, Afrane YA, Githeko AK, Zhong D, Yan G. Plasmodium falciparum Pfs47 haplotype compatibility to Anopheles gambiae in Kisumu, a malaria-endemic region of Kenya. Sci Rep 2025; 15:6550. [PMID: 39994226 PMCID: PMC11850800 DOI: 10.1038/s41598-024-84847-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 12/27/2024] [Indexed: 02/26/2025] Open
Abstract
Insecticide resistance and outdoor transmission have reduced the effectiveness of existing malaria transmission prevention strategies. As a result, targeted approaches to support continuing malaria control, such as transmission-blocking vaccines, are required. Cross-sectional mass blood screening in children between 5 and 15 years was conducted in Chulaimbo, Kisumu, during the dry and wet seasons in 2018 and 2019. Plasmodium falciparum gametocyte carriers were identified by Microscopy. Subsequently, carriers were used to feed colony bred Anopheles gambiae females in serum replacement and whole blood membrane feeding experiments. The infection prevalence was 19.7% (95% Cl 0.003-0.007) with 95% of the infections being caused by P. falciparum. Of all confirmed P. falciparum infections, 16.9% were gametocytes. Thirty-seven paired experiments showed infection rates of 0.9% and 0.5% in the serum replacement and whole blood experiments, respectively, with no significant difference (P = 0.738). Six Pfs47 haplotypes were identified from 24 sequenced infectious blood samples: Hap_1 (E27D and L240I), Hap_2 (S98T); Hap_3 (E27D); Hap_4 (L240I); Hap_5 (E188D); and Hap_6 without mutations. Haplotype 4 had the highest frequency of 29.2% followed by Hap_3 and Hap_6 at 20.8% each then Hap_1 with a frequency of 16.7%, whereas Hap_5 and Hap_2 had frequencies of 8.3% and 4.2% respectively. Varying frequencies of Pfs47 haplotypes observed from genetically heterogeneous parasite populations in endemic regions illuminates vector compatibility to refracting P. falciparum using the hypothesized lock and key analogy. This acts as a bottleneck that increases the frequency of P. falciparum haplotypes that escape elimination by vector immune responses. The interaction can be used as a potential target for transmission blocking through a refractory host.
Collapse
Affiliation(s)
- Shirley A Onyango
- School of Zoological Sciences, Kenyatta University, Nairobi, Kenya.
- International Centre of Excellence for Malaria Research, Tom Mboya University, Homa Bay, Kenya.
| | - Maxwell G Machani
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Kevin O Ochwedo
- International Centre of Excellence for Malaria Research, Tom Mboya University, Homa Bay, Kenya
- School of Biological Sciences, University of Nairobi, Nairobi, Kenya
| | - Robin M Oriango
- International Centre of Excellence for Malaria Research, Tom Mboya University, Homa Bay, Kenya
| | - Ming-Chieh Lee
- Program in Public Health, College of Health Sciences, University of California at Irvine, Irvine, CA, 92697, USA
| | | | - Yaw A Afrane
- Department of Medical Microbiology, Medical School, University of Ghana, Accra, Ghana
| | - Andrew K Githeko
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Daibin Zhong
- Program in Public Health, College of Health Sciences, University of California at Irvine, Irvine, CA, 92697, USA.
| | - Guiyun Yan
- Program in Public Health, College of Health Sciences, University of California at Irvine, Irvine, CA, 92697, USA
| |
Collapse
|
3
|
Liu T, Zheng D, Wang J, Li X, Yu S, Liu Z, Zheng F, Zhao C, Yang X, Wang Y. Dihydroartemisinin suppresses the susceptibility of Anopheles stephensi to Plasmodium yoelii by activating the Toll signaling pathway. Parasit Vectors 2024; 17:414. [PMID: 39363238 PMCID: PMC11451267 DOI: 10.1186/s13071-024-06497-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: 06/11/2024] [Accepted: 09/18/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND Malaria is a serious public health concern. Artemisinin and its derivatives are first-line drugs for the treatment of Plasmodium falciparum malaria. In mammals, artemisinin exhibits potent anti-inflammatory and immunoregulatory properties. However, it is unclear whether artemisinin plays a regulatory role in the innate immunity of mosquitoes, thereby affecting the development of Plasmodium in Anopheles when artemisinin and its metabolites enter mosquitoes. This study aims to determine the effect of dihydroartemisinin (DHA), a first-generation semisynthetic derivative of artemisinin, on innate immunity and malaria vector competence of Anopheles stephensi. METHODS Anopheles stephensi was fed Plasmodium-infected mice treated with DHA via gavage, Plasmodium-infected blood containing DHA in vitro, or DHA-containing sugar, followed by Plasmodium yoelii infection. The engorged female mosquitoes were separated and dissected 8 and 17 days after infection. Plasmodium oocysts and sporozoites were counted and compared between the control and DHA-treated groups. Additionally, total RNA and proteins were extracted from engorged mosquitoes 24 and 72 h post infection (hpi). Real-time polymerase chain reaction (PCR) and western blotting were performed to detect the transcriptional levels and protein expression of immune molecules in mosquitoes. Finally, the Toll signaling pathway was inhibited via RNA interference and the infection density was analyzed to confirm the role of the Toll signaling pathway in the effect of DHA on the vector competence of mosquitoes. RESULTS DHA treatment via different approaches significantly reduced the number of Plasmodium oocysts and sporozoites in mosquitoes. The transcriptional levels of anti-Plasmodium immune genes (including TEP1, LRIM1, and APL1C), Toll pathway genes (including Tube, MyD88, and Rel1), and the effector defensin 1 were upregulated by DHA treatment at 24 and 72 hpi. TEP1 and Rel1 protein expression was significantly induced under DHA treatment. However, Rel1 knockdown in DHA-treated mosquitoes abrogated DHA-mediated refractoriness to Plasmodium infection. CONCLUSIONS DHA treatment effectively inhibited the development of P. yoelii in A. stephensi by upregulating mosquitoes' Toll signaling pathway, thereby influencing the susceptibility of Anopheles to Plasmodium.
Collapse
Affiliation(s)
- Tingting Liu
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, No. 30 Gaotanyan St, Shapingba Dis, Chongqing, 400038, China
| | - Dan Zheng
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, No. 30 Gaotanyan St, Shapingba Dis, Chongqing, 400038, China
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China
| | - Jing Wang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, No. 30 Gaotanyan St, Shapingba Dis, Chongqing, 400038, China
| | - Xin Li
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, No. 30 Gaotanyan St, Shapingba Dis, Chongqing, 400038, China
| | - Shasha Yu
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, No. 30 Gaotanyan St, Shapingba Dis, Chongqing, 400038, China
| | - Zhilong Liu
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, No. 30 Gaotanyan St, Shapingba Dis, Chongqing, 400038, China
| | - Feifei Zheng
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, No. 30 Gaotanyan St, Shapingba Dis, Chongqing, 400038, China
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China
| | - Caizhi Zhao
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, No. 30 Gaotanyan St, Shapingba Dis, Chongqing, 400038, China
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China
| | - Xuesen Yang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, No. 30 Gaotanyan St, Shapingba Dis, Chongqing, 400038, China
| | - Ying Wang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, No. 30 Gaotanyan St, Shapingba Dis, Chongqing, 400038, China.
| |
Collapse
|
4
|
Mousavi Shafi ZS, Firouz ZM, Pirahmadi S. Gene expression analysis of Anopheles Meigen, 1818 (Diptera: Culicidae) innate immunity after Plasmodium Marchiafava & Celli, 1885 (Apicomplexa) infection: Toward developing new malaria control strategies. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2024; 123:105650. [PMID: 39089500 DOI: 10.1016/j.meegid.2024.105650] [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: 03/27/2024] [Revised: 07/27/2024] [Accepted: 07/28/2024] [Indexed: 08/04/2024]
Abstract
Despite the critical role of the Anopheles innate immune system in defending against Plasmodium infection, there is still limited information about the key immune mechanisms in Anopheles. This review assesses recent findings on the expression characteristics of immune-related genes in Anopheles following exposure to Plasmodium. A literature review, unrestricted by publication date, was conducted to evaluate immune-related gene expression in different organs of Anopheles after Plasmodium infection. Mosquito immune responses in the midgut are essential for reducing parasite populations. Additionally, innate immune responses in the salivary glands and hemocytes circulating in the hemocoel play key roles in defense against the parasite. Transcriptomic analysis of the mosquito's innate immune response to Plasmodium infection provides valuable insights into key immune mechanisms in mosquito defense. A deeper understanding of immune mechanisms in different organs of Anopheles following Plasmodium infection will aid in discovering critical targets for designing novel control strategies.
Collapse
Affiliation(s)
- Zahra Sadat Mousavi Shafi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Zeinab Mohammadi Firouz
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Sakineh Pirahmadi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
5
|
Liu T, Wang J, Li X, Yu S, Zheng D, Liu Z, Yang X, Wang Y. Human Defensin 5 Inhibits Plasmodium yoelii Development in Anopheles stephensi by Promoting Innate Immune Response. Trop Med Infect Dis 2024; 9:169. [PMID: 39195607 PMCID: PMC11360097 DOI: 10.3390/tropicalmed9080169] [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: 06/03/2024] [Revised: 07/23/2024] [Accepted: 07/23/2024] [Indexed: 08/29/2024] Open
Abstract
Malaria poses a serious threat to human health. Existing vector-based interventions have shortcomings, such as environmental pollution, strong resistance to chemical insecticides, and the slow effects of biological insecticides. Therefore, the need to develop novel strategies for controlling malaria, such as reducing mosquito vector competence, is escalating. Human defensin 5 (HD5) has broad-spectrum antimicrobial activity. To determine its effect on Plasmodium development in mosquitoes, HD5 was injected into Anopheles stephensi at various time points. The infection density of Plasmodium yoelii in An. stephensi was substantially reduced by HD5 treatment administered 24 h prior to infection or 6, 12, or 24 h post-infection (hpi). We found that HD5 treatment upregulated the expression of the innate immune effectors TEP1, MyD88, and Rel1 at 24 and 72 hpi. Furthermore, the RNA interference of MyD88, a key upstream molecule in the Toll signaling pathway, decreased the HD5-induced resistance of mosquitoes against Plasmodium infection. These results suggest that HD5 microinjection inhibits the development of malaria parasites in An. stephensi by activating the Toll signaling pathway.
Collapse
Affiliation(s)
- Tingting Liu
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing 400038, China; (T.L.)
| | - Jing Wang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing 400038, China; (T.L.)
| | - Xin Li
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing 400038, China; (T.L.)
| | - Shasha Yu
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing 400038, China; (T.L.)
| | - Dan Zheng
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing 400038, China; (T.L.)
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 550025, China
| | - Zhilong Liu
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing 400038, China; (T.L.)
| | - Xuesen Yang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing 400038, China; (T.L.)
| | - Ying Wang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing 400038, China; (T.L.)
| |
Collapse
|
6
|
Jiang S, Bao J, Chen Y, Liu Z, Liu R, Cheng Y, Zhang L, Jiang X, Kong H. Immunological regulation by Toll-1 and Spätzle-4 in larval density-dependent prophylaxis of the oriental armyworm, Mythimna separata. Int J Biol Macromol 2024; 264:130778. [PMID: 38467221 DOI: 10.1016/j.ijbiomac.2024.130778] [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: 06/04/2023] [Revised: 02/19/2024] [Accepted: 03/08/2024] [Indexed: 03/13/2024]
Abstract
High population density has been shown to alter insect prophylactic immunity. Toll-Spätzle pathway performs a key function in insect innate immune response. To determine the role of Toll and Spätzle, two main components of Toll-Spätzle pathway, in the density-dependent prophylaxis of Mythimna separata. We identified full-length cDNA encoding the Toll-1 and Spätzle-4 genes in M. separata (designed MsToll-1 and Ms Spätzle-4). Both MsToll-1 and MsSpätzle-4 were expressed throughout all developmental stages. MsToll-1 expression was highly in fat body and brain and MsSpätzle-4 was highly expressed in brain and Malpighian tubule. With increased larval density, MsToll-1 expression was markedly up-regulated. MsSpätzle-4 expression was found to be raised in larvae that were fed in high density (5 and 10 larvae per jar). Co-immunoprecipitation assays demonstrated that MsToll-1 interacted with MsSpätzle-4. Immune-related genes transcriptions were considerably reduced in high-density larvae MsToll-1 (or MsSpätzle-4) was silenced by dsRNA injection. Meanwhile, a discernible reduction in the survival rate of the larvae exposed to Bacillus thuringiensis infection with silence of MsToll-1 (or MsSpätzle-4) was observed. This study implies that prophylactic immunity was influenced by crowded larvae via modulating the Toll-Spätzle pathway in M. separata and allow for a new understanding of into density-dependent prophylaxis in insects.
Collapse
Affiliation(s)
- Suwan Jiang
- College of Plant Protection, Yangzhou University, Wenhui East Road, NO. 48, Yangzhou 225009, PR China
| | - Jianqiang Bao
- College of Plant Protection, Yangzhou University, Wenhui East Road, NO. 48, Yangzhou 225009, PR China
| | - Yuxuan Chen
- College of Plant Protection, Yangzhou University, Wenhui East Road, NO. 48, Yangzhou 225009, PR China
| | - Zhonglin Liu
- College of Plant Protection, Yangzhou University, Wenhui East Road, NO. 48, Yangzhou 225009, PR China
| | - Rui Liu
- College of Plant Protection, Yangzhou University, Wenhui East Road, NO. 48, Yangzhou 225009, PR China
| | - Yunxia Cheng
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, NO. 2 of West Yuanmingyuan Road, Beijing 100193, PR China
| | - Lei Zhang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, NO. 2 of West Yuanmingyuan Road, Beijing 100193, PR China
| | - Xingfu Jiang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, NO. 2 of West Yuanmingyuan Road, Beijing 100193, PR China.
| | - Hailong Kong
- College of Plant Protection, Yangzhou University, Wenhui East Road, NO. 48, Yangzhou 225009, PR China.
| |
Collapse
|
7
|
Li M, Zhou Y, Cheng J, Wang Y, Lan C, Shen Y. Response of the mosquito immune system and symbiotic bacteria to pathogen infection. Parasit Vectors 2024; 17:69. [PMID: 38368353 PMCID: PMC10874582 DOI: 10.1186/s13071-024-06161-4] [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: 12/01/2023] [Accepted: 01/24/2024] [Indexed: 02/19/2024] Open
Abstract
Mosquitoes are the deadliest animal in the word, transmitting a variety of insect-borne infectious diseases, such as malaria, dengue fever, yellow fever, and Zika, causing more deaths than any other vector-borne pathogen. Moreover, in the absence of effective drugs and vaccines to prevent and treat insect-borne diseases, mosquito control is particularly important as the primary measure. In recent decades, due to the gradual increase in mosquito resistance, increasing attention has fallen on the mechanisms and effects associated with pathogen infection. This review provides an overview of mosquito innate immune mechanisms in terms of physical and physiological barriers, pattern recognition receptors, signalling pathways, and cellular and humoral immunity, as well as the antipathogenic effects of mosquito symbiotic bacteria. This review contributes to an in-depth understanding of the interaction process between mosquitoes and pathogens and provides a theoretical basis for biological defence strategies against mosquito-borne infectious diseases.
