1
|
Elmorsy EA. Molecular host-parasite interaction at the site of vector bite. Exp Parasitol 2025; 270:108902. [PMID: 39826601 DOI: 10.1016/j.exppara.2025.108902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/19/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Affiliation(s)
- Eman Attia Elmorsy
- Medical Parasitology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt.
| |
Collapse
|
2
|
Wang LT, Idris AH, Kisalu NK, Crompton PD, Seder RA. Monoclonal antibodies to the circumsporozoite proteins as an emerging tool for malaria prevention. Nat Immunol 2024; 25:1530-1545. [PMID: 39198635 DOI: 10.1038/s41590-024-01938-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/17/2024] [Indexed: 09/01/2024]
Abstract
Despite various public health strategies, malaria caused by Plasmodium falciparum parasites remains a major global health challenge that requires development of new interventions. Extended half-life human monoclonal antibodies targeting the P. falciparum circumsporozoite protein on sporozoites, the infective form of malaria parasites, prevent malaria in rodents and humans and have been advanced into clinical development. The protective epitopes on the circumsporozoite protein targeted by monoclonal antibodies have been defined. Cryogenic electron and multiphoton microscopy have enabled mechanistic structural and functional investigations of how antibodies bind to the circumsporozoite protein and neutralize sporozoites. Moreover, innovations in bioinformatics and antibody engineering have facilitated enhancement of antibody potency and durability. Here, we summarize the latest scientific advances in understanding how monoclonal antibodies to the circumsporozoite protein prevent malaria and highlight existing clinical data and future plans for how this emerging intervention can be used alone or alongside existing antimalarial interventions to control malaria across at-risk populations.
Collapse
Affiliation(s)
- Lawrence T Wang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Azza H Idris
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA.
| | - Neville K Kisalu
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- PATH's Center for Vaccine Innovation and Access, Washington, DC, USA
| | - Peter D Crompton
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
3
|
Liu Z, Li S, Anantha P, Thanakornsombut T, Wu L, Chen J, Tsuchiya R, Tripathi AK, Chen Y, Barman I. Plasmodium sporozoite shows distinct motility patterns in responses to three-dimensional environments. iScience 2024; 27:110463. [PMID: 39129829 PMCID: PMC11315120 DOI: 10.1016/j.isci.2024.110463] [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: 09/28/2023] [Revised: 11/20/2023] [Accepted: 07/02/2024] [Indexed: 08/13/2024] Open
Abstract
During malaria infection, Plasmodium sporozoites, the fast-moving stage of the parasite, are injected by a mosquito into the skin of the mammalian host. In the skin, sporozoites need to migrate through the dermal tissue to enter the blood vessel. Sporozoite motility is critical for infection but not well understood. Here, we used collagen hydrogels with tunable fiber structures, as an in vitro model for the skin. After injecting sporozoites into the hydrogel, we analyzed their motility in three-dimension (3D). We found that sporozoites demonstrated chiral motility, in that they mostly follow right-handed helical trajectories. In high-concentration collagen gel, sporozoites have lower instantaneous speed, but exhibit straighter tracks compared to low-concentration collagen gel, which leads to longer net displacement and faster dissemination. Taken together, our study indicates an inner mechanism for sporozoites to adapt to the environment, which could help with their successful exit from the skin tissue.
Collapse
Affiliation(s)
- Zhenhui Liu
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Songman Li
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Pooja Anantha
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | | | - Lintong Wu
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Junjie Chen
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Center for Cell Dynamics, Johns Hopkins University, Baltimore, MD, USA
| | - Ryohma Tsuchiya
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Center for Cell Dynamics, Johns Hopkins University, Baltimore, MD, USA
| | - Abhai K. Tripathi
- Department of Molecular Microbiology & Immunology, Johns Hopkins University, Baltimore, MD, USA
| | - Yun Chen
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Center for Cell Dynamics, Johns Hopkins University, Baltimore, MD, USA
| | - Ishan Barman
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
4
|
Liffner B, Absalon S. Expansion microscopy of apicomplexan parasites. Mol Microbiol 2024; 121:619-635. [PMID: 37571814 DOI: 10.1111/mmi.15135] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 07/15/2023] [Accepted: 07/20/2023] [Indexed: 08/13/2023]
Abstract
Apicomplexan parasites comprise significant pathogens of humans, livestock and wildlife, but also represent a diverse group of eukaryotes with interesting and unique cell biology. The study of cell biology in apicomplexan parasites is complicated by their small size, and historically this has required the application of cutting-edge microscopy techniques to investigate fundamental processes like mitosis or cell division in these organisms. Recently, a technique called expansion microscopy has been developed, which rather than increasing instrument resolution like most imaging modalities, physically expands a biological sample. In only a few years since its development, a derivative of expansion microscopy known as ultrastructure-expansion microscopy (U-ExM) has been widely adopted and proven extremely useful for studying cell biology of Apicomplexa. Here, we review the insights into apicomplexan cell biology that have been enabled through the use of U-ExM, with a specific focus on Plasmodium, Toxoplasma and Cryptosporidium. Further, we summarize emerging expansion microscopy modifications and modalities and forecast how these may influence the field of parasite cell biology in future.
Collapse
Affiliation(s)
- Benjamin Liffner
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sabrina Absalon
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
5
|
Andolina C, Graumans W, Guelbeogo M, van Gemert GJ, Ramijth J, Harouna S, Soumanaba Z, Stoter R, Vegte-Bolmer M, Pangos M, Sinnis P, Collins K, Staedke SG, Tiono AB, Drakeley C, Lanke K, Bousema T. Quantification of sporozoite expelling by Anopheles mosquitoes infected with laboratory and naturally circulating P. falciparum gametocytes. eLife 2024; 12:RP90989. [PMID: 38517746 PMCID: PMC10959522 DOI: 10.7554/elife.90989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2024] Open
Abstract
It is currently unknown whether all Plasmodium falciparum-infected mosquitoes are equally infectious. We assessed sporogonic development using cultured gametocytes in the Netherlands and naturally circulating strains in Burkina Faso. We quantified the number of sporozoites expelled into artificial skin in relation to intact oocysts, ruptured oocysts, and residual salivary gland sporozoites. In laboratory conditions, higher total sporozoite burden was associated with shorter duration of sporogony (p<0.001). Overall, 53% (116/216) of infected Anopheles stephensi mosquitoes expelled sporozoites into artificial skin with a median of 136 expelled sporozoites (interquartile range [IQR], 34-501). There was a strong positive correlation between ruptured oocyst number and salivary gland sporozoite load (ρ = 0.8; p<0.0001) and a weaker positive correlation between salivary gland sporozoite load and number of sporozoites expelled (ρ = 0.35; p=0.0002). In Burkina Faso, Anopheles coluzzii mosquitoes were infected by natural gametocyte carriers. Among salivary gland sporozoite positive mosquitoes, 89% (33/37) expelled sporozoites with a median of 1035 expelled sporozoites (IQR, 171-2969). Again, we observed a strong correlation between ruptured oocyst number and salivary gland sporozoite load (ρ = 0.9; p<0.0001) and a positive correlation between salivary gland sporozoite load and the number of sporozoites expelled (ρ = 0.7; p<0.0001). Several mosquitoes expelled multiple parasite clones during probing. Whilst sporozoite expelling was regularly observed from mosquitoes with low infection burdens, our findings indicate that mosquito infection burden is positively associated with the number of expelled sporozoites. Future work is required to determine the direct implications of these findings for transmission potential.
Collapse
Affiliation(s)
- Chiara Andolina
- Department of Medical Microbiology, Radboud University Nijmegen Medical CentreNijmegenNetherlands
| | - Wouter Graumans
- Department of Medical Microbiology, Radboud University Nijmegen Medical CentreNijmegenNetherlands
| | - Moussa Guelbeogo
- Centre National de Recherche et de Formation sur le PaludismeOuagadougouBurkina Faso
| | - Geert-Jan van Gemert
- Department of Medical Microbiology, Radboud University Nijmegen Medical CentreNijmegenNetherlands
| | - Jordache Ramijth
- Department of Medical Microbiology, Radboud University Nijmegen Medical CentreNijmegenNetherlands
| | - Soré Harouna
- Centre National de Recherche et de Formation sur le PaludismeOuagadougouBurkina Faso
| | - Zongo Soumanaba
- Centre National de Recherche et de Formation sur le PaludismeOuagadougouBurkina Faso
| | - Rianne Stoter
- Department of Medical Microbiology, Radboud University Nijmegen Medical CentreNijmegenNetherlands
| | - Marga Vegte-Bolmer
- Department of Medical Microbiology, Radboud University Nijmegen Medical CentreNijmegenNetherlands
| | - Martina Pangos
- Department of Plastic and Reconstructive Surgery, Azienda Ospedaliero Universitaria GiulianoIsontina TriesteTriesteItaly
| | - Photini Sinnis
- Department of Molecular Microbiology and Immunology, Johns HopkinsBloomberg School of Public HealthBaltimoreUnited States
| | - Katharine Collins
- Department of Medical Microbiology, Radboud University Nijmegen Medical CentreNijmegenNetherlands
| | - Sarah G Staedke
- Liverpool School of Tropical MedicineLiverpoolUnited Kingdom
| | - Alfred B Tiono
- Centre National de Recherche et de Formation sur le PaludismeOuagadougouBurkina Faso
| | - Chris Drakeley
- Department of Immunology and Infection, London School of Hygiene and Tropical MedicineLondonUnited Kingdom
| | - Kjerstin Lanke
- Department of Medical Microbiology, Radboud University Nijmegen Medical CentreNijmegenNetherlands
| | - Teun Bousema
- Department of Medical Microbiology, Radboud University Nijmegen Medical CentreNijmegenNetherlands
- Department of Immunology and Infection, London School of Hygiene and Tropical MedicineLondonUnited Kingdom
| |
Collapse
|
6
|
Adedayo BC, Komolafe T, Ojueromi OO, Oboh G. Evaluation of Andrographis paniculata-supplemented Diet on the reproductive indices of Plasmodium berghei-infected mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117558. [PMID: 38092319 DOI: 10.1016/j.jep.2023.117558] [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: 10/19/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 12/19/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The King of Bitters (Andrographis paniculata) is a plant used to cure a wide range of infectious diseases which includes malaria, fever and others. However, there is a paucity of scientific evidence of its effect on male reproductive indices during malaria treatment. AIM OF THE STUDY The aim of this study is to evaluate the effect of supplemented diet on antiplasmodial, hematological and male reproductive indices in mice infected with Plasmodium berghei. MATERIALS AND METHODS Aqueous extract of A. paniculata (King of Bitters, KGB) was prepared and the total phenol and flavonoid contents were determined. Forty-two mice, weighing 20-25 g, were distributed into 7 groups consisting of 6 mice each. The mice were innoculated with strain NK65 Plasmodium berghei (Chloroquine, CQ sensitive) and the parasitemia suppression was assessed. The mice were fed with the dietary supplementation of KGB at varying inclusions (2.5%, 5%, 7.5%, and 10%) and administered 10 mg/kg CQ (which served as the positive control) for 5 consecutive days after infection was established. The reactive malondialdeahyde (MDA), antioxidant [superoxide dismutase (SOD), catalase (CAT), reduced glutathione (GSH)] and the hematological (hemoglobin, packed cell volume and red blood cell) parameters in the infected mice were determined. The reproductive indices (serum testosterone, luteinizing hormone (LH), follicle-stimulating hormone (FSH), sperm count, sperm motility, and sperm viability) and testis histopathology were also assessed. RESULT The result revealed that KGB had a total phenol content of 32.55 mgGAE/g and total flavonoid content of 19.71 mgQUE/g. The infected mice treated with the dietary supplementation of KGB showed significantly decreased (p < 0.05) parasitaemia and MDA levels. Furthermore, the 7.5% dietary inclusion showed significant improvement in the antioxidant, hematological and reproductive indices as well as the restoration of testis morphology as seen in the histopathology plate of the infected mice treated with KGB. Hence, this study suggests that the KGB- supplemented diet (7.5%) may be a potential alternative and complementary therapy in the treatment of malaria infection and reproductive disorders.
Collapse
Affiliation(s)
- Bukola Christiana Adedayo
- Functional Food and Nutraceutical Unit, Department of Biochemistry, Federal University of Technology, Akure, Ondo State, Nigeria.
| | - Toluwase Komolafe
- Functional Food and Nutraceutical Unit, Department of Biochemistry, Federal University of Technology, Akure, Ondo State, Nigeria
| | - Opeyemi Oluwafemi Ojueromi
- Functional Food and Nutraceutical Unit, Department of Biochemistry, Federal University of Technology, Akure, Ondo State, Nigeria; Department of Pure and Applied Sciences, Precious Cornerstone University, Ibadan, Oyo State, Nigeria
| | - Ganiyu Oboh
- Functional Food and Nutraceutical Unit, Department of Biochemistry, Federal University of Technology, Akure, Ondo State, Nigeria
| |
Collapse
|
7
|
Bron GM, Wichgers Schreur PJ, de Jong MCM, van Keulen L, Vloet RPM, Koenraadt CJM, Kortekaas J, ten Bosch QA. Quantifying Rift Valley fever virus transmission efficiency in a lamb-mosquito-lamb model. Front Cell Infect Microbiol 2023; 13:1206089. [PMID: 38170150 PMCID: PMC10759236 DOI: 10.3389/fcimb.2023.1206089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 10/30/2023] [Indexed: 01/05/2024] Open
Abstract
Rift Valley fever virus (RVFV) is a (re)emerging mosquito-borne pathogen impacting human and animal health. How RVFV spreads through a population depends on population-level and individual-level interactions between vector, host and pathogen. Here, we estimated the probability for RVFV to transmit to naive animals by experimentally exposing lambs to a bite of an infectious mosquito, and assessed if and how RVFV infection subsequently developed in the exposed animal. Aedes aegypti mosquitoes, previously infected via feeding on a viremic lamb, were used to expose naive lambs to the virus. Aedes aegypti colony mosquitoes were used as they are easy to maintain and readily feed in captivity. Other mosquito spp. could be examined with similar methodology. Lambs were exposed to either 1-3 (low exposure) or 7-9 (high exposure) infectious mosquitoes. All lambs in the high exposure group became viremic and showed characteristic signs of Rift Valley fever within 2-4 days post exposure. In contrast, 3 out of 12 lambs in the low exposure group developed viremia and disease, with similar peak-levels of viremia as the high exposure group but with some heterogeneity in the onset of viremia. These results suggest that the likelihood for successful infection of a ruminant host is affected by the number of infectious mosquitoes biting, but also highlights that a single bite of an infectious mosquito can result in disease. The per bite mosquito-to-host transmission efficiency was estimated at 28% (95% confidence interval: 15 - 47%). We subsequently combined this transmission efficiency with estimates for life traits of Aedes aegypti or related mosquitoes into a Ross-McDonald mathematical model to illustrate scenarios under which major RVFV outbreaks could occur in naïve populations (i.e., R0 >1). The model revealed that relatively high vector-to-host ratios as well as mosquitoes feeding preferably on competent hosts are required for R0 to exceed 1. Altogether, this study highlights the importance of experiments that mimic natural exposure to RVFV. The experiments facilitate a better understanding of the natural progression of disease and a direct way to obtain epidemiological parameters for mathematical models.