Collapse
Affiliation(s)
- Manjin Li
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China
| | - Yang Zhou
- Nanjing Medical University, Nanjing, 211166, China
| | - Jin Cheng
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China
| | - Yiqing Wang
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China
| | - Cejie Lan
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China.
| | - Yuan Shen
- The Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, 214023, China.
- Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
8
|
Garrigós M, Ylla G, Martínez-de la Puente J, Figuerola J, Ruiz-López MJ. Two avian Plasmodium species trigger different transcriptional responses on their vector Culex pipiens. Mol Ecol 2023:e17240. [PMID: 38108558 DOI: 10.1111/mec.17240] [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: 06/10/2023] [Revised: 11/01/2023] [Accepted: 11/27/2023] [Indexed: 12/19/2023]
Abstract
Malaria is a mosquito-borne disease caused by protozoans of the genus Plasmodium that affects both humans and wildlife. The fitness consequences of infections by avian malaria are well known in birds, however, little information exists on its impact on mosquitoes. Here we study how Culex pipiens mosquitoes transcriptionally respond to infection by two different Plasmodium species, P. relictum and P. cathemerium, differing in their virulence (mortality rate) and transmissibility (parasite presence in exposed mosquitoes' saliva). We studied the mosquito response to the infection at three critical stages of parasite development: the formation of ookinetes at 24 h post-infection (hpi), the release of sporozoites into the hemocoel at 10 days post-infection (dpi), and the storage of sporozoites in the salivary glands at 21 dpi. For each time point, we characterized the gene expression of mosquitoes infected with each P. relictum and P. cathemerium and mosquitoes fed on an uninfected bird and, subsequently, compared their transcriptomic responses. Differential gene expression analysis showed that most transcriptomic changes occurred during the early infection stage (24 hpi), especially when comparing P. relictum and P. cathemerium-infected mosquitoes. Differentially expressed genes in mosquitoes infected with each species were related mainly to the metabolism of the immune response, trypsin, and other serine-proteases. We conclude that these differences in response may partly play a role in the differential virulence and transmissibility previously observed between P. relictum and P. cathemerium in Cx. pipiens.
Collapse
Affiliation(s)
- Marta Garrigós
- Department of Parasitology, University of Granada, Granada, Spain
| | - Guillem Ylla
- Bioinformatics and Genome Biology Lab, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Josué Martínez-de la Puente
- Department of Parasitology, University of Granada, Granada, Spain
- CIBER Epidemiologia y Salud Pública (CIBERESP), Madrid, Spain
| | - Jordi Figuerola
- CIBER Epidemiologia y Salud Pública (CIBERESP), Madrid, Spain
- Department of Wetland Ecology, Estación Biológica de Doñana, CSIC, Sevilla, Spain
| | - María José Ruiz-López
- CIBER Epidemiologia y Salud Pública (CIBERESP), Madrid, Spain
- Department of Wetland Ecology, Estación Biológica de Doñana, CSIC, Sevilla, Spain
| |
Collapse
|
9
|
Katak RDM, Cintra AM, Burini BC, Marinotti O, Souza-Neto JA, Rocha EM. Biotechnological Potential of Microorganisms for Mosquito Population Control and Reduction in Vector Competence. INSECTS 2023; 14:718. [PMID: 37754686 PMCID: PMC10532289 DOI: 10.3390/insects14090718] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/11/2023] [Accepted: 08/19/2023] [Indexed: 09/28/2023]
Abstract
Mosquitoes transmit pathogens that cause human diseases such as malaria, dengue fever, chikungunya, yellow fever, Zika fever, and filariasis. Biotechnological approaches using microorganisms have a significant potential to control mosquito populations and reduce their vector competence, making them alternatives to synthetic insecticides. Ongoing research has identified many microorganisms that can be used effectively to control mosquito populations and disease transmission. However, the successful implementation of these newly proposed approaches requires a thorough understanding of the multipronged microorganism-mosquito-pathogen-environment interactions. Although much has been achieved in discovering new entomopathogenic microorganisms, antipathogen compounds, and their mechanisms of action, only a few have been turned into viable products for mosquito control. There is a discrepancy between the number of microorganisms with the potential for the development of new insecticides and/or antipathogen products and the actual available products, highlighting the need for investments in the intersection of basic research and biotechnology.
Collapse
Affiliation(s)
- Ricardo de Melo Katak
- Malaria and Dengue Laboratory, Instituto Nacional de Pesquisas da Amazônia-INPA, Manaus 69060-001, AM, Brazil;
| | - Amanda Montezano Cintra
- Multiuser Central Laboratory, Department of Bioprocesses and Biotechnology, School of Agricultural Sciences, São Paulo State University (UNESP), Botucatu 18610-034, SP, Brazil; (A.M.C.); (J.A.S.-N.)
| | - Bianca Correa Burini
- Florida Medical Entomology Laboratory, University of Florida, Vero Beach, FL 32962, USA;
| | - Osvaldo Marinotti
- Department of Biology, Indiana University, Bloomington, IN 47405, USA;
| | - Jayme A. Souza-Neto
- Multiuser Central Laboratory, Department of Bioprocesses and Biotechnology, School of Agricultural Sciences, São Paulo State University (UNESP), Botucatu 18610-034, SP, Brazil; (A.M.C.); (J.A.S.-N.)
| | - Elerson Matos Rocha
- Multiuser Central Laboratory, Department of Bioprocesses and Biotechnology, School of Agricultural Sciences, São Paulo State University (UNESP), Botucatu 18610-034, SP, Brazil; (A.M.C.); (J.A.S.-N.)
| |
Collapse
|
10
|
Lucas ER, Nagi SC, Egyir-Yawson A, Essandoh J, Dadzie S, Chabi J, Djogbénou LS, Medjigbodo AA, Edi CV, Kétoh GK, Koudou BG, Van't Hof AE, Rippon EJ, Pipini D, Harding NJ, Dyer NA, Cerdeira LT, Clarkson CS, Kwiatkowski DP, Miles A, Donnelly MJ, Weetman D. Genome-wide association studies reveal novel loci associated with pyrethroid and organophosphate resistance in Anopheles gambiae and Anopheles coluzzii. Nat Commun 2023; 14:4946. [PMID: 37587104 PMCID: PMC10432508 DOI: 10.1038/s41467-023-40693-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 08/03/2023] [Indexed: 08/18/2023] Open
Abstract
Resistance to insecticides in Anopheles mosquitoes threatens the effectiveness of malaria control, but the genetics of resistance are only partially understood. We performed a large scale multi-country genome-wide association study of resistance to two widely used insecticides: deltamethrin and pirimiphos-methyl, using sequencing data from An. gambiae and An. coluzzii from ten locations in West Africa. Resistance was highly multi-genic, multi-allelic and variable between populations. While the strongest and most consistent association with deltamethrin resistance came from Cyp6aa1, this was based on several independent copy number variants (CNVs) in An. coluzzii, and on a non-CNV haplotype in An. gambiae. For pirimiphos-methyl, signals included Ace1, cytochrome P450s, glutathione S-transferases and the nAChR target site of neonicotinoid insecticides. The regions around Cyp9k1 and the Tep family of immune genes showed evidence of cross-resistance to both insecticides. These locally-varying, multi-allelic patterns highlight the challenges involved in genomic monitoring of resistance, and may form the basis for improved surveillance methods.
Collapse
Affiliation(s)
- Eric R Lucas
- Department of Vector Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK.
| | - Sanjay C Nagi
- Department of Vector Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | | | - John Essandoh
- Department of Biomedical Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Samuel Dadzie
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Joseph Chabi
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Luc S Djogbénou
- Tropical Infectious Diseases Research Centre (TIDRC), Université d'Abomey-Calavi (UAC), 01 B.P. 526, Cotonou, Benin
| | - Adandé A Medjigbodo
- Tropical Infectious Diseases Research Centre (TIDRC), Université d'Abomey-Calavi (UAC), 01 B.P. 526, Cotonou, Benin
| | - Constant V Edi
- Centre Suisse de Recherches Scientifiques en Côte d'Ivoire, 01 BP 1303, Abidjan, Côte d'Ivoire
| | - Guillaume K Kétoh
- Laboratory of Ecology and Ecotoxicology, Department of Zoology, Faculty of Sciences, Université de Lomé, 01 B.P. 1515, Lomé, Togo
| | - Benjamin G Koudou
- Centre Suisse de Recherches Scientifiques en Côte d'Ivoire, 01 BP 1303, Abidjan, Côte d'Ivoire
| | - Arjen E Van't Hof
- Department of Vector Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
- Biology Centre of the Czech Academy of Sciences, Institute of Entomology, Branišovská 31, 370 05, České Budějovice, Czech Republic
| | - Emily J Rippon
- Department of Vector Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | - Dimitra Pipini
- Department of Vector Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | - Nicholas J Harding
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
| | - Naomi A Dyer
- Department of Vector Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | - Louise T Cerdeira
- Department of Vector Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | | | | | - Alistair Miles
- Wellcome Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK
| | - Martin J Donnelly
- Department of Vector Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK.
- Wellcome Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK.
| | - David Weetman
- Department of Vector Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK.
| |
Collapse
|
11
|
Abate A, Hassen J, Dembele L, Menard D, Golassa L. Differential transmissibility to Anopheles arabiensis of Plasmodium vivax gametocytes in patients with diverse Duffy blood group genotypes. Malar J 2023; 22:136. [PMID: 37098534 PMCID: PMC10131423 DOI: 10.1186/s12936-023-04570-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: 11/15/2022] [Accepted: 04/21/2023] [Indexed: 04/27/2023] Open
Abstract
BACKGROUND Measuring risk of malaria transmission is complex, especially in case of Plasmodium vivax. This may be overcome using membrane feeding assays in the field where P. vivax is endemic. However, mosquito-feeding assays are affected by a number of human, parasite and mosquito factors. Here, this study identified the contributions of Duffy blood group status of P. vivax-infected patients as a risk of parasite transmission to mosquitoes. METHODS A membrane feeding assay was conducted on a total of 44 conveniently recruited P. vivax infected patients in Adama city and its surroundings in East Shewa Zone, Oromia region, Ethiopia from October, 2019 to January, 2021. The assay was performed in Adama City administration. Mosquito infection rates were determined by midgut dissections at seven to 8 days post-infection. Duffy genotyping was defined for each of the 44 P. vivax infected patients. RESULTS The infection rate of Anopheles mosquitoes was 32.6% (296/907) with 77.3% proportion of infectious participants (34/44). Infectiousness of participants to Anopheles mosquitoes appeared to be higher among individuals with homozygous Duffy positive blood group (TCT/TCT) than heterozygous (TCT/CCT), but the difference was not statistically significant. The mean oocyst density was significantly higher among mosquitoes fed on blood of participants with FY*B/FY*BES than other genotypes (P = 0.001). CONCLUSION Duffy antigen polymorphisms appears to contribute to transmissibility difference of P. vivax gametocytes to Anopheles mosquitoes, but further studies are required.
Collapse
Affiliation(s)
- Andargie Abate
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia.
- College of Medicine and Health Sciences, Bahir Dar University, Bahir Dar, Ethiopia.
| | - Jifar Hassen
- School of Applied Natural Science, Adama Science and Technology University, Adama, Ethiopia
| | - Laurent Dembele
- Malaria Research and Training Centre (MRTC), Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Bamako, Mali
| | - Didier Menard
- Institut Pasteur, Malaria Genetics and Resistance Unit, Université Paris Cité, INSERM U1201, 75015, Paris, France
- Federation of Translational Medicine, Institute of Parasitology and Tropical Diseases, University of Strasbourg, UR7292 Dynamics of Host-Pathogen Interactions, 67000, Strasbourg, France
- Laboratory of Parasitology and Medical Mycology, Strasbourg University Hospital, Strasbourg, France
| | - Lemu Golassa
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
12
|
García-Longoria L, Ahrén D, Berthomieu A, Kalbskopf V, Rivero A, Hellgren O. Immune gene expression in the mosquito vector Culex quinquefasciatus during an avian malaria infection. Mol Ecol 2023; 32:904-919. [PMID: 36448733 PMCID: PMC10108303 DOI: 10.1111/mec.16799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 12/02/2022]
Abstract
Plasmodium relictum is the most widespread avian malaria parasite in the world. It is listed as one of the 100 most dangerous invasive species, having been responsible for the extinction of several endemic bird species, and the near-demise of several others. Here we present the first transcriptomic study focused on the effect of P. relictum on the immune system of its vector (the mosquito Culex quinquefasciatus) at different times post-infection. We show that over 50% of immune genes identified as being part of the Toll pathway and 30%-40% of the immune genes identified within the Imd pathway are overexpressed during the critical period spanning the parasite's oocyst and sporozoite formation (8-12 days), revealing the crucial role played by both these pathways in this natural mosquito-Plasmodium combination. Comparison of infected mosquitoes with their uninfected counterparts also revealed some unexpected immune RNA expression patterns earlier and later in the infection: significant differences in expression of several immune effectors were observed as early as 30 min after ingestion of the infected blood meal. In addition, in the later stages of the infection (towards the end of the mosquito lifespan), we observed an unexpected increase in immune investment in uninfected, but not in infected, mosquitoes. In conclusion, our work extends the comparative transcriptomic analyses of malaria-infected mosquitoes beyond human and rodent parasites and provides insights into the degree of conservation of immune pathways and into the selective pressures exerted by Plasmodium parasites on their vectors.