Collapse
Affiliation(s)
- Gebbiena M. Bron
- Quantitative Veterinary Epidemiology, Wageningen University and Research, Wageningen, Netherlands
| | | | - Mart C. M. de Jong
- Quantitative Veterinary Epidemiology, Wageningen University and Research, Wageningen, Netherlands
| | - Lucien van Keulen
- Wageningen Bioveterinary Research, Wageningen University and Research, Lelystad, Netherlands
| | - Rianka P. M. Vloet
- Wageningen Bioveterinary Research, Wageningen University and Research, Lelystad, Netherlands
| | | | - Jeroen Kortekaas
- Wageningen Bioveterinary Research, Wageningen University and Research, Lelystad, Netherlands
| | - Quirine A. ten Bosch
- Quantitative Veterinary Epidemiology, Wageningen University and Research, Wageningen, Netherlands
| |
Collapse
|
8
|
Vigário AM, Pamplona A. γδ T cells as immunotherapy for malaria: balancing challenges and opportunities. Front Immunol 2023; 14:1242306. [PMID: 38124746 PMCID: PMC10731019 DOI: 10.3389/fimmu.2023.1242306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023] Open
Affiliation(s)
- Ana M. Vigário
- Projecto Medicina, Faculdade de Ciências da Vida, Universidade da Madeira, Funchal, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina de Lisboa, Lisboa, Portugal
| | - Ana Pamplona
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina de Lisboa, Lisboa, Portugal
| |
Collapse
|
9
|
Nasir G, Sinnis P. Transport of antibody into the skin is only partially dependent upon the neonatal Fc-receptor. PLoS One 2023; 18:e0273960. [PMID: 37093800 PMCID: PMC10124839 DOI: 10.1371/journal.pone.0273960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 03/30/2023] [Indexed: 04/25/2023] Open
Abstract
The dermis is the portal of entry for most vector-transmitted pathogens, making the host's immune response at this site critical in mitigating the magnitude of infection. For malaria, antibody-mediated neutralization of Plasmodium parasites in the dermis was recently demonstrated. However, surprisingly little is known about the mechanisms that govern antibody transport into the skin. Since the neonatal Fc receptor (FcRn) has been shown to transcytose IgG into various tissues, we sought to understand its contribution to IgG transport into the skin and antibody-mediated inhibition of Plasmodium parasites following mosquito bite inoculation. Using confocal imaging, we show that the transport of an anti-Langerin mAb into the skin occurs but is only partially reduced in mice lacking FcRn. To understand the relevance of FcRn in the context of malaria infection, we use the rodent parasite Plasmodium berghei and show that passively-administered anti-malarial antibody in FcRn deficient mice, does not reduce parasite burden to the same extent as previously observed in wildtype mice. Overall, our data suggest that FcRn plays a role in the transport of IgG into the skin but is not the major driver of IgG transport into this tissue. These findings have implications for the rational design of antibody-based therapeutics for malaria as well as other vector-transmitted pathogens.
Collapse
Affiliation(s)
- Gibran Nasir
- Johns Hopkins Malaria Institute and Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Photini Sinnis
- Johns Hopkins Malaria Institute and Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| |
Collapse
|
10
|
Barthelemy J, Bogard G, Wolowczuk I. Beyond energy balance regulation: The underestimated role of adipose tissues in host defense against pathogens. Front Immunol 2023; 14:1083191. [PMID: 36936928 PMCID: PMC10019896 DOI: 10.3389/fimmu.2023.1083191] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/09/2023] [Indexed: 03/06/2023] Open
Abstract
Although the adipose tissue (AT) is a central metabolic organ in the regulation of whole-body energy homeostasis, it is also an important endocrine and immunological organ. As an endocrine organ, AT secretes a variety of bioactive peptides known as adipokines - some of which have inflammatory and immunoregulatory properties. As an immunological organ, AT contains a broad spectrum of innate and adaptive immune cells that have mostly been studied in the context of obesity. However, overwhelming evidence supports the notion that AT is a genuine immunological effector site, which contains all cell subsets required to induce and generate specific and effective immune responses against pathogens. Indeed, AT was reported to be an immune reservoir in the host's response to infection, and a site of parasitic, bacterial and viral infections. In addition, besides AT's immune cells, preadipocytes and adipocytes were shown to express innate immune receptors, and adipocytes were reported as antigen-presenting cells to regulate T-cell-mediated adaptive immunity. Here we review the current knowledge on the role of AT and AT's immune system in host defense against pathogens. First, we will summarize the main characteristics of AT: type, distribution, function, and extraordinary plasticity. Second, we will describe the intimate contact AT has with lymph nodes and vessels, and AT immune cell composition. Finally, we will present a comprehensive and up-to-date overview of the current research on the contribution of AT to host defense against pathogens, including the respiratory viruses influenza and SARS-CoV-2.
Collapse
Affiliation(s)
| | | | - Isabelle Wolowczuk
- Univ. Lille, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (Inserm), Centre Hospitalier Universitaire de Lille (CHU Lille), Institut Pasteur de Lille, U1019 - UMR 9017 - Center for Infection and Immunity of Lille (CIIL), Lille, France
| |
Collapse
|
11
|
Ojueromi OO, Oboh G, Ademosun AO. Effect of black seeds (Nigella sativa) on inflammatory and immunomodulatory markers in Plasmodium berghei-infected mice. J Food Biochem 2022; 46:e14300. [PMID: 35833536 DOI: 10.1111/jfbc.14300] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 05/14/2022] [Accepted: 05/25/2022] [Indexed: 12/29/2022]
Abstract
Nigella sativa, a core dietary supplement and food additive in folklore is one of the most broadly studied seed plants in the global nutraceutical sector. Malaria infection impairs the ability of principal cells of the immune system to trigger an efficient inflammatory and immune response. Ninety-six mice, weighing 20-25 g, were grouped into 12 consisting of 8 animals each. The mice were infected with standard inoculum of the strain NK65 Plasmodium berghei (chloroquine sensitive) and the percentage parasitemia suppression were evaluated. The individual effect of black seed supplemented diet and its combinatory effect with chloroquine (CQ) were investigated on reactive oxygen species (ROS), glutathione peroxidase (GPx), reduced glutathione (GSH), glutathione-S-transferase (GST), serum immunoglobulins (IgG and IgM), and the hematological parameters (hemoglobin, packed cell volume, and red blood cell count) in P. berghei infected mice. The inflammatory cytokines, tumor necrosis factor (TNF-α), interleukin (IL-6 and IL-10), as well as IgG and IgM were assayed in the serum. The mice temperature and behavioral changes were observed. Infected mice treated with the dietary supplementation of black seed with a percentage inclusion (2.5%, 5%, 10%) showed significantly decreased parasitemia and ROS levels (p < 0.05) compared with the untreated mice. The result demonstrated a significant suppression in the pro-inflammatory cytokines (TNF-α, IL-6) levels and a notable elevation in the anti-inflammatory cytokine (IL-10), antioxidant markers as well as the immunoglobulin levels of the P. berghei-infected mice treated with black seed. The study revealed that black seed enhanced host antioxidant status, modulated inflammatory and immune response by regulating some inflammatory cytokines and immunomodulatory mediators. PRACTICAL APPLICATIONS: Black seed (Nigella sativa) has been a dietary supplement and natural remedy for many centuries. Inflammatory and immune diseases are the most notable cause of mortality in the world and more than 50% of deaths have been attributed to it. However, there is paucity of information on the effect of N. sativa on anti-inflammatory and immunomodulatory ability during malaria infection. The result suggests that N. sativa produced antioxidant, anti-inflammatory, and immunomodulatory effect in Plasmodium berghei-infected mice via the participation of glutathione antioxidant system, serum antibodies, and some inflammatory cytokines.
Collapse
Affiliation(s)
- Opeyemi Oluwafemi Ojueromi
- Functional Foods and Nutraceuticals Unit, Department of Biochemistry, Federal University of Technology, Akure, Ondo State, Nigeria
| | - Ganiyu Oboh
- Functional Foods and Nutraceuticals Unit, Department of Biochemistry, Federal University of Technology, Akure, Ondo State, Nigeria
| | - Ayokunle Olubode Ademosun
- Functional Foods and Nutraceuticals Unit, Department of Biochemistry, Federal University of Technology, Akure, Ondo State, Nigeria
| |
Collapse
|
12
|
Andoh NE, Gyan BA. The Potential Roles of Glial Cells in the Neuropathogenesis of Cerebral Malaria. Front Cell Infect Microbiol 2021; 11:741370. [PMID: 34692564 PMCID: PMC8529055 DOI: 10.3389/fcimb.2021.741370] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/17/2021] [Indexed: 01/02/2023] Open
Abstract
Cerebral malaria (CM) is a severe neurological complication of malaria caused by the Plasmodium falciparum parasite. It is one of the leading causes of death in children under 5 years of age in Sub-Saharan Africa. CM is associated with blood-brain barrier disruption and long-term neurological sequelae in survivors of CM. Despite the vast amount of research on cerebral malaria, the cause of neurological sequelae observed in CM patients is poorly understood. In this article, the potential roles of glial cells, astrocytes, and microglia, in cerebral malaria pathogenesis are reviewed. The possible mechanisms by which glial cells contribute to neurological damage in CM patients are also examined.