Collapse
Affiliation(s)
- Luz García-Longoria
- Department of Anatomy, Cellular Biology and Zoology, University of Extremadura, Badajoz, Spain
| | - Dag Ahrén
- Molecular Ecology and Evolution Lab, Department of Biology, Lund University, Lund, Sweden
| | | | - Victor Kalbskopf
- Molecular Ecology and Evolution Lab, Department of Biology, Lund University, Lund, Sweden
| | - Ana Rivero
- MIVEGEC (CNRS, Université de Montpellier, IRD), Montpellier, France
| | - Olof Hellgren
- Molecular Ecology and Evolution Lab, Department of Biology, Lund University, Lund, Sweden
| |
Collapse
|
13
|
Lucas ER, Nagi SC, Egyir-Yawson A, Essandoh J, Dadzie S, Chabi J, Djogbénou LS, Medjigbodo AA, Edi CV, Ketoh GK, Koudou BG, Van't Hof AE, Rippon EJ, Pipini D, Harding NJ, Dyer NA, Cerdeira LT, Clarkson CS, Kwiatkowski DP, Miles A, Donnelly MJ, Weetman D. Genome-wide association studies reveal novel loci associated with pyrethroid and organophosphate resistance in Anopheles gambiae s.l. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.13.523889. [PMID: 36712022 PMCID: PMC9882144 DOI: 10.1101/2023.01.13.523889] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Resistance to insecticides in Anopheles mosquitoes threatens the effectiveness of the most widespread tools currently used to control malaria. The genetic underpinnings of resistance are still only partially understood, with much of the variance in resistance phenotype left unexplained. We performed a multi-country large scale genome-wide association study of resistance to two insecticides widely used in malaria control: deltamethrin and pirimiphos-methyl. Using a bioassay methodology designed to maximise the phenotypic difference between resistant and susceptible samples, we sequenced 969 phenotyped female An. gambiae and An. coluzzii from ten locations across four countries in West Africa (Benin, Côte d'Ivoire, Ghana and Togo), identifying single nucleotide polymorphisms (SNPs) and copy number variants (CNVs) segregating in the populations. The patterns of resistance association were highly multiallelic and variable between populations, with different genomic regions contributing to resistance, as well as different mutations within a given region. While the strongest and most consistent association with deltamethrin resistance came from the region around Cyp6aa1 , this resistance was based on a combination of several independent CNVs in An. coluzzii , and on a non-CNV bearing haplotype in An. gambiae . Further signals involved a range of cytochrome P450, mitochondrial, and immunity genes. Similarly, for pirimiphos-methyl, while the strongest signal came from the region of Ace1 , more widespread signals included cytochrome P450s, glutathione S-transferases, and a subunit of the nAChR target site of neonicotinoid insecticides. The regions around Cyp9k1 and the Tep family of immune genes were associated with resistance to both insecticide classes, suggesting possible cross-resistance mechanisms. These locally-varying, multigenic and multiallelic patterns highlight the challenges involved in genomic monitoring and surveillance of resistance, and form the basis for improvement of methods used to detect and predict resistance. Based on simulations of resistance variants, we recommend that yet larger scale studies, exceeding 500 phenotyped samples per population, are required to better identify associated genomic regions.
Collapse
Affiliation(s)
- Eric R Lucas
- Department of Vector Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
| | - Sanjay C Nagi
- Department of Vector Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
| | | | - John Essandoh
- Department of Biomedical Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Sam Dadzie
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Joseph Chabi
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Luc S Djogbénou
- Tropical Infectious Diseases Research Centre (TIDRC), Université d'Abomey-Calavi (UAC), 01 B.P. 526, Cotonou, Benin
| | - Adandé A Medjigbodo
- Tropical Infectious Diseases Research Centre (TIDRC), Université d'Abomey-Calavi (UAC), 01 B.P. 526, Cotonou, Benin
| | - Constant V Edi
- Centre Suisse de Recherches Scientifiques en Côte d'Ivoire, 01 BP 1303 Abidjan 01, Côte d'Ivoire
| | - Guillaume K Ketoh
- Laboratory of Ecology and Ecotoxicology, Department of Zoology, Faculty of Sciences, Université de Lomé, 01 B.P: 1515 Lomé 01, Togo
| | - Benjamin G Koudou
- Centre Suisse de Recherches Scientifiques en Côte d'Ivoire, 01 BP 1303 Abidjan 01, Côte d'Ivoire
| | - Arjen E Van't Hof
- Department of Vector Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
- Biology Centre of the Czech Academy of Sciences, Institute of Entomology, Branišovská 31, 370 05 České Budějovice, Czech Republic
| | - Emily J Rippon
- Department of Vector Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
| | - Dimitra Pipini
- Department of Vector Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
| | - Nicholas J Harding
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, United Kingdom
| | - Naomi A Dyer
- Department of Vector Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
| | - Louise T Cerdeira
- Department of Vector Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
| | - Chris S Clarkson
- Wellcome Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | | | - Alistair Miles
- Wellcome Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Martin J Donnelly
- Department of Vector Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
- Wellcome Sanger Institute, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - David Weetman
- Department of Vector Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK
| |
Collapse
|
14
|
Ferreira FC, Videvall E, Seidl CM, Wagner NE, Kilpatrick AM, Fleischer RC, Fonseca DM. Transcriptional response of individual Hawaiian Culex quinquefasciatus mosquitoes to the avian malaria parasite Plasmodium relictum. Malar J 2022; 21:249. [PMID: 36038897 PMCID: PMC9422152 DOI: 10.1186/s12936-022-04271-x] [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: 06/17/2022] [Accepted: 08/16/2022] [Indexed: 11/10/2022] Open
Abstract
Background Plasmodium parasites that cause bird malaria occur in all continents except Antarctica and are primarily transmitted by mosquitoes in the genus Culex. Culex quinquefasciatus, the mosquito vector of avian malaria in Hawaiʻi, became established in the islands in the 1820s. While the deadly effects of malaria on endemic bird species have been documented for many decades, vector-parasite interactions in avian malaria systems are relatively understudied. Methods To evaluate the gene expression response of mosquitoes exposed to a Plasmodium infection intensity known to occur naturally in Hawaiʻi, offspring of wild-collected Hawaiian Cx. quinquefasciatus were fed on a domestic canary infected with a fresh isolate of Plasmodium relictum GRW4 from a wild-caught Hawaiian honeycreeper. Control mosquitoes were fed on an uninfected canary. Transcriptomes of five infected and three uninfected individual mosquitoes were sequenced at each of three stages of the parasite life cycle: 24 h post feeding (hpf) during ookinete invasion; 5 days post feeding (dpf) when oocysts are developing; 10 dpf when sporozoites are released and invade the salivary glands. Results Differential gene expression analyses showed that during ookinete invasion (24 hpf), genes related to oxidoreductase activity and galactose catabolism had lower expression levels in infected mosquitoes compared to controls. Oocyst development (5 dpf) was associated with reduced expression of a gene with a predicted innate immune function. At 10 dpf, infected mosquitoes had reduced expression levels of a serine protease inhibitor, and further studies should assess its role as a Plasmodium agonist in C. quinquefasciatus. Overall, the differential gene expression response of Hawaiian Culex exposed to a Plasmodium infection intensity known to occur naturally in Hawaiʻi was low, but more pronounced during ookinete invasion. Conclusions This is the first analysis of the transcriptional responses of vectors to malaria parasites in non-mammalian systems. Interestingly, few similarities were found between the response of Culex infected with a bird Plasmodium and those reported in Anopheles infected with human Plasmodium. The relatively small transcriptional changes observed in mosquito genes related to immune response and nutrient metabolism support conclusions of low fitness costs often documented in experimental challenges of Culex with avian Plasmodium. Supplementary Information The online version contains supplementary material available at 10.1186/s12936-022-04271-x.
Collapse
Affiliation(s)
- Francisco C Ferreira
- Center for Conservation Genomics, Smithsonian Conservation Biology Institute, Washington, DC, USA. .,Center for Vector Biology, Entomology Department, Rutgers University, New Brunswick, NJ, 08901, USA.
| | - Elin Videvall
- Center for Conservation Genomics, Smithsonian Conservation Biology Institute, Washington, DC, USA.,Department of Ecology, Evolution and Organismal Biology, Brown University, Providence, RI, USA.,Institute at Brown for Environment and Society, Brown University, Providence, RI, USA.,Animal Ecology, Department of Ecology and Genetics, Uppsala University, Uppsala, Sweden
| | - Christa M Seidl
- Department of Ecology and Evolutionary Biology, University of California, Santa Cruz, CA, USA
| | - Nicole E Wagner
- Center for Vector Biology, Entomology Department, Rutgers University, New Brunswick, NJ, 08901, USA
| | - A Marm Kilpatrick
- Department of Ecology and Evolutionary Biology, University of California, Santa Cruz, CA, USA
| | - Robert C Fleischer
- Center for Conservation Genomics, Smithsonian Conservation Biology Institute, Washington, DC, USA
| | - Dina M Fonseca
- Center for Vector Biology, Entomology Department, Rutgers University, New Brunswick, NJ, 08901, USA
| |
Collapse
|
15
|
Gómez-Govea MA, Ramírez-Ahuja MDL, Contreras-Perera Y, Jiménez-Camacho AJ, Ruiz-Ayma G, Villanueva-Segura OK, Trujillo-Rodríguez GDJ, Delgado-Enciso I, Martínez-Fierro ML, Manrique-Saide P, Puerta-Guardo H, Flores-Suárez AE, Ponce-García G, Rodríguez-Sánchez IP. Suppression of Midgut Microbiota Impact Pyrethroid Susceptibility in Aedes aegypti. Front Microbiol 2022; 13:761459. [PMID: 35979482 PMCID: PMC9376455 DOI: 10.3389/fmicb.2022.761459] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 05/16/2022] [Indexed: 11/16/2022] Open
Abstract
Aedes aegypti is a mosquito that transmits viral diseases such as dengue, chikungunya, Zika, and yellow fever. The insect’s microbiota is recognized for regulating several biological processes, including digestion, metabolism, egg production, development, and immune response. However, the role of the bacteria involved in insecticide susceptibility has not been established. Therefore, the objective of this study was to characterize the resident microbiota in a field population of A. aegypti to evaluate its role associated with susceptibility to the insecticides permethrin and deltamethrin. Mosquitoes were fed 10% sucrose mixed with antibiotics and then exposed to insecticides using a diagnostic dose. DNA was extracted, and sequencing of bacterial 16S rRNA was carried out on Illumina® MiSeq™. Proteobacteria (92.4%) and Bacteroidetes (7.6%) were the phyla, which are most abundant in mosquitoes fed with sucrose 10%. After exposure to permethrin, the most abundant bacterial species were Pantoea agglomerans (38.4%) and Pseudomonas azotoformans-fluorescens-synxantha (14.2%). Elizabethkingia meningoseptica (38.4%) and Ps. azotoformans-fluorescens-synxantha (26.1%) were the most abundant after exposure to deltamethrin. Our results showed a decrease in mosquitoes’ survival when exposed to permethrin, while no difference in survival when exposed to deltamethrin when the microbiota was modified. We found that the change in microbiota modifies the response of mosquitoes to permethrin. These results are essential for a better understanding of mosquito physiology in response to insecticides.
Collapse
Affiliation(s)
- Mayra A. Gómez-Govea
- Universidad Autónoma de Nuevo León, Facultad de Ciencias Biológicas, Laboratorio de Fisiología Molecular y Estructural, San Nicolás de los Garza, Mexico
| | - María de Lourdes Ramírez-Ahuja
- Universidad Autónoma de Nuevo León, Facultad de Ciencias Biológicas, Laboratorio de Fisiología Molecular y Estructural, San Nicolás de los Garza, Mexico
| | - Yamili Contreras-Perera
- Unidad Colaborativa de Bioensayos Entomológicos (UCBE) y del Laboratorio de Control Biológico (LCB) para Ae. aegypti, Universidad Autónoma de Yucatán (UADY), Mérida, Mexico
| | - Armando J. Jiménez-Camacho
- Universidad Autónoma de Nuevo León, Facultad de Ciencias Biológicas, Laboratorio de Fisiología Molecular y Estructural, San Nicolás de los Garza, Mexico
| | - Gabriel Ruiz-Ayma
- Universidad Autónoma de Nuevo León, Facultad de Ciencias Biologicas, Laboratorio de Biológía de la Conservación, San Nicolás de los Garza, Mexico
| | - Olga Karina Villanueva-Segura
- Universidad Autónoma de Nuevo León, Facultad de Ciencias Biológicas, Laboratorio de Fisiología Molecular y Estructural, San Nicolás de los Garza, Mexico
| | - Gerardo de Jesús Trujillo-Rodríguez
- Universidad Autónoma de Nuevo León, Facultad de Ciencias Biológicas, Laboratorio de Fisiología Molecular y Estructural, San Nicolás de los Garza, Mexico
| | | | - Margarita L. Martínez-Fierro
- Universidad Autónoma de Zacatecas, Laboratorio de Medicina Molecular, Unidad Académica de Medicina Humana, Zacatecas, Mexico
| | - Pablo Manrique-Saide
- Unidad Colaborativa de Bioensayos Entomológicos (UCBE) y del Laboratorio de Control Biológico (LCB) para Ae. aegypti, Universidad Autónoma de Yucatán (UADY), Mérida, Mexico
| | - Henry Puerta-Guardo
- Unidad Colaborativa de Bioensayos Entomológicos (UCBE) y del Laboratorio de Control Biológico (LCB) para Ae. aegypti, Universidad Autónoma de Yucatán (UADY), Mérida, Mexico
| | - Adriana E. Flores-Suárez
- Universidad Autónoma de Nuevo León, Facultad de Ciencias Biológicas, Departamento de Zoología de Invertebrados, San Nicolás de los Garza, Mexico
| | - Gustavo Ponce-García
- Universidad Autónoma de Nuevo León, Facultad de Ciencias Biológicas, Departamento de Zoología de Invertebrados, San Nicolás de los Garza, Mexico
| | - Iram P. Rodríguez-Sánchez
- Universidad Autónoma de Nuevo León, Facultad de Ciencias Biológicas, Laboratorio de Fisiología Molecular y Estructural, San Nicolás de los Garza, Mexico
- *Correspondence: Iram P. Rodríguez-Sánchez,
| |
Collapse
|
16
|
Dong Y, Dong S, Dizaji NB, Rutkowski N, Pohlenz T, Myles K, Dimopoulos G. The Aedes aegypti siRNA pathway mediates broad-spectrum defense against human pathogenic viruses and modulates antibacterial and antifungal defenses. PLoS Biol 2022; 20:e3001668. [PMID: 35679279 PMCID: PMC9182253 DOI: 10.1371/journal.pbio.3001668] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 05/11/2022] [Indexed: 01/08/2023] Open
Abstract
The mosquito's innate immune system defends against a variety of pathogens, and the conserved siRNA pathway plays a central role in the control of viral infections. Here, we show that transgenic overexpression of Dicer2 (Dcr2) or R2d2 resulted in an accumulation of 21-nucleotide viral sequences that was accompanied by a significant suppression of dengue virus (DENV), Zika virus (ZIKV), and chikungunya virus (CHIKV) replication, thus indicating the broad-spectrum antiviral response mediated by the siRNA pathway that can be applied for the development of novel arbovirus control strategies. Interestingly, overexpression of Dcr2 or R2d2 regulated the mRNA abundance of a variety of antimicrobial immune genes, pointing to additional functions of DCR2 and R2D2 as well as cross-talk between the siRNA pathway and other immune pathways. Accordingly, transgenic overexpression of Dcr2 or R2d2 resulted in a lesser proliferation of the midgut microbiota and increased resistance to bacterial and fungal infections.