Collapse
Affiliation(s)
- Nana Efua Andoh
- Noguchi Memorial Institute for Medical Research, Department of Parasitology, University of Ghana, Accra, Ghana
| | - Ben Adu Gyan
- Noguchi Memorial Institute for Medical Research, Department of Immunology, University of Ghana, Accra, Ghana
| |
Collapse
|
13
|
Thongsripong P, Hyman JM, Kapan DD, Bennett SN. Human-Mosquito Contact: A Missing Link in Our Understanding of Mosquito-Borne Disease Transmission Dynamics. ANNALS OF THE ENTOMOLOGICAL SOCIETY OF AMERICA 2021; 114:397-414. [PMID: 34249219 PMCID: PMC8266639 DOI: 10.1093/aesa/saab011] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Indexed: 05/26/2023]
Abstract
Despite the critical role that contact between hosts and vectors, through vector bites, plays in driving vector-borne disease (VBD) transmission, transmission risk is primarily studied through the lens of vector density and overlooks host-vector contact dynamics. This review article synthesizes current knowledge of host-vector contact with an emphasis on mosquito bites. It provides a framework including biological and mathematical definitions of host-mosquito contact rate, blood-feeding rate, and per capita biting rates. We describe how contact rates vary and how this variation is influenced by mosquito and vertebrate factors. Our framework challenges a classic assumption that mosquitoes bite at a fixed rate determined by the duration of their gonotrophic cycle. We explore alternative ecological assumptions based on the functional response, blood index, forage ratio, and ideal free distribution within a mechanistic host-vector contact model. We highlight that host-vector contact is a critical parameter that integrates many factors driving disease transmission. A renewed focus on contact dynamics between hosts and vectors will contribute new insights into the mechanisms behind VBD spread and emergence that are sorely lacking. Given the framework for including contact rates as an explicit component of mathematical models of VBD, as well as different methods to study contact rates empirically to move the field forward, researchers should explicitly test contact rate models with empirical studies. Such integrative studies promise to enhance understanding of extrinsic and intrinsic factors affecting host-vector contact rates and thus are critical to understand both the mechanisms driving VBD emergence and guiding their prevention and control.
Collapse
Affiliation(s)
- Panpim Thongsripong
- Department of Microbiology, California Academy of Sciences, 55 Music Concourse Drive, San Francisco, CA 94118, USA
| | - James M Hyman
- Department of Mathematics, Tulane University, 6823 St. Charles Avenue, New Orleans, LA 70118, USA
| | - Durrell D Kapan
- Department of Entomology and Center for Comparative Genomics, Institute of Biodiversity Sciences and Sustainability, California Academy of Sciences, 55 Music Concourse Drive, San Francisco, CA 94118, USA
- Center for Conservation and Research Training, Pacific Biosciences Research Center, University of Hawai’i at Manoa, 3050 Maile Way, Honolulu, HI 96822
| | - Shannon N Bennett
- Department of Microbiology, California Academy of Sciences, 55 Music Concourse Drive, San Francisco, CA 94118, USA
| |
Collapse
|
14
|
Dehghankar M, Maleki-Ravasan N, Tahghighi A, Karimian F, Karami M. Bioactivities of rose-scented geranium nanoemulsions against the larvae of Anopheles stephensi and their gut bacteria. PLoS One 2021; 16:e0246470. [PMID: 33556110 PMCID: PMC7870081 DOI: 10.1371/journal.pone.0246470] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 01/19/2021] [Indexed: 11/24/2022] Open
Abstract
Anopheles stephensi with three different biotypes is a major vector of malaria in Asia. It breeds in a wide range of habitats. Therefore, safer and more sustainable methods are needed to control its immature stages rather than chemical pesticides. The larvicidal and antibacterial properties of the Pelargonium roseum essential oil (PREO) formulations were investigated against mysorensis and intermediate forms of An. stephensi in laboratory conditions. A series of nanoemulsions containing different amounts of PREO, equivalent to the calculated LC50 values for each An. stephensi form, and various quantities of surfactants and co-surfactants were developed. The physical and morphological properties of the most lethal formulations were also determined. PREO and its major components, i.e. citronellol (21.34%), L-menthone (6.41%), linalool (4.214%), and geraniol (2.19%), showed potent larvicidal activity against the studied mosquitoes. The LC50/90 values for mysorensis and intermediate forms were computed as 11.44/42.42 ppm and 12.55/47.69 ppm, respectively. The F48/F44 nanoformulations with 94% and 88% lethality for the mysorensis and intermediate forms were designated as optimized formulations. The droplet size, polydispersity index, and zeta-potential for F48/F44 were determined as 172.8/90.95 nm, 0.123/0.183, and -1.08/-2.08 mV, respectively. These results were also confirmed by TEM analysis. Prepared formulations displayed antibacterial activity against larval gut bacteria in the following order of decreasing inhibitory: LC90, optimized nanoemulsions, and LC50. PREO-based formulations were more effective against mysorensis than intermediate. Compared to the crude PREO, the overall larvicidal activity of all nanoformulations boosted by 20% and the optimized formulations by 50%. The sensitivity of insect gut bacteria may be a crucial factor in determining the outcome of the effect of toxins on target insects. The formulations designed in the present study may be a good option as a potent and selective larvicide for An. stephensi.
Collapse
Affiliation(s)
- Maryam Dehghankar
- Faculty of Basic Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
- Malaria and Vector Research Group, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Naseh Maleki-Ravasan
- Malaria and Vector Research Group, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
- Department of Parasitology, Pasteur Institute of Iran, Tehran, Iran
- * E-mail: (NMR); (AT)
| | - Azar Tahghighi
- Malaria and Vector Research Group, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
- Laboratory of Medicinal Chemistry, Department of Clinical Research, Pasteur Institute of Iran, Tehran, Iran
- * E-mail: (NMR); (AT)
| | - Fateh Karimian
- Department of Medical Entomology and Vector Control, School of Public Health, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mohsen Karami
- Department of Parasitology and Mycology, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
15
|
Graumans W, Jacobs E, Bousema T, Sinnis P. When Is a Plasmodium-Infected Mosquito an Infectious Mosquito? Trends Parasitol 2020; 36:705-716. [PMID: 32620501 DOI: 10.1016/j.pt.2020.05.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 12/19/2022]
Abstract
Plasmodium parasites experience significant bottlenecks as they transit through the mosquito and are transmitted to their mammalian host. Oocyst prevalence on mosquito midguts and sporozoite prevalence in salivary glands are nevertheless commonly used to confirm successful malaria transmission, assuming that these are reliable indicators of the mosquito's capacity to give rise to secondary infections. Here we discuss recent insights in sporogonic development and transmission bottlenecks for Plasmodium. We highlight critical gaps in our knowledge and frame their importance in understanding the human and mosquito reservoirs of infection. A better understanding of the events that lead to successful inoculation of infectious sporozoites by mosquitoes is critical to designing effective interventions to shrink the malaria map.
Collapse
Affiliation(s)
- Wouter Graumans
- Radboud University Medical Center, Radboud Institute for Health Sciences, Department of Medical Microbiology, Nijmegen, The Netherlands
| | - Ella Jacobs
- Department of Molecular Microbiology and Immunology, and Johns Hopkins Malaria Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Teun Bousema
- Radboud University Medical Center, Radboud Institute for Health Sciences, Department of Medical Microbiology, Nijmegen, The Netherlands; Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK.