Collapse
Affiliation(s)
- Yuemei Dong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Shengzhang Dong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Nahid Borhani Dizaji
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Natalie Rutkowski
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Tyler Pohlenz
- Texas A & M University, Department of Entomology, TAMU College Station, Texas, United States of America
| | - Kevin Myles
- Texas A & M University, Department of Entomology, TAMU College Station, Texas, United States of America
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
17
|
Hendrickx S, Caljon G. The effect of the sugar metabolism on Leishmania infantum promastigotes inside the gut of Lutzomyia longipalpis: A sweet relationship? PLoS Negl Trop Dis 2022; 16:e0010293. [PMID: 35385472 PMCID: PMC8985994 DOI: 10.1371/journal.pntd.0010293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 03/02/2022] [Indexed: 11/26/2022] Open
Abstract
It is well-known that Leishmania parasites can alter the behavior of the sand fly vector in order to increase their transmission potential. However, little is known about the contribution of the infecting host’s blood composition on subsequent sand fly infection and survival. This study focused on the host’s glucose metabolism and the insulin/insulin-like growth factor 1 (IGF-1) pathway as both metabolic processes are known to impact vector-parasite interactions of other protozoa and insect species. The focus of this study was inspired by the observation that the glycemic levels in the blood of infected Syrian golden hamsters inversely correlated to splenic and hepatic parasite burdens. To evaluate the biological impact of these findings on further transmission, Lutzomyia longipalpis sand flies were infected with blood that was artificially supplemented with different physiological concentrations of several monosaccharides, insulin or IGF-1. Normoglycemic levels resulted in transiently higher parasite loads and faster appearance of metacyclics, whereas higher carbohydrate and insulin/IGF-1 levels favored sand fly survival. Although the recorded effects were modest or transient of nature, these observations support the concept that the host blood biochemistry may affect Leishmania transmission and sand fly longevity. Past research on the interaction between the Leishmania parasite and the sand fly vector has revealed that Leishmania is capable of changing vector behavior to favor transmission of parasites in the environment. Little is known about the impact of host blood composition on parasite development inside the vector and on vector survival. Here, we showed that parasite burdens in the spleen and the liver inversely correlated to the serum blood glucose levels of infected animals, which triggered us to further investigate the effect of blood monosaccharides, insulin and insulin-like growth factor 1 (IGF-1) on sand fly infection and survival. We demonstrated that normal serum glucose levels in the initial parasitized blood meal resulted in transiently higher parasite loads and a faster appearance of infectious parasites, whereas higher sugar and insulin/IGF-1 levels favored sand fly survival, which supports the concept that the host blood biochemistry may affect Leishmania transmission and sand fly longevity.
Collapse
Affiliation(s)
- Sarah Hendrickx
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
- * E-mail: (SH); (GC)
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
- * E-mail: (SH); (GC)
| |
Collapse
|
18
|
Dehghan H, Mosa-Kazemi SH, Yakhchali B, Maleki-Ravasan N, Vatandoost H, Oshaghi MA. Evaluation of anti-malaria potency of wild and genetically modified Enterobacter cloacae expressing effector proteins in Anopheles stephensi. Parasit Vectors 2022; 15:63. [PMID: 35183231 PMCID: PMC8858508 DOI: 10.1186/s13071-022-05183-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 01/28/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Malaria is one of the most lethal infectious diseases in tropical and subtropical areas of the world. Paratransgenesis using symbiotic bacteria offers a sustainable and environmentally friendly strategy to combat this disease. In the study reported here, we evaluated the disruption of malaria transmission in the Anopheles stephensi-Plasmodium berghei assemblage using the wild-type (WT) and three modified strains of the insect gut bacterium, Enterobacter cloacae. METHODS The assay was carried out using the E. cloacae dissolvens WT and three engineered strains (expressing green fluorescent protein-defensin (GFP-D), scorpine-HasA (S-HasA) and HasA only, respectively). Cotton wool soaked in a solution of 5% (wt/vol) fructose + red dye (1/50 ml) laced with one of the bacterial strains (1 × 109cells/ml) was placed overnight in cages containing female An. stephensi mosquitoes (age: 3-5 days). Each group of sugar-fed mosquitoes was then starved for 4-6 h, following which time they were allowed to blood-feed on P. berghei-infected mice for 20 min in the dark at 17-20 °C. The blood-fed mosquitoes were kept at 19 ± 1 °C and 80 ± 5% relative humidity, and parasite infection was measured by midgut dissection and oocyst counting 10 days post-infection (dpi). RESULTS Exposure to both WT and genetically modified E. cloacae dissolvens strains significantly (P < 0.0001) disrupted P. berghei development in the midgut of An. stephensi, in comparison with the control group. The mean parasite inhibition of E. cloacaeWT, E. cloacaeHasA, E. cloacaeS-HasA and E. cloacaeGFP-D was measured as 72, 86, 92.5 and 92.8 respectively. CONCLUSIONS The WT and modified strains of E. cloacae have the potential to abolish oocyst development by providing a physical barrier or through the excretion of intrinsic effector molecules. These findings reinforce the case for the use of either WT or genetically modified strains of E. cloacae bacteria as a powerful tool to combat malaria.
Collapse
Affiliation(s)
- Hossein Dehghan
- Department of Public Health, School of Public Health, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Seyed Hassan Mosa-Kazemi
- Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Yakhchali
- Department Industrial and of Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Naseh Maleki-Ravasan
- Malaria and Vector Research Group, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Hassan Vatandoost
- Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Department of Chemical Pollutants and Pesticides, Institute for Environmental Research, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Oshaghi
- Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Oduma CO, Koepfli C. Plasmodium falciparum and Plasmodium vivax Adjust Investment in Transmission in Response to Change in Transmission Intensity: A Review of the Current State of Research. Front Cell Infect Microbiol 2021; 11:786317. [PMID: 34956934 PMCID: PMC8692836 DOI: 10.3389/fcimb.2021.786317] [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: 09/30/2021] [Accepted: 11/19/2021] [Indexed: 12/02/2022] Open
Abstract
Malaria parasites can adjust the proportion of parasites that develop into gametocytes, and thus the probability for human-to-vector transmission, through changes in the gametocyte conversion rate. Understanding the factors that impact the commitment of malaria parasites to transmission is required to design better control interventions. Plasmodium spp. persist across countries with vast differences in transmission intensities, and in sites where transmission is highly seasonal. Mounting evidence shows that Plasmodium spp. adjusts the investment in transmission according to seasonality of vector abundance, and transmission intensity. Various techniques to determine the investment in transmission are available, i.e., short-term culture, where the conversion rate can be measured most directly, genome and transcriptome studies, quantification of mature gametocytes, and mosquito feeding assays. In sites with seasonal transmission, the proportion of gametocytes, their densities and infectivity are higher during the wet season, when vectors are plentiful. When countries with pronounced differences in transmission intensity were compared, the investment in transmission was higher when transmission was low, thus maximizing the parasite’s chances to be transmitted to mosquitoes. Increased transmissibility of residual infections after a successful reduction of malaria transmission levels need to be considered when designing intervention measures.
Collapse
Affiliation(s)
- Colins O Oduma
- Department of Biochemistry and Molecular Biology, Egerton University, Nakuru, Kenya.,Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Cristian Koepfli
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States
| |
Collapse
|
20
|
Wang HC, Wang QH, Bhowmick B, Li YX, Han Q. Functional characterization of two clip domain serine proteases in innate immune responses of Aedes aegypti. Parasit Vectors 2021; 14:584. [PMID: 34819136 PMCID: PMC8611957 DOI: 10.1186/s13071-021-05091-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/08/2021] [Indexed: 11/12/2022] Open
Abstract
Background Clip domain serine proteases (CLIPs), a very diverse group of proteolytic enzymes, play a crucial role in the innate immunity of insects. Innate immune responses are the first line of defense in mosquitoes against the invasion of pathogenic microorganisms. The Toll pathway, immunodeficiency (IMD) pathway and melanization are the main processes of innate immunity in Aedes aegypti. CLIPS are classified into five subfamilies—CLIPA, CLIPB, CLIPC, CLIPD, and CLIPE—based on their sequence specificity and phylogenetic relationships. We report the functional characterization of the genes that code for two CLIPs in Ae. aegypti (Ae): Ae-CLIPB15 and Ae-CLIPB22. Methods Clustal Omega was used for multiple amino acid sequence alignment of Ae-CLIPB15 and Ae-CLIPB22 with different CLIP genes from other insect species. The spatiotemporal expression profiles of Ae-CLIPB15 and Ae-CLIPB22 were examined. We determined whether Ae-CLIPB15 and Ae-CLIPB22 respond to microbial challenge and tissue injury. RNA interference (RNAi) was used to explore the function of Ae-CLIPB15 and Ae-CLIPB22 in the defense of Ae. aegypti against bacterial and fungal infections. The expression levels of nuclear factor kappa B (NF-κB) transcription factors REL1 and REL2 in the Toll pathway and IMD pathway after bacterial infection were investigated. Finally, the change in phenoloxidase (PO) activity in Ae-CLIPB15 and Ae-CLIPB22 knockdown adults was investigated. Results We performed spatiotemporal gene expression profiling of Ae-CLIPB15 and Ae-CLIPB22 genes in Ae. aegypti using quantitative real-time polymerase chain reaction. These genes were expressed in different stages and tissues. The messenger RNA (mRNA) levels for both genes were also up-regulated by Gram-negative bacteria Escherichia coli, Gram-positive bacteria Staphylococcus aureus and fungal Beauveria bassiana infections, as well as in the tissue injury experiments. RNAi-mediated knockdown of Ae-CLIPB15 led to a significant decrease of PO activity in the hemolymph of Ae. aegypti, while other RNAi experiments revealed that both Ae-CLIPB15 and Ae-CLIPB22 were involved in immune defense against bacterial and fungal infections. The mRNA expression of NF-κB transcription factors REL1 and REL2 in the Toll pathway and IMD pathway differed between Ae-CLIPB15 and Ae-CLIPB22 knockdown mosquitoes infected with bacteria and wild type mosquitoes infected with bacteria. Conclusions Our findings suggest that Ae-CLIPB15 and Ae-CLIPB22 play a critical role in mosquito innate immunity, and that they are involved in immune responses to injury and infection. Their regulation of transcription factors and PO activity indicates that they also play a specific role in the regulation of innate immunity. Graphical Abstract ![]()
Collapse
Affiliation(s)
- Hao-Cheng Wang
- Laboratory of Tropical Veterinary Medicine and Vector Biology, School of Life Sciences, Hainan University, Haikou, Hainan, 570228, People's Republic of China.,One Health Institute, Hainan University, Haikou, Hainan, 570228, People's Republic of China
| | - Qiu-Hui Wang
- Laboratory of Tropical Veterinary Medicine and Vector Biology, School of Life Sciences, Hainan University, Haikou, Hainan, 570228, People's Republic of China.,One Health Institute, Hainan University, Haikou, Hainan, 570228, People's Republic of China
| | - Biswajit Bhowmick
- Laboratory of Tropical Veterinary Medicine and Vector Biology, School of Life Sciences, Hainan University, Haikou, Hainan, 570228, People's Republic of China.,One Health Institute, Hainan University, Haikou, Hainan, 570228, People's Republic of China
| | - Yi-Xun Li
- Laboratory of Tropical Veterinary Medicine and Vector Biology, School of Life Sciences, Hainan University, Haikou, Hainan, 570228, People's Republic of China.,One Health Institute, Hainan University, Haikou, Hainan, 570228, People's Republic of China
| | - Qian Han
- Laboratory of Tropical Veterinary Medicine and Vector Biology, School of Life Sciences, Hainan University, Haikou, Hainan, 570228, People's Republic of China. .,One Health Institute, Hainan University, Haikou, Hainan, 570228, People's Republic of China.
| |
Collapse
|
21
|
Singh S, Singh A, Baweja V, Roy A, Chakraborty A, Singh IK. Molecular Rationale of Insect-Microbes Symbiosis-From Insect Behaviour to Mechanism. Microorganisms 2021; 9:microorganisms9122422. [PMID: 34946024 PMCID: PMC8707026 DOI: 10.3390/microorganisms9122422] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/18/2021] [Accepted: 11/21/2021] [Indexed: 12/27/2022] Open
Abstract
Insects nurture a panoply of microbial populations that are often obligatory and exist mutually with their hosts. Symbionts not only impact their host fitness but also shape the trajectory of their phenotype. This co-constructed niche successfully evolved long in the past to mark advanced ecological specialization. The resident microbes regulate insect nutrition by controlling their host plant specialization and immunity. It enhances the host fitness and performance by detoxifying toxins secreted by the predators and abstains them. The profound effect of a microbial population on insect physiology and behaviour is exploited to understand the host–microbial system in diverse taxa. Emergent research of insect-associated microbes has revealed their potential to modulate insect brain functions and, ultimately, control their behaviours, including social interactions. The revelation of the gut microbiota–brain axis has now unravelled insects as a cost-effective potential model to study neurodegenerative disorders and behavioural dysfunctions in humans. This article reviewed our knowledge about the insect–microbial system, an exquisite network of interactions operating between insects and microbes, its mechanistic insight that holds intricate multi-organismal systems in harmony, and its future perspectives. The demystification of molecular networks governing insect–microbial symbiosis will reveal the perplexing behaviours of insects that could be utilized in managing insect pests.
Collapse
Affiliation(s)
- Sujata Singh
- Molecular Biology Research Lab, Department of Zoology, Deshbandhu College, University of Delhi, Kalkaji, New Delhi 110019, India; (S.S.); (V.B.)
- Department of Botany, Hansraj College, University of Delhi, New Delhi 110007, India;
| | - Archana Singh
- Department of Botany, Hansraj College, University of Delhi, New Delhi 110007, India;
| | - Varsha Baweja
- Molecular Biology Research Lab, Department of Zoology, Deshbandhu College, University of Delhi, Kalkaji, New Delhi 110019, India; (S.S.); (V.B.)
- DBC i4 Center, Deshbandhu College, University of Delhi, Kalkaji, New Delhi 110019, India
| | - Amit Roy
- EVA 4.0 Unit, Faculty of Forestry and Wood Sciences, Czech University of Life Sciences, Kamýcká 129, Suchdol, 16521 Prague 6, Czech Republic;
- Excelentní Tým pro Mitigaci (ETM), Faculty of Forestry and Wood Sciences, Czech University of Life Sciences, Kamýcká 129, Suchdol, 16521 Prague 6, Czech Republic
| | - Amrita Chakraborty
- EVA 4.0 Unit, Faculty of Forestry and Wood Sciences, Czech University of Life Sciences, Kamýcká 129, Suchdol, 16521 Prague 6, Czech Republic;
- Correspondence: (A.C.); (I.K.S.)
| | - Indrakant Kumar Singh
- Molecular Biology Research Lab, Department of Zoology, Deshbandhu College, University of Delhi, Kalkaji, New Delhi 110019, India; (S.S.); (V.B.)
- DBC i4 Center, Deshbandhu College, University of Delhi, Kalkaji, New Delhi 110019, India
- Correspondence: (A.C.); (I.K.S.)
| |
Collapse
|
22
|
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
|
23
|
Kwon H, Mohammed M, Franzén O, Ankarklev J, Smith RC. Single-cell analysis of mosquito hemocytes identifies signatures of immune cell subtypes and cell differentiation. eLife 2021; 10:66192. [PMID: 34318744 PMCID: PMC8376254 DOI: 10.7554/elife.66192] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 07/27/2021] [Indexed: 12/16/2022] Open
Abstract
Mosquito immune cells, known as hemocytes, are integral to cellular and humoral responses that limit pathogen survival and mediate immune priming. However, without reliable cell markers and genetic tools, studies of mosquito immune cells have been limited to morphological observations, leaving several aspects of their biology uncharacterized. Here, we use single-cell RNA sequencing (scRNA-seq) to characterize mosquito immune cells, demonstrating an increased complexity to previously defined prohemocyte, oenocytoid, and granulocyte subtypes. Through functional assays relying on phagocytosis, phagocyte depletion, and RNA-FISH experiments, we define markers to accurately distinguish immune cell subtypes and provide evidence for immune cell maturation and differentiation. In addition, gene-silencing experiments demonstrate the importance of lozenge in defining the mosquito oenocytoid cell fate. Together, our scRNA-seq analysis provides an important foundation for future studies of mosquito immune cell biology and a valuable resource for comparative invertebrate immunology.
Collapse
Affiliation(s)
- Hyeogsun Kwon
- Department of Entomology, Iowa State University, Ames, United States
| | - Mubasher Mohammed
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Oscar Franzén
- Integrated Cardio Metabolic Centre, Department of Medicine, Karolinska Institutet, Novum, Huddinge, Sweden
| | - Johan Ankarklev
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden.,Microbial Single Cell Genomics facility, SciLifeLab, Biomedical Center (BMC) Uppsala University, Uppsala, Sweden
| | - Ryan C Smith
- Department of Entomology, Iowa State University, Ames, United States
| |
Collapse
|
24
|
Cannon MV, Bogale HN, Bhalerao D, Keita K, Camara D, Barry Y, Keita M, Coulibaly D, Kone AK, Doumbo OK, Thera MA, Plowe CV, Travassos MA, Irish SR, Yeroshefsky J, Dorothy J, Prendergast B, St. Laurent B, Fritz ML, Serre D. High-throughput detection of eukaryotic parasites and arboviruses in mosquitoes. Biol Open 2021; 10:bio058855. [PMID: 34156069 PMCID: PMC8325944 DOI: 10.1242/bio.058855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 06/01/2021] [Indexed: 11/20/2022] Open
Abstract
Vector-borne pathogens cause many human infectious diseases and are responsible for high mortality and morbidity throughout the world. They can also cause livestock epidemics with dramatic social and economic consequences. Due to its high costs, vector-borne disease surveillance is often limited to current threats, and the investigation of emerging pathogens typically occurs after the reports of clinical cases. Here, we use high-throughput sequencing to detect and identify a wide range of parasites and viruses carried by mosquitoes from Cambodia, Guinea, Mali and the USA. We apply this approach to individual Anopheles mosquitoes as well as pools of mosquitoes captured in traps; and compare the outcomes of this assay when applied to DNA or RNA. We identified known human and animal pathogens and mosquito parasites belonging to a wide range of taxa, as well as DNA sequences from previously uncharacterized organisms. Our results also revealed that analysis of the content of an entire trap could be an efficient approach to monitor and identify rare vector-borne pathogens in large surveillance studies. Overall, we describe a high-throughput and easy-to-customize assay to screen for a wide range of pathogens and efficiently complement current vector-borne disease surveillance approaches.
Collapse
Affiliation(s)
- Matthew V. Cannon
- Institute for Genome Sciences, Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Haikel N. Bogale
- Institute for Genome Sciences, Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Devika Bhalerao
- Institute for Genome Sciences, Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Kalil Keita
- Programme National de Lutte contre le Paludisme, Guinea
| | - Denka Camara
- Programme National de Lutte contre le Paludisme, Guinea
| | - Yaya Barry
- Programme National de Lutte contre le Paludisme, Guinea
| | - Moussa Keita
- Programme National de Lutte contre le Paludisme, Guinea
| | - Drissa Coulibaly
- Malaria Research and Training Center, University Science, Techniques and Technologies of Bamako, Mali
| | - Abdoulaye K. Kone
- Malaria Research and Training Center, University Science, Techniques and Technologies of Bamako, Mali
| | - Ogobara K. Doumbo
- Malaria Research and Training Center, University Science, Techniques and Technologies of Bamako, Mali
| | - Mahamadou A. Thera
- Malaria Research and Training Center, University Science, Techniques and Technologies of Bamako, Mali
| | - Christopher V. Plowe
- Malaria Research Program, Department of Medicine, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Mark A. Travassos
- Malaria Research Program, Department of Medicine, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Seth R. Irish
- U.S. President's Malaria Initiative and Entomology Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Joshua Yeroshefsky
- Department of Entomology, University of Maryland College Park, College Park, MD 20742, USA
| | - Jeannine Dorothy
- Mosquito Control Program, Maryland Department of Agriculture, Annapolis, MD 21401, USA
| | - Brian Prendergast
- Mosquito Control Program, Maryland Department of Agriculture, Annapolis, MD 21401, USA
| | - Brandyce St. Laurent
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, MD 20852, USA
| | - Megan L. Fritz
- Department of Entomology, University of Maryland College Park, College Park, MD 20742, USA
| | - David Serre
- Institute for Genome Sciences, Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
25
|
Wang M, An Y, Gao L, Dong S, Zhou X, Feng Y, Wang P, Dimopoulos G, Tang H, Wang J. Glucose-mediated proliferation of a gut commensal bacterium promotes Plasmodium infection by increasing mosquito midgut pH. Cell Rep 2021; 35:108992. [PMID: 33882310 PMCID: PMC8116483 DOI: 10.1016/j.celrep.2021.108992] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 12/06/2020] [Accepted: 03/24/2021] [Indexed: 12/30/2022] Open
Abstract
Plant-nectar-derived sugar is the major energy source for mosquitoes, but its influence on vector competence for malaria parasites remains unclear. Here, we show that Plasmodium berghei infection of Anopheles stephensi results in global metabolome changes, with the most significant impact on glucose metabolism. Feeding on glucose or trehalose (the main hemolymph sugars) renders the mosquito more susceptible to Plasmodium infection by alkalizing the mosquito midgut. The glucose/trehalose diets promote proliferation of a commensal bacterium, Asaia bogorensis, that remodels glucose metabolism in a way that increases midgut pH, thereby promoting Plasmodium gametogenesis. We also demonstrate that the sugar composition from different natural plant nectars influences A. bogorensis growth, resulting in a greater permissiveness to Plasmodium. Altogether, our results demonstrate that dietary glucose is an important determinant of mosquito vector competency for Plasmodium, further highlighting a key role for mosquito-microbiota interactions in regulating the development of the malaria parasite.
Collapse
Affiliation(s)
- Mengfei Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, PRC; Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai 200438, PRC
| | - Yanpeng An
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, PRC; State Key Laboratory of Genetic Engineering, Zhongshan Hospital and School of Life Sciences, Human Phenome Institute, Metabonomics and Systems Biology Laboratory at Shanghai International Centre for Molecular Phenomics, Fudan University, Shanghai 200438, PRC
| | - Li Gao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, PRC; Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai 200438, PRC
| | - Shengzhang Dong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Xiaofeng Zhou
- Human Phenome Institute, Fudan University, Shanghai 200433, PRC
| | - Yuebiao Feng
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, PRC; Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai 200438, PRC
| | - Penghua Wang
- Department of Immunology, School of Medicine, The University of Connecticut Health Center, Farmington, CT 06030, USA
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Huiru Tang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, PRC; State Key Laboratory of Genetic Engineering, Zhongshan Hospital and School of Life Sciences, Human Phenome Institute, Metabonomics and Systems Biology Laboratory at Shanghai International Centre for Molecular Phenomics, Fudan University, Shanghai 200438, PRC.
| | - Jingwen Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, PRC; Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai 200438, PRC.
| |
Collapse
|
26
|
Response of Leucine-Rich Repeat Domain-Containing Protein in Haemaphysalis longicornis to Babesia microti Infection and Its Ligand Identification. Infect Immun 2021; 89:IAI.00268-20. [PMID: 33593890 DOI: 10.1128/iai.00268-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 02/05/2021] [Indexed: 11/20/2022] Open
Abstract
Haemaphysalis longicornis is a blood-feeding hard tick known for transmitting a variety of pathogens, including Babesia How the parasites in the imbibed blood become anchored in the midgut of ticks is still unknown. Leucine-rich repeat domain (LRR)-containing protein, which is associated with the innate immune reaction and conserved in many species, has been detected in H. longicornis and has previously been indicated in inhibiting the growth of Babesia gibsoni However, the detailed mechanism is unknown. In this study, one of the ligands for LRR from H. longicornis (HlLRR) was identified in Babesia microti, designated BmActin, using glutathione transferase (GST) pulldown experiments and immunofluorescence assays. Moreover, RNA interference of HlLRR led to a decrease in the BmActin mRNA expression in the midgut of fully engorged ticks which fed on B. microti-infected mice. We also found that the expression level of the innate immune molecules in H. longicornis, defensin, antimicrobial peptides (AMPs), and lysozyme, were downregulated after the knockdown of HlLRR. However, subolesin expression was upregulated. These results indicate that HlLRR not only recognizes BmActin but may also modulate innate immunity in ticks to influence Babesia growth, which will further benefit the development of anti-Babesia vaccines or drugs.
Collapse
|
27
|
Gabrieli P, Caccia S, Varotto-Boccazzi I, Arnoldi I, Barbieri G, Comandatore F, Epis S. Mosquito Trilogy: Microbiota, Immunity and Pathogens, and Their Implications for the Control of Disease Transmission. Front Microbiol 2021; 12:630438. [PMID: 33889137 PMCID: PMC8056039 DOI: 10.3389/fmicb.2021.630438] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/02/2021] [Indexed: 11/16/2022] Open
Abstract
In mosquitoes, the interaction between the gut microbiota, the immune system, and the pathogens that these insects transmit to humans and animals is regarded as a key component toward the development of control strategies, aimed at reducing the burden of severe diseases, such as malaria and dengue fever. Indeed, different microorganisms from the mosquito microbiota have been investigated for their ability to affect important traits of the biology of the host insect, related with its survival, development and reproduction. Furthermore, some microorganisms have been shown to modulate the immune response of mosquito females, significantly shaping their vector competence. Here, we will review current knowledge in this field, focusing on i) the complex interaction between the intestinal microbiota and mosquito females defenses, both in the gut and at humoral level; ii) how knowledge on these issues contributes to the development of novel and targeted strategies for the control of mosquito-borne diseases such as the use of paratransgenesis or taking advantage of the relationship between Wolbachia and mosquito hosts. We conclude by providing a brief overview of available knowledge on microbiota-immune system interplay in major insect vectors.
Collapse
Affiliation(s)
- Paolo Gabrieli
- Department of Biosciences and Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", University of Milan, Milan, Italy
| | - Silvia Caccia
- Department of Agricultural Sciences, University of Naples "Federico II", Naples, Italy.,Task Force on Microbiome Studies, University of Naples "Federico II", Naples, Italy
| | - Ilaria Varotto-Boccazzi
- Department of Biosciences and Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", University of Milan, Milan, Italy
| | - Irene Arnoldi
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Giulia Barbieri
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Francesco Comandatore
- "L. Sacco" Department of Biomedical and Clinical Sciences, Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", University of Milan, Milan, Italy
| | - Sara Epis
- Department of Biosciences and Pediatric Clinical Research Center "Romeo ed Enrica Invernizzi", University of Milan, Milan, Italy
| |
Collapse
|
28
|
Kumari S, Chauhan C, Tevatiya S, Singla D, De TD, Sharma P, Thomas T, Rani J, Savargaonkar D, Pandey KC, Pande V, Dixit R. Genetic changes of Plasmodium vivax tempers host tissue-specific responses in Anopheles stephensi. CURRENT RESEARCH IN IMMUNOLOGY 2021; 2:12-22. [PMID: 35492403 PMCID: PMC9040150 DOI: 10.1016/j.crimmu.2021.02.002] [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: 10/13/2020] [Revised: 01/13/2021] [Accepted: 02/14/2021] [Indexed: 02/02/2023] Open
Abstract
Recently, we showed how an early restriction of gut flora proliferation by Plasmodium vivax favors immune-suppression and Plasmodium survival in the gut lumen (Sharma et al., 2020). Here, we asked post gut invasion how P. vivax interacts with individual tissues such as the midgut, hemocyte, and salivary glands, and manages its survival in the mosquito host. Our data from tissue-specific comparative RNA-Seq analysis and extensive temporal/spatial expression profiling of selected mosquito transcripts in the uninfected and P. vivax infected mosquito's tissues indicated that (i) a transient suppression of gut metabolic machinery by early oocysts; (ii) enriched expression of nutritional responsive proteins and immune proteins against late oocysts, together may ensure optimal parasite development and gut homeostasis restoration; (iii) pre-immune activation of hemocyte by early gut-oocysts infection via REL induction (p < 0.003); and altered expression of hemocyte-encoded immune proteins may cause rapid removal of free circulating sporozoites from hemolymph; (iv) while a strong suppression of salivary metabolic activities, and elevated expression of salivary specific secretory, as well as immune proteins together, may favor the long-term storage and survival of invaded sporozoites. Finally, our RNA-Seq-based discovery of 4449 transcripts of Plasmodium vivax origin, and their developmental stage-specific expression modulation in the corresponding infected mosquito tissues, predicts a possible mechanism of mosquito responses evasion by P. vivax. Conclusively, our system-wide RNA-Seq analysis provides the first genetic evidence of direct mosquito-Plasmodium interaction and establishes a functional correlation.