| | - Photini Sinnis
- Department of Molecular Microbiology and Immunology, and Johns Hopkins Malaria Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
16
|
Mayton EH, Tramonte AR, Wearing HJ, Christofferson RC. Age-structured vectorial capacity reveals timing, not magnitude of within-mosquito dynamics is critical for arbovirus fitness assessment. Parasit Vectors 2020; 13:310. [PMID: 32539759 PMCID: PMC7296759 DOI: 10.1186/s13071-020-04181-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 06/10/2020] [Indexed: 12/20/2022] Open
Abstract
Background Transmission dynamics of arboviruses like Zika virus are often evaluated by vector competence (the proportion of infectious vectors given exposure) and the extrinsic incubation period (EIP, the time it takes for a vector to become infectious), but vector age is another critical driver of transmission dynamics. Vectorial capacity (VC) is a measure of transmission potential of a vector-pathogen system, but how these three components, EIP, vector competence and vector age, affect VC in concert still needs study. Methods The interaction of vector competence, EIP, and mosquito age at the time of infection acquisition (Ageacquisition) was experimentally measured in an Aedes aegypti-ZIKV model system, as well as the age-dependence of probability of survival and the willingness to bite. An age-structured vectorial capacity framework (VCage) was then developed using both EIPMin and EIPMax, defined as the time to first observed minimum proportion of transmitting mosquitoes and the time to observed maximum proportion of transmitting mosquitoes. Results The within-mosquito dynamics of vector competence/EIP were not significant among treatments where mosquitoes were exposed at different ages. However, VCage revealed: (i) age-dependence in vector-virus interactions is important for transmission success; (ii) lower vector competence but at shorter EIPs was sufficient for transmission perpetuation; and (iii) R0 may be overestimated by using non-age-structured VC. Conclusions The results indicate that ultimately the temporal component of the virus-vector dynamics is most critical, especially when exposure occurred at advanced mosquito age. While our study is limited to a single virus-vector system, and a multitude of other factors affect both vector competence and mosquito mortality, our methods can be extrapolated to these other scenarios. Results indicate that how ‘highly’ or ‘negligibly’ competent vectors are categorized may need adjustment.![]()
Collapse
Affiliation(s)
- E Handly Mayton
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - A Ryan Tramonte
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA
| | - Helen J Wearing
- Departments of Biology and Mathematics & Statistics, University of New Mexico, Albuquerque, NM, USA
| | - Rebecca C Christofferson
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, USA. .,Center for Computation and Technology, Louisiana State University, Baton Rouge, LA, USA.
| |
Collapse
|
17
|
Aleshnick M, Ganusov VV, Nasir G, Yenokyan G, Sinnis P. Experimental determination of the force of malaria infection reveals a non-linear relationship to mosquito sporozoite loads. PLoS Pathog 2020; 16:e1008181. [PMID: 32453765 PMCID: PMC7295235 DOI: 10.1371/journal.ppat.1008181] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 06/15/2020] [Accepted: 04/04/2020] [Indexed: 11/30/2022] Open
Abstract
Plasmodium sporozoites are the infective stage of the malaria parasite. Though this is a bottleneck for the parasite, the quantitative dynamics of transmission, from mosquito inoculation of sporozoites to patent blood-stage infection in the mammalian host, are poorly understood. Here we utilize a rodent model to determine the probability of malaria infection after infectious mosquito bite, and consider the impact of mosquito parasite load, blood-meal acquisition, probe-time, and probe location, on infection probability. We found that infection likelihood correlates with mosquito sporozoite load and, to a lesser degree, the duration of probing, and is not dependent upon the mosquito’s ability to find blood. The relationship between sporozoite load and infection probability is non-linear and can be described by a set of models that include a threshold, with mosquitoes harboring over 10,000 salivary gland sporozoites being significantly more likely to initiate a malaria infection. Overall, our data suggest that the small subset of highly infected mosquitoes may contribute disproportionally to malaria transmission in the field and that quantifying mosquito sporozoite loads could aid in predicting the force of infection in different transmission settings. Malaria is a leading cause of death in many parts of the world. Infection is initiated when infected Anopheles mosquitoes inject sporozoites as they look for blood. Though transmission is a bottleneck for the parasite and thus a good point for intervention, many aspects of transmission remain poorly understood. In this study, using a rodent model of malaria, we found that the majority of infective bites do not result in malaria infection. Furthermore, we found that the bites of mosquitoes with heavy parasite burdens are significantly more likely to result in blood stage infection. These data have important implications for designing interventions targeting transmission stages of the malaria parasite as they suggest that reducing parasite loads, even without completely eliminating them, could be effective against disease spread. We also found that mosquitoes that probe but do not succeed in finding blood are equally likely to initiate infection, an important finding for human vaccine trials. Overall this work contributes to our understanding of the epidemiology of malaria and should aid in the development of malaria elimination strategies.
Collapse
Affiliation(s)
- Maya Aleshnick
- Johns Hopkins Malaria Research Institute and Department of Molecular Microbiology & Immunology, Baltimore, Maryland, United States of America
| | - Vitaly V. Ganusov
- Departments of Microbiology and Mathematics, University of Tennessee, Knoxville Tennessee, United States of America
| | - Gibran Nasir
- Johns Hopkins Malaria Research Institute and Department of Molecular Microbiology & Immunology, Baltimore, Maryland, United States of America
| | - Gayane Yenokyan
- Johns Hopkins Biostatistics Center, Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Photini Sinnis
- Johns Hopkins Malaria Research Institute and Department of Molecular Microbiology & Immunology, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
18
|
Horta MF, Andrade LO, Martins-Duarte ÉS, Castro-Gomes T. Cell invasion by intracellular parasites - the many roads to infection. J Cell Sci 2020; 133:133/4/jcs232488. [PMID: 32079731 DOI: 10.1242/jcs.232488] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Intracellular parasites from the genera Toxoplasma, Plasmodium, Trypanosoma, Leishmania and from the phylum Microsporidia are, respectively, the causative agents of toxoplasmosis, malaria, Chagas disease, leishmaniasis and microsporidiosis, illnesses that kill millions of people around the globe. Crossing the host cell plasma membrane (PM) is an obstacle these parasites must overcome to establish themselves intracellularly and so cause diseases. The mechanisms of cell invasion are quite diverse and include (1) formation of moving junctions that drive parasites into host cells, as for the protozoans Toxoplasma gondii and Plasmodium spp., (2) subversion of endocytic pathways used by the host cell to repair PM, as for Trypanosoma cruzi and Leishmania, (3) induction of phagocytosis as for Leishmania or (4) endocytosis of parasites induced by specialized structures, such as the polar tubes present in microsporidian species. Understanding the early steps of cell entry is essential for the development of vaccines and drugs for the prevention or treatment of these diseases, and thus enormous research efforts have been made to unveil their underlying biological mechanisms. This Review will focus on these mechanisms and the factors involved, with an emphasis on the recent insights into the cell biology of invasion by these pathogens.
Collapse
Affiliation(s)
- Maria Fátima Horta
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil
| | - Luciana Oliveira Andrade
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil
| | - Érica Santos Martins-Duarte
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil
| | - Thiago Castro-Gomes
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil
| |
Collapse
|
19
|
Julien JP, Wardemann H. Antibodies against Plasmodium falciparum malaria at the molecular level. Nat Rev Immunol 2019; 19:761-775. [DOI: 10.1038/s41577-019-0209-5] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2019] [Indexed: 12/11/2022]
|
20
|
Amelia F, Iyori M, Emran TB, Yamamoto DS, Genshi K, Otsuka H, Onoue Y, Yusuf Y, Islam A, Yoshida S. Down-selecting circumsporozoite protein-based malaria vaccine: A comparison of malaria sporozoite challenge model. Parasite Immunol 2019; 41:e12624. [PMID: 30883819 DOI: 10.1111/pim.12624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 02/28/2019] [Accepted: 03/08/2019] [Indexed: 12/18/2022]
Abstract
Plasmodium falciparum circumsporozoite protein (PfCSP) is the main target antigen in development of pre-erythrocytic malaria vaccines. To evaluate PfCSP vaccines in animal models, challenge by intravenous sporozoite injection is preferentially used. However, in clinical trials, vaccinated human volunteers are exposed to the bites of malaria-infected mosquitoes. In this study, we down-selected Escherichia coli-produced full-length PfCSP (PfCSP-F) and its three truncated PfCSPs based on their abilities to elicit immune response and protection in mice against two challenge models. We showed that immunization with three doses of PfCSP-F elicited high anti-PfCSP antibody titres and 100% protection against the bites of infected mosquitoes. Meanwhile, three-dose truncated PfCSP induced 60%-70% protection after immunization with each truncated PfCSP. Heterologous prime-boost immunization regimen with adenovirus-PfCSP-F and R32LR greatly induced complete protection against intravenous sporozoite injection. Our results suggest that Abs to both anti-repeat and anti-nonrepeat regions induced by PfCSP-F are required to confer complete protection against challenge by the bites of infected mosquitoes, whereas anti-repeat Abs play an important role in protection against intravenous sporozoite injection. Our findings provide a potential clinical application that PfCSP-F vaccine induces potent Abs capable of neutralizing sporozoites in the dermis inoculated by infected mosquitoes and subsequently sporozoites in the blood circulation.