Collapse
Affiliation(s)
- Seena Kumari
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
| | - Charu Chauhan
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
| | - Sanjay Tevatiya
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
| | - Deepak Singla
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
- School of Agricultural Biotechnology, Punjab Agricultural University, Ludhiana, Punjab, India
| | - Tanwee Das De
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
| | - Punita Sharma
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
| | - Tina Thomas
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
| | - Jyoti Rani
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
- Bio and Nanotechnology Department, Guru Jambheshwar University of Science and Technology, Haryana, India
| | - Deepali Savargaonkar
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
| | - Kailash C. Pandey
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
| | - Veena Pande
- Department of Biotechnology, Kumaun University, Nainital, Uttarakhand, India
| | - Rajnikant Dixit
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
| |
Collapse
|
29
|
Kumari S, Chauhan C, Tevatiya S, Singla D, De TD, Sharma P, Thomas T, Rani J, Savargaonkar D, Pandey KC, Pande V, Dixit R. Genetic changes of Plasmodium vivax tempers host tissue-specific responses in Anopheles stephensi. CURRENT RESEARCH IN IMMUNOLOGY 2021. [DOI: https:/doi.org/10.1016/j.crimmu.2021.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
|
30
|
Kumari S, Chauhan C, Tevatiya S, Singla D, De TD, Sharma P, Thomas T, Rani J, Savargaonkar D, Pandey KC, Pande V, Dixit R. Genetic changes of Plasmodium vivax tempers host tissue-specific responses in Anopheles stephensi. CURRENT RESEARCH IN IMMUNOLOGY 2021. [DOI: https://doi.org/10.1016/j.crimmu.2021.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
31
|
Plasmodium's journey through the Anopheles mosquito: A comprehensive review. Biochimie 2020; 181:176-190. [PMID: 33346039 DOI: 10.1016/j.biochi.2020.12.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 12/13/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023]
Abstract
The malaria parasite has an extraordinary ability to evade the immune system due to which the development of a malaria vaccine is a challenging task. Extensive research on malarial infection in the human host particularly during the liver stage has resulted in the discovery of potential candidate vaccines including RTS,S/AS01 and R21. However, complete elimination of malaria would require a holistic multi-component approach. In line with this, under the World Health Organization's PATH Malaria Vaccine Initiative (MVI), the research focus has shifted towards the sexual stages of malaria in the mosquito host. Last two decades of scientific research obtained seminal information regarding the sexual/mosquito stages of the malaria. This updated and comprehensive review would provide the basis for consolidated understanding of cellular, biochemical, molecular and immunological aspects of parasite transmission right from the sexual stage commitment in the human host to the sporozoite delivery back into subsequent vertebrate host by the female Anopheles mosquito.
Collapse
|
32
|
Núñez AI, Esteve-Codina A, Gómez-Garrido J, Brustolin M, Talavera S, Berdugo M, Dabad M, Alioto T, Bensaid A, Busquets N. Alteration in the Culex pipiens transcriptome reveals diverse mechanisms of the mosquito immune system implicated upon Rift Valley fever phlebovirus exposure. PLoS Negl Trop Dis 2020; 14:e0008870. [PMID: 33301456 PMCID: PMC7755283 DOI: 10.1371/journal.pntd.0008870] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 12/22/2020] [Accepted: 10/10/2020] [Indexed: 01/02/2023] Open
Abstract
Rift Valley fever phlebovirus (RVFV) causes an emerging zoonotic disease and is mainly transmitted by Culex and Aedes mosquitoes. While Aedes aegypti-dengue virus (DENV) is the most studied model, less is known about the genes involved in infection-responses in other mosquito-arboviruses pairing. The main objective was to investigate the molecular responses of Cx. pipiens to RVFV exposure focusing mainly on genes implicated in innate immune responses. Mosquitoes were fed with blood spiked with RVFV. The fully-engorged females were pooled at 3 different time points: 2 hours post-exposure (hpe), 3- and 14-days post-exposure (dpe). Pools of mosquitoes fed with non-infected blood were also collected for comparisons. Total RNA from each mosquito pool was subjected to RNA-seq analysis and a de novo transcriptome was constructed. A total of 451 differentially expressed genes (DEG) were identified. Most of the transcriptomic alterations were found at an early infection stage after RVFV exposure. Forty-eight DEG related to immune infection-response were characterized. Most of them were related with the RNAi system, Toll and IMD pathways, ubiquitination pathway and apoptosis. Our findings provide for the first time a comprehensive view on Cx. pipiens-RVFV interactions at the molecular level. The early depletion of RNAi pathway genes at the onset of the RVFV infection would allow viral replication in mosquitoes. While genes from the Toll and IMD immune pathways were altered in response to RVFV none of the DEG were related to the JAK/STAT pathway. The fact that most of the DEG involved in the Ubiquitin-proteasome pathway (UPP) or apoptosis were found at an early stage of infection would suggest that apoptosis plays a regulatory role in infected Cx. pipiens midguts. This study provides a number of target genes that could be used to identify new molecular targets for vector control.
Collapse
Affiliation(s)
- Ana I. Núñez
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Anna Esteve-Codina
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Catalonia, Spain
| | - Jèssica Gómez-Garrido
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Catalonia, Spain
| | - Marco Brustolin
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Spain
- Department of Entomology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Sandra Talavera
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Miguel Berdugo
- Instituto de Biología Evolutiva, Universitat Pompeu i Fabra-CSIC, Dr. Aigüader 88, Barcelona, Spain
| | - Marc Dabad
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Catalonia, Spain
| | - Tyler Alioto
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Catalonia, Spain
- Universitat Pompeu i Fabra (UPF), Barcelona, Catalonia, Spain
| | - Albert Bensaid
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Núria Busquets
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Spain
| |
Collapse
|
33
|
Wang M, Wang J. Glucose transporter GLUT1 influences Plasmodium berghei infection in Anopheles stephensi. Parasit Vectors 2020; 13:285. [PMID: 32503601 PMCID: PMC7275331 DOI: 10.1186/s13071-020-04155-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 05/28/2020] [Indexed: 12/04/2022] Open
Abstract
Background Sugar-feeding provides energy for mosquitoes. Facilitated glucose transporters (GLUTs) are responsible for the uptake of glucose in animals. However, knowledge of GLUTs function in Anopheles spp. is limited. Methods Phylogenetic analysis of GLUTs in Anopheles stephensi was performed by the maximum likelihood and Bayesian inference methods. The spatial and temporal expression patterns of four Asteglut genes were analyzed by qPCR. The function of Asteglut1 was examined using a dsRNA-mediated RNA interference method. Transcriptome analysis was used to investigate the global influence of Asteglut1 on mosquito physiology. Results We identified 4 glut genes, Asteglut1, Asteglutx, Asteglut3 and Asteglut4 in An. stephensi. Asteglut1, Asteglut3 and Asteglut4 were mainly expressed in the midgut. Plasmodium berghei infection differentially regulated the expression of Asteglut genes with significant downregulation of Asteglut1 and Asteglut4, while upregulation of Asteglutx. Only knocking-down Asteglut1 facilitated Plasmodium berghei infection in An. stephensi. This might be due to the accumulation of glucose prior to blood-feeding in dsAsteglut1-treated mosquitoes. Our transcriptome analysis revealed that knockdown of Asteglut1 differentially regulated expression of genes associated with multiple functional clusters, especially those related to detoxification and immunity. The dysregulation of multiple pathways might contribute to the increased P. berghei infection. Conclusions Our study shows that Asteglut1 participates in defense against P. berghei in An. stephensi. The regulation of Asteglut1 on vector competence might through modulating multiple biological processes, such as detoxification and immunity.![]()
Collapse
|
34
|
Li-Byarlay H, Boncristiani H, Howell G, Herman J, Clark L, Strand MK, Tarpy D, Rueppell O. Transcriptomic and Epigenomic Dynamics of Honey Bees in Response to Lethal Viral Infection. Front Genet 2020; 11:566320. [PMID: 33101388 PMCID: PMC7546774 DOI: 10.3389/fgene.2020.566320] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 08/17/2020] [Indexed: 12/28/2022] Open
Abstract
Honey bees (Apis mellifera L.) suffer from many brood pathogens, including viruses. Despite considerable research, the molecular responses and dynamics of honey bee pupae to viral pathogens remain poorly understood. Israeli Acute Paralysis Virus (IAPV) is emerging as a model virus since its association with severe colony losses. Using worker pupae, we studied the transcriptomic and methylomic consequences of IAPV infection over three distinct time points after inoculation. Contrasts of gene expression and 5 mC DNA methylation profiles between IAPV-infected and control individuals at these time points - corresponding to the pre-replicative (5 h), replicative (20 h), and terminal (48 h) phase of infection - indicate that profound immune responses and distinct manipulation of host molecular processes accompany the lethal progression of this virus. We identify the temporal dynamics of the transcriptomic response to with more genes differentially expressed in the replicative and terminal phases than in the pre-replicative phase. However, the number of differentially methylated regions decreased dramatically from the pre-replicative to the replicative and terminal phase. Several cellular pathways experienced hyper- and hypo-methylation in the pre-replicative phase and later dramatically increased in gene expression at the terminal phase, including the MAPK, Jak-STAT, Hippo, mTOR, TGF-beta signaling pathways, ubiquitin mediated proteolysis, and spliceosome. These affected biological functions suggest that adaptive host responses to combat the virus are mixed with viral manipulations of the host to increase its own reproduction, all of which are involved in anti-viral immune response, cell growth, and proliferation. Comparative genomic analyses with other studies of viral infections of honey bees and fruit flies indicated that similar immune pathways are shared. Our results further suggest that dynamic DNA methylation responds to viral infections quickly, regulating subsequent gene activities. Our study provides new insights of molecular mechanisms involved in epigenetic that can serve as foundation for the long-term goal to develop anti-viral strategies for honey bees, the most important commercial pollinator.
Collapse
Affiliation(s)
- Hongmei Li-Byarlay
- Department of Entomology and Plant Pathology, North Carolina State University, Raleigh, NC, United States
| | - Humberto Boncristiani
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC, United States
| | - Gary Howell
- High Performance Cluster, Office of Information Technology, North Carolina State University, Raleigh, NC, United States
| | - Jake Herman
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC, United States
| | - Lindsay Clark
- High Performance Computing in Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Micheline K. Strand
- Army Research Office, Army Research Laboratory, Research Triangle Park, NC, United States
| | - David Tarpy
- Department of Entomology and Plant Pathology, North Carolina State University, Raleigh, NC, United States
- W.M. Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC, United States
| | - Olav Rueppell
- Department of Biology, University of North Carolina at Greensboro, Greensboro, NC, United States
| |
Collapse
|
35
|
Ezemuoka LC, Akorli EA, Aboagye-Antwi F, Akorli J. Mosquito midgut Enterobacter cloacae and Serratia marcescens affect the fitness of adult female Anopheles gambiae s.l. PLoS One 2020; 15:e0238931. [PMID: 32946471 PMCID: PMC7500640 DOI: 10.1371/journal.pone.0238931] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 08/26/2020] [Indexed: 01/03/2023] Open
Abstract
Some bacteria species found in the mosquito midgut have demonstrated their role in interrupting the development of Plasmodium within the midgut of the Anopheles mosquito and have been identified as potential candidates for novel bacteria-mediated disease control. However, to use these bacteria successfully in biocontrol mechanisms their effect on the fitness of the vector into which they have been introduced has to be evaluated. This study investigated the effect of two such bacteria candidates, Enterobacter cloacae and Serratia marcescens, on Anopheles gambiae s.l. fitness. Pupae and larvae of Anopheles gambiae s.l. mosquitoes were collected by dipping method and reared to adults. The effect of these bacteria on mosquito fitness was assessed by reintroducing isolates of each bacteria separately into antibiotic-treated female adult mosquitoes through sugar meal. Wild type (non-antibiotic-treated) mosquitoes and those antibiotic-treated with no bacteria reintroduction were used as controls. The mosquitoes were monitored on longevity/survival, fecundity, hatch rate, and larval survival. The antibiotic-treated adult mosquitoes had reduced life span with median survival of 14 days while the bacteria-reintroduced groups and the wild type survived to day 22 (p< 0.0001). Treatment with Enterobacter and Serratia did not affect the average egg deposition (p>0.05) but they affected hatch rates positively (p = 0.008). There was, however, some evidence that suggests Enterobacter could have a positive effect on larval development (p < 0.0001). With no observed negative effect on survival/longevity of Anopheles gambiae, introducing E. cloacae and S. marcescens in future bacteria-associated control strategies is unlikely to result in mosquitoes that will be outlived by the wild population. This, however, requires evaluations under field conditions.
Collapse
Affiliation(s)
- Lilian Chiamaka Ezemuoka
- African Regional Postgraduate Programme in Insect Science (ARPPIS), University of Ghana, Legon, Accra, Ghana
| | - Esinam Abla Akorli
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Accra, Ghana
| | - Fred Aboagye-Antwi
- Department of Animal Biology and Conservation Science, University of Ghana, Legon, Accra, Ghana
| | - Jewelna Akorli
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Accra, Ghana
- West African Centre for Cell Biology of Infectious Diseases, University of Ghana, Legon, Accra, Ghana
| |
Collapse
|
36
|
Yu S, Wang P, Qin J, Zheng H, Wang J, Liu T, Yang X, Wang Y. Bacillus sphaericus exposure reduced vector competence of Anopheles dirus to Plasmodium yoelii by upregulating the Imd signaling pathway. Parasit Vectors 2020; 13:446. [PMID: 32891162 PMCID: PMC7487769 DOI: 10.1186/s13071-020-04321-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/30/2020] [Indexed: 12/05/2022] Open
Abstract
Background Vector control with Bacillus sphaericus (Bs) is an effective way to block the transmission of malaria. However, in practical application of Bs agents, a sublethal dose effect was often caused by insufficient dosing, and it is little known whether the Bs exposure would affect the surviving mosquitoes’ vector capacity to malaria. Methods A sublethal dose of the Bs 2362 strain was administrated to the early fourth-instar larvae of Anopheles dirus to simulate shortage use of Bs in field circumstance. To determine vector competence, mosquitoes were dissected and the oocysts in the midguts were examined on day 9–11 post-infection with Plasmodium yoelii. Meanwhile, a SYBR quantitative PCR assay was conducted to examine the transcriptional level of the key immune molecules of mosquitoes, and RNA interference was utilized to validate the role of key immune effector molecule TEP1. Results The sublethal dose of Bs treatment significantly reduced susceptibility of An. dirus to P. yoelii, with the decrease of P. yoelii infection intensity and rate. Although there existed a melanization response of adult An. dirus following challenge with P. yoelii, it was not involved in the decrease of vector competence as no significant difference of melanization rates and densities between the control and Bs groups was found. Further studies showed that Bs treatment significantly increased TEP1 expression in the fourth-instar larvae (L4), pupae (Pu), 48 h post-infection (hpi) and 72 hpi (P < 0.001). Further, gene-silencing of TEP1 resulted in disappearance of the Bs impact on vector competence of An. dirus to P. yoelii. Moreover, the transcriptional level of PGRP-LC and Rel2 were significantly elevated by Bs treatment with decreased expression of the negative regulator Caspar at 48 hpi, which implied that the Imd signaling pathway was upregulated by Bs exposure. Conclusions Bs exposure can reduce the vector competence of An. dirus to malaria parasites through upregulating Imd signaling pathway and enhancing the expression of TEP1. The data could not only help us to understand the impact and mechanism of Bs exposure on Anopheles’ vector competence to malaria but also provide us with novel clues for wiping out malaria using vector control.![]()
Collapse
Affiliation(s)
- Shasha Yu
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Pan Wang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Jie Qin
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Hong Zheng
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Jing Wang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Tingting Liu
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Xuesen Yang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, 400038, China
| | - Ying Wang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
37
|
Dong Y, Simões ML, Dimopoulos G. Versatile transgenic multistage effector-gene combinations for Plasmodium falciparum suppression in Anopheles. SCIENCE ADVANCES 2020; 6:eaay5898. [PMID: 32426491 PMCID: PMC7220273 DOI: 10.1126/sciadv.aay5898] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 02/27/2020] [Indexed: 05/14/2023]
Abstract
The malaria parasite's complex journey through the Anopheles mosquito vector provides multiple opportunities for targeting Plasmodium with recombinant effectors at different developmental stages and different host tissues. We have designed and expressed transgenes that efficiently suppress Plasmodium infection by targeting the parasite with multiple independent endogenous and exogenous effectors at multiple infection stages to potentiate suppression and minimize the probability for development of resistance to develop. We have also addressed the fitness impact of transgene expression on the mosquito. We show that highly potent suppression can be achieved by targeting both pre-oocyst stages by transgenically overexpressing either the endogenous immune deficiency immune pathway transcription factor Rel2 or a polycistronic mRNA encoding multiple antiparasitic effectors and simultaneously targeting the sporozoite stages with an anti-sporozoite single-chain antibody fused to the antiparasitic protein Scorpine. Expression of the selected endogenous effector systems appears to pose a lower fitness cost than does the use of foreign genes.
Collapse
|
38
|
Gao L, Song X, Wang J. Gut microbiota is essential in PGRP-LA regulated immune protection against Plasmodium berghei infection. Parasit Vectors 2020; 13:3. [PMID: 31907025 PMCID: PMC6945779 DOI: 10.1186/s13071-019-3876-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 12/30/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Malaria remains to be one of the deadliest infectious diseases and imposes substantial financial and social costs in the world. Mosquitoes rely on the immune system to control parasite infection. Peptidoglycan recognition proteins (PGRPs), a family of pattern-recognition receptors (PRR), are responsible for initiating and regulating immune signaling pathways. PGRP-LA is involved in the regulation of immune defense against the Plasmodium parasite, however, the underlying mechanism needs to be further elucidated. METHODS The spatial and temporal expression patterns of pgrp-la in Anopheles stephensi were analyzed by qPCR. The function of PGRP-LA was examined using a dsRNA-based RNA interference strategy. Western blot and periodic acid schiff (PAS) staining were used to assess the structural integrity of peritrophic matrix (PM). RESULTS The expression of pgrp-la in An. stephensi was induced in the midgut in response to the rapid proliferating gut microbiota post-blood meal. Knocking down of pgrp-la led to the downregulation of immune effectors that control gut microbiota growth. The decreased expression of these immune genes also facilitated P. berghei infection. However, such dsLA treatment did not influence the structural integrity of PM. When gut microbiota was removed by antibiotic treatment, the regulation of PGRP-LA on immune effectors was abolished and the knock down of pgrp-la failed to increase susceptibility of mosquitoes to parasite infection. CONCLUSIONS PGRP-LA regulates the immune responses by sensing the dynamics of gut microbiota. A mutual interaction between gut microbiota and PGRP-LA contributes to the immune defense against Plasmodium parasites in An. stephensi.
Collapse
Affiliation(s)
- Li Gao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200438, People's Republic of China.,Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200438, People's Republic of China
| | - Xiumei Song
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200438, People's Republic of China.,Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200438, People's Republic of China
| | - Jingwen Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200438, People's Republic of China. .,Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, 200438, People's Republic of China.
| |
Collapse
|
39
|
Abstract
The composition of insect hemolymph can change depending on many factors, e.g. access to nutrients, stress conditions, and current needs of the insect. In this chapter, insect immune-related polypeptides, which can be permanently or occasionally present in the hemolymph, are described. Their division into peptides or low-molecular weight proteins is not always determined by the length or secondary structure of a given molecule but also depends on the mode of action in insect immunity and, therefore, it is rather arbitrary. Antimicrobial peptides (AMPs) with their role in immunity, modes of action, and classification are presented in the chapter, followed by a short description of some examples: cecropins, moricins, defensins, proline- and glycine-rich peptides. Further, we will describe selected immune-related proteins that may participate in immune recognition, may possess direct antimicrobial properties, or can be involved in the modulation of insect immunity by both abiotic and biotic factors. We briefly cover Fibrinogen-Related Proteins (FREPs), Down Syndrome Cell Adhesion Molecules (Dscam), Hemolin, Lipophorins, Lysozyme, Insect Metalloproteinase Inhibitor (IMPI), and Heat Shock Proteins. The reader will obtain a partial picture presenting molecules participating in one of the most efficient immune strategies found in the animal world, which allow insects to inhabit all ecological land niches in the world.
Collapse
Affiliation(s)
- Iwona Wojda
- Department of Immunobiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Lublin, Poland.
| | - Małgorzata Cytryńska
- Department of Immunobiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Lublin, Poland
| | - Agnieszka Zdybicka-Barabas
- Department of Immunobiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Lublin, Poland
| | - Jakub Kordaczuk
- Department of Immunobiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Lublin, Poland
| |
Collapse
|
40
|
Bacterial communities associated with the midgut microbiota of wild Anopheles gambiae complex in Burkina Faso. Mol Biol Rep 2019; 47:211-224. [PMID: 31643044 DOI: 10.1007/s11033-019-05121-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 10/09/2019] [Indexed: 10/25/2022]
Abstract
Plasmodium falciparum is transmitted by mosquitoes from the Anopheles gambiae sensu lato (s.l) species complex and is responsible for severe forms of malaria. The composition of the mosquitoes' microbiota plays a role in P. falciparum transmission, so we studied midgut bacterial communities of An. gambiae s.l from Burkina Faso. DNA was extracted from 17 pools of midgut of mosquitoes from the Anopheles gambiae complex from six localities in three climatic areas, including cotton-growing and cotton-free localities to include potential differences in insecticide selection pressure. The v3-v4 region of the 16S rRNA gene was targeted and sequenced using Illumina Miseq (2 × 250 nt). Diversity analysis was performed using QIIME and R software programs. The major bacterial phylum was Proteobacteria (97.2%) in all samples. The most abundant genera were Enterobacter (32.8%) and Aeromonas (29.8%), followed by Pseudomonas (11.8%), Acinetobacter (5.9%) and Thorsellia (2.2%). No statistical difference in operational taxonomic units (OTUs) was found (Kruskal-Wallis FDR-p > 0.05) among the different areas, fields or localities. Richness and diversity indexes (observed OTUs, Chao1, Simpson and Shannon indexes) showed significant differences in the cotton-growing fields and in the agroclimatic zones, mainly in the Sudano-Sahelian area. OTUs from seven bacterial species that mediate refractoriness to Plasmodium infection in An. gambiae s.l were detected. The beta diversity analysis did not show any significant difference. Therefore, a same control strategy of using bacterial species refractoriness to Plasmodium to target mosquito midgut bacterial community and affect their fitness in malaria transmission may be valuable tool for future malaria control efforts in Burkina Faso.
Collapse
|
41
|
Santana RAG, Oliveira MC, Cabral I, Junior RCAS, de Sousa DRT, Ferreira L, Lacerda MVG, Monteiro WM, Abrantes P, Guerra MDGVB, Silveira H. Anopheles aquasalis transcriptome reveals autophagic responses to Plasmodium vivax midgut invasion. Parasit Vectors 2019; 12:261. [PMID: 31126324 PMCID: PMC6534896 DOI: 10.1186/s13071-019-3506-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 05/14/2019] [Indexed: 01/23/2023] Open
Abstract
Background Elimination of malaria depends on mastering transmission and understanding the biological basis of Plasmodium infection in the vector. The first mosquito organ to interact with the parasite is the midgut and its transcriptomic characterization during infection can reveal effective antiplasmodial responses able to limit the survival of the parasite. The vector response to Plasmodium vivax is not fully characterized, and its specificities when compared with other malaria parasites can be of fundamental interest for specific control measures. Methods Experimental infections were performed using a membrane-feeding device. Three groups were used: P. vivax-blood-fed, blood-fed on inactivated gametocytes, and unfed mosquitoes. Twenty-four hours after feeding, the mosquitoes were dissected and the midgut collected for transcriptomic analysis using RNAseq. Nine cDNA libraries were generated and sequenced on an Illumina HiSeq2500. Readings were checked for quality control and analysed using the Trinity platform for de novo transcriptome assembly. Transcript quantification was performed and the transcriptome was functionally annotated. Differential expression gene analysis was carried out. The role of the identified mechanisms was further explored using functional approaches. Results Forty-nine genes were identified as being differentially expressed with P. vivax infection: 34 were upregulated and 15 were downregulated. Half of the P. vivax-related differentially expressed genes could be related to autophagy; therefore, the effect of the known inhibitor (wortmannin) and activator (spermidine) was tested on the infection outcome. Autophagic activation significantly reduced the intensity and prevalence of infection. This was associated with transcription alterations of the autophagy regulating genes Beclin, DRAM and Apg8. Conclusions Our data indicate that P. vivax invasion of An. aquasalis midgut epithelium triggers an autophagic response and its activation reduces infection. This suggests a novel mechanism that mosquitoes can use to fight Plasmodium infection. Electronic supplementary material The online version of this article (10.1186/s13071-019-3506-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rosa Amélia Gonçalves Santana
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas/Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Maurício Costa Oliveira
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas/Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Iria Cabral
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas/Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Rubens Celso Andrade Silva Junior
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas/Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Débora Raysa Teixeira de Sousa
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas/Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Lucas Ferreira
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas/Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Marcus Vinícius Guimarães Lacerda
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas/Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil.,Instituto Leônidas & Maria Deane, Fundação Oswaldo Cruz, Manaus, Brazil
| | - Wuelton Marcelo Monteiro
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas/Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Patrícia Abrantes
- Instituto de Higiene e Medicina Tropical, Global Health and Tropical Medicine, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Maria das Graças Vale Barbosa Guerra
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas/Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Henrique Silveira
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas/Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil. .,Instituto de Higiene e Medicina Tropical, Global Health and Tropical Medicine, Universidade Nova de Lisboa, Lisboa, Portugal.
| |
Collapse
|
42
|
Alonso-Palomares LA, Moreno-García M, Lanz-Mendoza H, Salazar MI. Molecular Basis for Arbovirus Transmission by Aedes aegypti Mosquitoes. Intervirology 2019; 61:255-264. [PMID: 31082816 DOI: 10.1159/000499128] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 02/17/2019] [Indexed: 11/19/2022] Open
Abstract
Mosquitoes are considered the most important vectors for the transmission of pathogens to humans. Aedes aegypti is a unique species, not only by its highly anthropophilic and peridomestic habits but also because it can transmit an important variety of pathogenic viruses. Examples are dengue, yellow fever, chikungunya, Zika, and Mayaro viruses. After ingesting viremic blood, a wide range of mechanisms are activated in the mosquito to counteract viral infection. Nevertheless, these arboviruses possess strategies to overcome barriers in the mosquito and eventually reach the salivary glands to continue the transmission cycle. However, the infection and eventual transmission of arbovirus depends on multiple factors. The current review focuses in detail on the anatomic, physiological, and molecular characteristics of the mosquito A. aegypti that participate in response to a viral infection. In the past decades, the awareness of the importance of this mosquito as a disease vector and its impact on human health was largely recognized. We need to improve our comprehension of molecular mechanisms that determine the outcome of successful virus replication or control of infection for each arbovirus in the vector; this could lead to the design of effective control strategies in the future.