Collapse
Affiliation(s)
- Fitri Amelia
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa, Japan
- Department of Chemistry, Universitas Negeri Padang, Padang, Indonesia
| | - Mitsuhiro Iyori
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa, Japan
| | - Talha Bin Emran
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa, Japan
| | - Daisuke S Yamamoto
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Shimotsuke, Japan
| | - Kento Genshi
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa, Japan
| | - Hiromu Otsuka
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa, Japan
| | - Yutaro Onoue
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa, Japan
| | - Yenni Yusuf
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa, Japan
| | - Ashekul Islam
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa, Japan
| | - Shigeto Yoshida
- Laboratory of Vaccinology and Applied Immunology, Kanazawa University School of Pharmacy, Kanazawa, Japan
| |
Collapse
|
21
|
De Niz M, Meehan GR, Brancucci NM, Marti M, Rotureau B, Figueiredo LM, Frischknecht F. Intravital imaging of host-parasite interactions in skin and adipose tissues. Cell Microbiol 2019; 21:e13023. [PMID: 30825872 PMCID: PMC6590052 DOI: 10.1111/cmi.13023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 02/06/2019] [Accepted: 02/08/2019] [Indexed: 12/20/2022]
Abstract
Intravital microscopy allows the visualisation of how pathogens interact with host cells and tissues in living animals in real time. This method has enabled key advances in our understanding of host-parasite interactions under physiological conditions. A combination of genetics, microscopy techniques, and image analysis have recently facilitated the understanding of biological phenomena in living animals at cellular and subcellular resolution. In this review, we summarise findings achieved by intravital microscopy of the skin and adipose tissues upon infection with various parasites, and we present a view into possible future applications of this method.
Collapse
Affiliation(s)
- Mariana De Niz
- Institute of Cell Biology, Heussler GroupUniversity of BernBernSwitzerland
- Wellcome Centre for Integrative ParasitologyUniversity of GlasgowGlasgowUK
| | - Gavin R. Meehan
- Wellcome Centre for Integrative ParasitologyUniversity of GlasgowGlasgowUK
| | - Nicolas M.B. Brancucci
- Malaria Gene Regulation Unit, Department of Medical Parasitology and Infection BiologySwiss Tropical and Public Health InstituteBaselSwitzerland
- University of BaselBaselSwitzerland
| | - Matthias Marti
- Wellcome Centre for Integrative ParasitologyUniversity of GlasgowGlasgowUK
| | - Brice Rotureau
- Trypanosome Transmission Group, Trypanosome Cell Biology Unit, Department of Parasites and Insect Vectors, INSERM U1201Institut PasteurParisFrance
| | - Luisa M. Figueiredo
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo AntunesUniversidade de LisboaLisbonPortugal
| | - Friedrich Frischknecht
- Integrative Parasitology, Centre for Infectious DiseasesUniversity of Heidelberg Medical SchoolHeidelbergGermany
| |
Collapse
|
22
|
Shimada M, Hirose Y, Shimizu K, Yamamoto DS, Hayakawa EH, Matsuoka H. Upper gastrointestinal pathophysiology due to mouse malaria Plasmodium berghei ANKA infection. Trop Med Health 2019; 47:18. [PMID: 30872946 PMCID: PMC6399856 DOI: 10.1186/s41182-019-0146-9] [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: 12/31/2018] [Accepted: 02/20/2019] [Indexed: 12/11/2022] Open
Abstract
Background Epigastric pain, vomiting, and other gastrointestinal problems are among the most important symptoms of malaria infection as they suggest the possibility that the condition is serious. Pathophysiologies such as gastric mucosal changes and delayed gastric emptying have been reported in serious cases of malaria infection. However, it is unclear whether or not pathophysiological involvement of the upper gastrointestinal tract occurs in Plasmodium berghei ANKA (PbA)-infected mice. Methods PbA-infective Anopheles mosquitoes were used to infect mice via the natural route of infection. Fifteen PbA-C57BL/6 mice were used as a cerebral malaria model and the same numbers of PbA-BALB/c mice were used as a cerebral malaria-resistant model, and then we investigated the pathophysiological involvement of the stomach and small intestine. Results On day 8 post infection, six PbA-C57BL/6 mice showed cerebral malaria and nine others had uncomplicated infection. All the PbA-C57BL/6 mice on that same day showed severe weight loss with multiple, red gastric patches and changes to the course of the small intestine with villus goblet cell enlargement. In addition, cerebral malaria cases showed gastric gas retention with submucosal edema and small intestinal shortening. In PbA-BALB/c mice, overextension of the stomach and gas retention were evident from week 2 after PbA infection, as well as changes to the course of the small intestine and mesenteric thinning with fragility. Conclusions We described the upper gastrointestinal pathophysiology representing new findings directly linked to malarial severity and subsequent death in PbA-infected mice as a mouse model of malaria infection.
Collapse
Affiliation(s)
- Mizuho Shimada
- 1Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke City, Tochigi 329-0498 Japan
| | - Yoshie Hirose
- 2Department of Pathology, Ashikaga Red Cross Hospital, 284-1, Yobe-cho, Ashikaga City, Tochigi 326-0843 Japan
| | - Kazuhiko Shimizu
- 2Department of Pathology, Ashikaga Red Cross Hospital, 284-1, Yobe-cho, Ashikaga City, Tochigi 326-0843 Japan
| | - Daisuke S Yamamoto
- 1Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke City, Tochigi 329-0498 Japan
| | - Eri H Hayakawa
- 1Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke City, Tochigi 329-0498 Japan
| | - Hiroyuki Matsuoka
- 1Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, 3311-1, Yakushiji, Shimotsuke City, Tochigi 329-0498 Japan
| |
Collapse
|
23
|
Abstract
Plasmodium sporozoites are injected into the skin as mosquitoes probe for blood. From here, they migrate through the dermis to find blood vessels which they enter in order to be rapidly carried to the liver, where they invade hepatocytes and develop into the next life cycle stage, the exoerythrocytic stage. Once sporozoites enter the blood circulation, they are found in hepatocytes within minutes. In contrast, sporozoite exit from the inoculation site resembles a slow trickle and occurs over several hours. Thus, sporozoites spend the majority of their extracellular time at the inoculation site, raising the hypothesis that this is when the malarial parasite is most vulnerable to antibody-mediated destruction. Here, we investigate this hypothesis and demonstrate that the neutralizing capacity of circulating antibodies is greater at the inoculation site than in the blood circulation. Furthermore, these antibodies are working, at least in part, by impacting sporozoite motility at the inoculation site. Using actively and passively immunized mice, we found that most parasites are either immobilized at the site of injection or display reduced motility, particularly in their net displacement. We also found that antibodies severely impair the entry of sporozoites into the bloodstream. Overall, our data suggest that antibodies targeting the migratory sporozoite exert a large proportion of their protective effect at the inoculation site.IMPORTANCE Studies in experimental animal models and humans have shown that antibodies against Plasmodium sporozoites abolish parasite infectivity and provide sterile immunity. While it is well documented that these antibodies can be induced after immunization with attenuated parasites or subunit vaccines, the mechanisms by and location in which they neutralize parasites have not been fully elucidated. Here, we report studies indicating that these antibodies display a significant portion of their protective effect in the skin after injection of sporozoites and that one mechanism by which they work is by impairing sporozoite motility, thus diminishing their ability to reach blood vessels. These results suggest that immune protection against malaria begins at the earliest stages of parasite infection and emphasize the need of performing parasite challenge in the skin for the evaluation of protective immunity.