Collapse
Affiliation(s)
- Luis A Alonso-Palomares
- Laboratorio de Virología e Inmunovirología, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional (ENCB-IPN), Unidad Profesional "Lázaro Cárdenas", Mexico City, Mexico.,Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública (CISEI-INSP), Cuernavaca, Mexico
| | - Miguel Moreno-García
- Centro Regional de Control de Vectores, Secretaría de Salud (CERECOVE-SS), Panchimalco, Mexico
| | - Humberto Lanz-Mendoza
- Centro de Investigación sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública (CISEI-INSP), Cuernavaca, Mexico
| | - Ma Isabel Salazar
- Laboratorio de Virología e Inmunovirología, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional (ENCB-IPN), Unidad Profesional "Lázaro Cárdenas", Mexico City, Mexico,
| |
Collapse
|
43
|
Wu P, Shang Q, Dweteh OA, Huang H, Zhang S, Zhong J, Hou Q, Guo X. Over expression of bmo-miR-2819 suppresses BmNPV replication by regulating the BmNPV ie-1 gene in Bombyx mori. Mol Immunol 2019; 109:134-139. [PMID: 30947109 DOI: 10.1016/j.molimm.2019.03.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 02/07/2023]
Abstract
Bombyx mori nucleopolyhedrovirus (BmNPV) is a major pathogen that threatens the growth and sustainability of the sericulture industry. Accumulating studies in recent years suggest that insect viruses infection can change the host microRNAs (miRNAs) expression profile and both cellular and viral miRNAs play roles in host-pathogen interactions. Until now, the functional analysis of miRNA encoded by silkworm for host-virus interaction is limited. In this study, we validate the down-regulation of bmo-miR-2819 upon BmNPV infection by qRT-PCR and confirm the BmNPV immediately early 1 gene, ie-1 is one of the targets for bmo-miR-2819 based on the results of dual luciferase report assay. Overexpression of bmo-miR-2819 can significantly decline the abundance of IE-1 protein level in BmNPV-infected silkworm larvae. Further, the expression level of polyhedrin gene and VP39 protein of BmNPV in the infected larvae after applying bmo-miR-2819 mimics was significantly decreased comparing with that of larvae with mimic control. Our results suggest that overexpression of bmo-miR-2819 could suppress BmNPV replication by down-regulating the expression of BmNPV ie-1 gene, which demonstrate that cellular miRNAs could affect virus infection by regulating the expression of virus genes.
Collapse
Affiliation(s)
- Ping Wu
- Sericultural Research Institute, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, 212018, China.
| | - Qi Shang
- Sericultural Research Institute, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, 212018, China
| | - Owusu Amanfo Dweteh
- Sericultural Research Institute, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, 212018, China
| | - Haoling Huang
- Sericultural Research Institute, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, 212018, China
| | - Shaolun Zhang
- Sericultural Research Institute, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, 212018, China
| | - Jinbo Zhong
- Sericultural Research Institute, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, 212018, China
| | - Qirui Hou
- Sericultural Research Institute, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, 212018, China
| | - Xijie Guo
- Sericultural Research Institute, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu, 212018, China.
| |
Collapse
|
44
|
Lu Y, Johnston PR, Dennis SR, Monaghan MT, John U, Spaak P, Wolinska J. Daphnia galeata responds to the exposure to an ichthyosporean gut parasite by down-regulation of immunity and lipid metabolism. BMC Genomics 2018; 19:932. [PMID: 30547741 PMCID: PMC6295042 DOI: 10.1186/s12864-018-5312-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 11/27/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Regulatory circuits of infection in the emerging experimental model system, water flea Daphnia and their microparasites, remain largely unknown. Here we provide the first molecular insights into the response of Daphnia galeata to its highly virulent and common parasite Caullerya mesnili, an ichthyosporean that infects the gut epithelium. We generated a transcriptomic dataset using RNAseq from parasite-exposed (vs. control) Daphnia, at two time points (4 and 48 h) after parasite exposure. RESULTS We found a down-regulation of metabolism and immunity-related genes, at 48 h (but not 4 h) after parasite exposure. These genes are involved in lipid metabolism and fatty acid biosynthesis, as well as microbe recognition (e.g. c-type lectins) and pathogen attack (e.g. gut chitin). CONCLUSIONS General metabolic suppression implies host energy shift from reproduction to survival, which is in agreement with the known drastic reduction in Daphnia fecundity after Caullerya infection. The down-regulation of gut chitin indicates a possible interaction between the peritrophic matrix and the evading host immune system. Our study provides the first description of host transcriptional responses in this very promising host-parasite experimental system.
Collapse
Affiliation(s)
- Yameng Lu
- Leibniz Institute of Freshwater Ecology and Inland Fisheries (IGB), Berlin, Germany. .,Berlin Center for Genomics in Biodiversity Research (BeGenDiv), Berlin, Germany.
| | - Paul R Johnston
- Leibniz Institute of Freshwater Ecology and Inland Fisheries (IGB), Berlin, Germany.,Berlin Center for Genomics in Biodiversity Research (BeGenDiv), Berlin, Germany.,Freie Universität Berlin, Berlin, Germany
| | - Stuart R Dennis
- Swiss Federal Institute of Aquatic Science and Technology (Eawag), Dübendorf, Switzerland
| | - Michael T Monaghan
- Leibniz Institute of Freshwater Ecology and Inland Fisheries (IGB), Berlin, Germany.,Berlin Center for Genomics in Biodiversity Research (BeGenDiv), Berlin, Germany
| | - Uwe John
- Alfred Wegener Institut Helmholtz Zentrum für Polar und Meeresforschung (AWI), Bremerhaven, Germany.,Helmholtz Institute for Functional Marine Biodiversity (HIFMB), Oldenburg, Germany
| | - Piet Spaak
- Swiss Federal Institute of Aquatic Science and Technology (Eawag), Dübendorf, Switzerland
| | - Justyna Wolinska
- Leibniz Institute of Freshwater Ecology and Inland Fisheries (IGB), Berlin, Germany.,Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
45
|
Ogaugwu CE, Agbo SO, Adekoya MA. CRISPR in Sub-Saharan Africa: Applications and Education. Trends Biotechnol 2018; 37:234-237. [PMID: 30100229 PMCID: PMC6378648 DOI: 10.1016/j.tibtech.2018.07.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 07/18/2018] [Accepted: 07/18/2018] [Indexed: 01/12/2023]
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR) technology has enabled genetic engineering feats previously considered impracticable, offering great hopes for solutions to problems facing society. We consider it timely to highlight how CRISPR can benefit public health, medicine, and agriculture in sub-Saharan Africa (SSA) and offer recommendations for successful implementation.
Collapse
Affiliation(s)
- Christian E Ogaugwu
- Department of Microbiology and Molecular Genetics, University of California, Irvine, CA 92697-4500, USA; Department of Animal and Environmental Biology, Federal University Oye-Ekiti, 371010 Ekiti State, Nigeria; Laboratory website: http://sites.uci.edu/jameslab.
| | - Stanley O Agbo
- Department of Animal and Environmental Biology, Federal University Oye-Ekiti, 371010 Ekiti State, Nigeria
| | - Modinat A Adekoya
- Department of Plant Science and Biotechnology, Federal University Oye-Ekiti, 371010 Ekiti State, Nigeria
| |
Collapse
|
46
|
Abstract
In this review, we explore the state-of-the-art of sand fly relationships with microbiota, viruses and Leishmania, with particular emphasis on the vector immune responses. Insect-borne diseases are a major public health problem in the world. Phlebotomine sand flies are proven vectors of several aetiological agents including viruses, bacteria and the trypanosomatid Leishmania, which are responsible for diseases such as viral encephalitis, bartonellosis and leishmaniasis, respectively. All metazoans in nature coexist intimately with a community of commensal microorganisms known as microbiota. The microbiota has a fundamental role in the induction, maturation and function of the host immune system, which can modulate host protection from pathogens and infectious diseases. We briefly review viruses of public health importance present in sand flies and revisit studies done on bacterial and fungal gut contents of these vectors. We bring this information into the context of sand fly development and immune responses. We highlight the immunity mechanisms that the insect utilizes to survive the potential threats involved in these interactions and discuss the recently discovered complex interactions among microbiota, sand fly, Leishmania and virus. Additionally, some of the alternative control strategies that could benefit from the current knowledge are considered.
Collapse
|
47
|
Wang YH, Chang MM, Wang XL, Zheng AH, Zou Z. The immune strategies of mosquito Aedes aegypti against microbial infection. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 83:12-21. [PMID: 29217264 DOI: 10.1016/j.dci.2017.12.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/30/2017] [Accepted: 12/03/2017] [Indexed: 06/07/2023]
Abstract
Yellow fever mosquito Aedes aegypti transmits many devastating arthropod-borne viruses (arboviruses), such as dengue virus, yellow fever virus, Chikungunya virus, and Zika virus, which cause great concern to human health. Mosquito control is an effective method to block the spread of infectious diseases. Ae. aegypti uses its innate immune system to fight against arboviruses, parasites, and fungi. In this review, we briefly summarize the recent findings in the immune response of Ae. aegypti against arboviral and entomopathogenic infections. This review enriches our understanding of the mosquito immune system and provides evidence to support the development of novel mosquito control strategies.
Collapse
Affiliation(s)
- Yan-Hong Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Meng-Meng Chang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xue-Li Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Ai-Hua Zheng
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhen Zou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
48
|
Simões ML, Caragata EP, Dimopoulos G. Diverse Host and Restriction Factors Regulate Mosquito-Pathogen Interactions. Trends Parasitol 2018; 34:603-616. [PMID: 29793806 DOI: 10.1016/j.pt.2018.04.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 04/25/2018] [Accepted: 04/26/2018] [Indexed: 12/12/2022]
Abstract
Mosquitoes transmit diseases that seriously impact global human health. Despite extensive knowledge of the life cycles of mosquito-borne parasites and viruses within their hosts, control strategies have proven insufficient to halt their spread. An understanding of the relationships established between such pathogens and the host tissues they inhabit is therefore paramount for the development of new strategies that specifically target these interactions, to prevent the pathogens' maturation and transmission. Here we present an updated account of the antagonists and host factors that affect the development of Plasmodium, the parasite causing malaria, and mosquito-borne viruses, such as dengue virus and Zika virus, within their mosquito vectors, and we discuss the similarities and differences between Plasmodium and viral systems, looking toward the elucidation of new targets for disease control.
Collapse
Affiliation(s)
- Maria L Simões
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA; These authors contributed equally
| | - Eric P Caragata
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA; These authors contributed equally
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
49
|
microRNA profiles and functions in mosquitoes. PLoS Negl Trop Dis 2018; 12:e0006463. [PMID: 29718912 PMCID: PMC5951587 DOI: 10.1371/journal.pntd.0006463] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 05/14/2018] [Accepted: 04/19/2018] [Indexed: 11/19/2022] Open
Abstract
Mosquitoes are incriminated as vectors for many crippling diseases, including malaria, West Nile fever, Dengue fever, and other neglected tropical diseases (NTDs). microRNAs (miRNAs) can interact with multiple target genes to elicit biological functions in the mosquitoes. However, characterization and function of individual miRNAs and their potential targets have not been fully determined to date. We conducted a systematic review of published literature following PRISMA guidelines. We summarize the information about miRNAs in mosquitoes to better understand their metabolism, development, and responses to microorganisms. Depending on the study, we found that miRNAs were dysregulated in a species-, sex-, stage-, and tissue/organ-specific manner. Aberrant miRNA expressions were observed in development, metabolism, host-pathogen interactions, and insecticide resistance. Of note, many miRNAs were down-regulated upon pathogen infection. The experimental studies have expanded the identification of miRNA target from the 3' untranslated regions (UTRs) of mRNAs of mosquitoes to the 5' UTRs of mRNAs of the virus. In addition, we discuss current trends in mosquito miRNA research and offer suggestions for future studies.
Collapse
|
50
|
Lefevre T, Ohm J, Dabiré KR, Cohuet A, Choisy M, Thomas MB, Cator L. Transmission traits of malaria parasites within the mosquito: Genetic variation, phenotypic plasticity, and consequences for control. Evol Appl 2018; 11:456-469. [PMID: 29636799 PMCID: PMC5891056 DOI: 10.1111/eva.12571] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 10/23/2017] [Indexed: 12/16/2022] Open
Abstract
Evaluating the risk of emergence and transmission of vector-borne diseases requires knowledge of the genetic and environmental contributions to pathogen transmission traits. Compared to the significant effort devoted to understanding the biology of malaria transmission from vertebrate hosts to mosquito vectors, the strategies that malaria parasites have evolved to maximize transmission from vectors to vertebrate hosts have been largely overlooked. While determinants of infection success within the mosquito host have recently received attention, the causes of variability for other key transmission traits of malaria, namely the duration of parasite development and its virulence within the vector, as well as its ability to alter mosquito behavior, remain largely unknown. This important gap in our knowledge needs to be bridged in order to obtain an integrative view of the ecology and evolution of malaria transmission strategies. Associations between transmission traits also need to be characterized, as they trade-offs and constraints could have important implications for understanding the evolution of parasite transmission. Finally, theoretical studies are required to evaluate how genetic and environmental influences on parasite transmission traits can shape malaria dynamics and evolution in response to disease control.
Collapse
Affiliation(s)
- Thierry Lefevre
- MIVEGEC, IRD, CNRSUniversity of MontpellierMontpellierFrance
- Institut de Recherche en Sciences de la Santé (IRSS)Bobo DioulassoBurkina Faso
- Laboratoire Mixte International sur les Vecteurs (LAMIVECT)Bobo DioulassoBurkina Faso
| | - Johanna Ohm
- Department of Entomology and Center for Infectious Disease DynamicsPenn State UniversityUniversity ParkPAUSA
| | - Kounbobr R. Dabiré
- Institut de Recherche en Sciences de la Santé (IRSS)Bobo DioulassoBurkina Faso
- Laboratoire Mixte International sur les Vecteurs (LAMIVECT)Bobo DioulassoBurkina Faso
| | - Anna Cohuet
- MIVEGEC, IRD, CNRSUniversity of MontpellierMontpellierFrance
| | - Marc Choisy
- MIVEGEC, IRD, CNRSUniversity of MontpellierMontpellierFrance
- Oxford University Clinical Research UnitHanoiVietnam
| | - Matthew B. Thomas
- Department of Entomology and Center for Infectious Disease DynamicsPenn State UniversityUniversity ParkPAUSA
| | - Lauren Cator
- Grand Challenges in Ecosystems and EnvironmentImperial College LondonAscotUK
| |
Collapse
|