Collapse
|
24
|
A recombinant antibody against Plasmodium vivax UIS4 for distinguishing replicating from dormant liver stages. Malar J 2018; 17:370. [PMID: 30333026 PMCID: PMC6192329 DOI: 10.1186/s12936-018-2519-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 10/08/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Plasmodium vivax is the most geographically widespread of the human malaria parasites, causing 50,000 to 100,000 deaths annually. Plasmodium vivax parasites have the unique feature of forming dormant liver stages (hypnozoites) that can reactivate weeks or months after a parasite-infected mosquito bite, leading to new symptomatic blood stage infections. Efforts to eliminate P. vivax malaria likely will need to target the persistent hypnozoites in the liver. Therefore, research on P. vivax liver stages necessitates a marker for clearly distinguishing between actively replicating parasites and dormant hypnozoites. Hypnozoites possess a densely fluorescent prominence in the parasitophorous vacuole membrane (PVM) when stained with antibodies against the PVM-resident protein Upregulated in Infectious Sporozoites 4 (PvUIS4), resulting in a key feature recognizable for quantification of hypnozoites. Thus, PvUIS4 staining, in combination with the characteristic small size of the parasite, is currently the only hypnozoite-specific morphological marker available. RESULTS Here, the generation and validation of a recombinant monoclonal antibody against PvUIS4 (α-rUIS4 mAb) is described. The variable heavy and light chain domains of an α-PvUIS4 hybridoma were cloned into murine IgG1 and IgK expression vectors. These expression plasmids were co-transfected into HEK293 cells and mature IgG was purified from culture supernatants. It is shown that the α-rUIS4 mAb binds to its target with high affinity. It reliably stains the schizont PVM and the hypnozoite-specific PVM prominence, enabling the visual differentiation of hypnozoites from replicating liver stages by immunofluorescence assays in different in vitro settings, as well as in liver sections from P. vivax infected liver-chimeric mice. The antibody functions reliably against all four parasite isolates tested and will be an important tool in the identification of the elusive hypnozoite. CONCLUSIONS The α-rUIS4 mAb is a versatile tool for distinguishing replicating P. vivax liver stages from dormant hypnozoites, making it a valuable resource that can be deployed throughout laboratories worldwide.
Collapse
|
25
|
Douglas RG, Reinig M, Neale M, Frischknecht F. Screening for potential prophylactics targeting sporozoite motility through the skin. Malar J 2018; 17:319. [PMID: 30170589 PMCID: PMC6119338 DOI: 10.1186/s12936-018-2469-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 08/27/2018] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Anti-malarial compounds have not yet been identified that target the first obligatory step of infection in humans: the migration of Plasmodium sporozoites in the host dermis. This movement is essential to find and invade a blood vessel in order to be passively transported to the liver. Here, an imaging screening pipeline was established to screen for compounds capable of inhibiting extracellular sporozoites. METHODS Sporozoites expressing the green fluorescent protein were isolated from infected Anopheles mosquitoes, incubated with compounds from two libraries (MMV Malaria Box and a FDA-approved library) and imaged. Effects on in vitro motility or morphology were scored. In vivo efficacy of a candidate drug was investigated by treating mice ears with a gel prior to infectious mosquito bites. Motility was analysed by in vivo imaging and the progress of infection was monitored by daily blood smears. RESULTS Several compounds had a pronounced effect on in vitro sporozoite gliding or morphology. Notably, monensin sodium potently affected sporozoite movement while gramicidin S resulted in rounding up of sporozoites. However, pre-treatment of mice with a topical gel containing gramicidin did not reduce sporozoite motility and infection. CONCLUSIONS This approach shows that it is possible to screen libraries for inhibitors of sporozoite motility and highlighted the paucity of compounds in currently available libraries that inhibit this initial step of a malaria infection. Screening of diverse libraries is suggested to identify more compounds that could serve as leads in developing 'skin-based' malaria prophylactics. Further, strategies need to be developed that will allow compounds to effectively penetrate the dermis and thereby prevent exit of sporozoites from the skin.
Collapse
Affiliation(s)
- Ross G Douglas
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany.
| | - Miriam Reinig
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
| | - Matthew Neale
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
| | - Friedrich Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany.
| |
Collapse
|
26
|
Roth A, Adapa SR, Zhang M, Liao X, Saxena V, Goffe R, Li S, Ubalee R, Saggu GS, Pala ZR, Garg S, Davidson S, Jiang RHY, Adams JH. Unraveling the Plasmodium vivax sporozoite transcriptional journey from mosquito vector to human host. Sci Rep 2018; 8:12183. [PMID: 30111801 PMCID: PMC6093925 DOI: 10.1038/s41598-018-30713-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 08/03/2018] [Indexed: 02/07/2023] Open
Abstract
Malaria parasites transmitted by mosquito bite are remarkably efficient in establishing human infections. The infection process requires roughly 30 minutes and is highly complex as quiescent sporozoites injected with mosquito saliva must be rapidly activated in the skin, migrate through the body, and infect the liver. This process is poorly understood for Plasmodium vivax due to low infectivity in the in vitro models. To study this skin-to-liver-stage of malaria, we used quantitative bioassays coupled with transcriptomics to evaluate parasite changes linked with mammalian microenvironmental factors. Our in vitro phenotyping and RNA-seq analyses revealed key microenvironmental relationships with distinct biological functions. Most notable, preservation of sporozoite quiescence by exposure to insect-like factors coupled with strategic activation limits untimely activation of invasion-associated genes to dramatically increase hepatocyte invasion rates. We also report the first transcriptomic analysis of the P. vivax sporozoite interaction in salivary glands identifying 118 infection-related differentially-regulated Anopheles dirus genes. These results provide important new insights in malaria parasite biology and identify priority targets for antimalarial therapeutic interventions to block P. vivax infection.
Collapse
Affiliation(s)
- Alison Roth
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Swamy R Adapa
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Min Zhang
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Xiangyun Liao
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Vishal Saxena
- Molecular Parasitology and System Biology Lab, Department of Biological Sciences, Birla Institute of Technology and Science, Pilani, Rajasthan, India
| | - Raaven Goffe
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Suzanne Li
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Ratawan Ubalee
- Department of Entomology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Gagandeep S Saggu
- Laboratory of Malaria and Vector Research, National Institute of Allergic and Infectious Diseases, National Institute of Health, Rockville, Maryland, USA
| | - Zarna R Pala
- Molecular Parasitology and System Biology Lab, Department of Biological Sciences, Birla Institute of Technology and Science, Pilani, Rajasthan, India
| | - Shilpi Garg
- Molecular Parasitology and System Biology Lab, Department of Biological Sciences, Birla Institute of Technology and Science, Pilani, Rajasthan, India
| | - Silas Davidson
- Department of Entomology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Rays H Y Jiang
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, USA.
| | - John H Adams
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, USA.
| |
Collapse
|
27
|
Minkah NK, Schafer C, Kappe SHI. Humanized Mouse Models for the Study of Human Malaria Parasite Biology, Pathogenesis, and Immunity. Front Immunol 2018; 9:807. [PMID: 29725334 PMCID: PMC5917005 DOI: 10.3389/fimmu.2018.00807] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 04/03/2018] [Indexed: 12/25/2022] Open
Abstract
Malaria parasite infection continues to inflict extensive morbidity and mortality in resource-poor countries. The insufficiently understood parasite biology, continuously evolving drug resistance and the lack of an effective vaccine necessitate intensive research on human malaria parasites that can inform the development of new intervention tools. Humanized mouse models have been greatly improved over the last decade and enable the direct study of human malaria parasites in vivo in the laboratory. Nevertheless, no small animal model developed so far is capable of maintaining the complete life cycle of Plasmodium parasites that infect humans. The ultimate goal is to develop humanized mouse systems in which a Plasmodium infection closely reproduces all stages of a parasite infection in humans, including pre-erythrocytic infection, blood stage infection and its associated pathology, transmission as well as the human immune response to infection. Here, we discuss current humanized mouse models and the future directions that should be taken to develop next-generation models for human malaria parasite research.
Collapse
Affiliation(s)
- Nana K Minkah
- Center for Infectious Disease Research, Seattle, WA, United States
| | - Carola Schafer
- Center for Infectious Disease Research, Seattle, WA, United States
| | - Stefan H I Kappe
- Center for Infectious Disease Research, Seattle, WA, United States.,Department of Global Health, University of Washington, Seattle, WA, United States
| |
Collapse
|
28
|
Frischknecht F, Matuschewski K. Plasmodium Sporozoite Biology. Cold Spring Harb Perspect Med 2017; 7:cshperspect.a025478. [PMID: 28108531 DOI: 10.1101/cshperspect.a025478] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Plasmodium sporozoite transmission is a critical population bottleneck in parasite life-cycle progression and, hence, a target for prophylactic drugs and vaccines. The recent progress of a candidate antisporozoite subunit vaccine formulation to licensure highlights the importance of sporozoite transmission intervention in the malaria control portfolio. Sporozoites colonize mosquito salivary glands, migrate through the skin, penetrate blood vessels, breach the liver sinusoid, and invade hepatocytes. Understanding the molecular and cellular mechanisms that mediate the remarkable sporozoite journey in the invertebrate vector and the vertebrate host can inform evidence-based next-generation drug development programs and immune intervention strategies.
Collapse
Affiliation(s)
- Friedrich Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, 69120 Heidelberg, Germany
| | - Kai Matuschewski
- Department of Molecular Parasitology, Institute of Biology, Humboldt University Berlin, 10115 Berlin, Germany
| |
Collapse
|
29
|
Alfrd Mavondo GA, Tagumirwa MC. Asiatic acid-pectin hydrogel matrix patch transdermal delivery system influences parasitaemia suppression and inflammation reduction in P. berghei murine malaria infected Sprague-Dawley rats. ASIAN PAC J TROP MED 2016; 9:1172-1180. [PMID: 27955745 DOI: 10.1016/j.apjtm.2016.10.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 09/10/2016] [Accepted: 10/12/2016] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE To report the influence of transdermal delivery of asiatic acid (AA) in Plasmodium berghei-infected Sprague Dawley rats on physicochemical changes, %parasitaemia and associated pathophysiology. METHODS A topical once-off AA (5, 10, and 20 mg/kg)- or chloroquine (CHQ)-pectin patch was applied on the shaven dorsal neck region of Plasmodium berghei-infected Sprague Dawley rats (90-120 g) on day 7 after infection. Eating and drinking habits, weight changes, malaria effects and %parasitaemia were compared among animal groups over 21 d. RESULTS AA-pectin patch application preserved food and water intake together with %weight gain. All animals developed stable parasitaemia (15-20%) by day 7. AA doses suppressed parasitaemia significantly. AA 5 mg/kg patch was most effective. AA and CHQ displayed bimodal time-spaced peaks. CHQ patch had a longer time course to clear parasitaemia. CONCLUSIONS AA influences bio-physicochemical changes and parasitaemia suppression in dose dependent manner. In comparison by dose administered, AA has much better efficacy than CHQ. AA may be a useful antimalarial. AA and CHQ displays bimodal peaks suggesting possible synergism if used in combination therapy.
Collapse
Affiliation(s)
- Greanious Alfred Alfrd Mavondo
- Discipline of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu Natal, Westville Campus, Durban, 4000, South Africa; Pathology Department, Faculty of Medicine, National University of Science and Technology, Mpilo Hospital NUST Complex, Vera Road, P.O. AC939, Ascot, Bulawayo, Zimbabwe.
| | | |
Collapse
|
30
|
Salivary Gland Proteome during Adult Development and after Blood Feeding of Female Anopheles dissidens Mosquitoes (Diptera: Culicidae). PLoS One 2016; 11:e0163810. [PMID: 27669021 PMCID: PMC5036837 DOI: 10.1371/journal.pone.0163810] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 09/14/2016] [Indexed: 12/28/2022] Open
Abstract
Understanding changes in mosquito salivary proteins during the time that sporozoite maturation occurs and after blood feeding may give information regarding the roles of salivary proteins during the malarial transmission. Anopheles dissidens (formerly Anopheles barbirostris species A1) is a potential vector of Plasmodium vivax in Thailand. In this study, analyses of the proteomic profiles of female An. dissidens salivary glands during adult development and after blood feeding were carried out using two-dimensional gel electrophoresis coupled with nano-liquid chromatography-mass spectrometry. Results showed at least 17 major salivary gland proteins present from day one to day 21 post emergence at 8 different time points sampled. Although there was variation observed, the patterns of protein expression could be placed into one of four groups. Fifteen protein spots showed significant depletion after blood feeding with the percentages of the amount of depletion ranging from 8.5% to 68.11%. The overall results identified various proteins, including a putative mucin-like protein, an anti-platelet protein, a long form D7 salivary protein, a putative gVAG protein precursor, a D7-related 3.2 protein, gSG7 salivary proteins, and a gSG6 protein. These results allow better understanding of the changes of the salivary proteins during the adult mosquito development. They also provide candidate proteins to investigate any possible link or not between sporozoite maturation, or survival of skin stage sporozoites, and salivary proteins.
Collapse
|
31
|
Mavondo GA, Mkhwananzi BN, Mabandla MV, Musabayane CT. Asiatic acid influences parasitaemia reduction and ameliorates malaria anaemia in P. berghei infected Sprague-Dawley male rats. Altern Ther Health Med 2016; 16:357. [PMID: 27618936 PMCID: PMC5020548 DOI: 10.1186/s12906-016-1338-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 09/03/2016] [Indexed: 01/11/2023]
Abstract
BACKGROUND Current malaria treatment is either "anti-parasitic", "anti-infectivity" or both without addressing the pathophysiological derangement (anti-disease aspect) associated with the disease. Asiatic acid is a natural phytochemical with oxidant, antioxidant and anti-inflammatory properties whose effect on malarial and accompanying pathophysiology are yet to be investigated. Asiatic acid influence in P. berghei-infected Sprague Dawley rats on %parasitaemia and malarial anaemia were investigated. METHODS Plasmodium berghei-infected rats (90-120 g) were orally administered with Asiatic acid (5, 10, 20 mg/kg) and 30 mg/kg chloroquine as a positive control. Changes in %parasitaemia and haematological parameters in Asiatic acid administered rats were monitored in a 21 day study and compared to controls. RESULTS All animals developed stable parasitaemia (15-20 %) by day 7. Asiatic acid doses suppressed parasitaemia, normalised haematological measurements and influenced biophysical characteristics changes. Most positive changes were associated with intragastric administration of 10 mg/kg Asiatic acid dose. Peak %parasitaemia in Asiatic acid administration occurred at days 12 with a shorter time course compared to day 9 for chloroquine (30 mg/kg) treatment with a longer time course. CONCLUSIONS Oral Asiatic acid administration influenced %parasitaemia suppression, ameliorated malarial anaemia and increased biophysical properties on infected animals. Asiatic acid may be a replacement alternative for chloroquine treatment with concomitant amelioration of malaria pathophysiology. Due to different action time courses, Asiatic acid and chloroquine may be possible candidates in combination therapy.
Collapse
|
32
|
Wu JH, Li B, Wu MX. Laser-induced capillary leakage for blood biomarker detection and vaccine delivery via the skin. JOURNAL OF BIOPHOTONICS 2016; 9:676-682. [PMID: 26776718 PMCID: PMC4929029 DOI: 10.1002/jbio.201500226] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 11/06/2015] [Accepted: 01/02/2016] [Indexed: 06/05/2023]
Abstract
Circulation system is the center for coordination and communication of all organs in our body. Examination of any change in its analytes or delivery of therapeutic drugs into the system consists of important medical practice in today's medicine. Two recent studies prove that brief illumination of skin with a low powered laser, at wavelengths preferentially absorbed by hemoglobin, increases the amount of circulating biomarkers in the epidermis and upper dermis by more than 1,000-fold. When probe-coated microneedle arrays are applied into laser-treated skin, plasma blood biomarkers can be reliably, accurately, and sufficiently quantified in 15∼30 min assays, with a maximal detection in one hr in a manner independent of penetration depth or a molecular mass of the biomarker. Moreover, the laser treatment permits a high efficient delivery of radiation-attenuated malarial sporozoites (RAS) into the circulation, leading to robust immunity against malaria infections, whereas similar immunization at sham-treated skin elicits poor immune responses. Thus this technology can potentially instruct designs of small, portable devices for onsite, in mobile clinics, or at home for point-of-care diagnosis and drug/vaccine delivery via the skin. Laser-induced capillary leakage (a) to induce extravasation of circualing molecules only (b) or facilitate entry of attenuated malaria sporozoites into the capillary (c). Skin illumination with a laser preferably absorbed by hemoglobin causes dilation of the capillary beneath the skin. The extravasated molecules can be sufficiently measured in the skin or guide sporozoites to enter the vessel.
Collapse
Affiliation(s)
- Jeffrey H Wu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, 50 Blossom Street, Edwards 222, Boston, MA 02114, USA
| | - Bo Li
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, 50 Blossom Street, Edwards 222, Boston, MA 02114, USA
| | - Mei X Wu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School, 50 Blossom Street, Edwards 222, Boston, MA 02114, USA.
| |
Collapse
|
33
|
Mavondo GA, Mkhwananzi BN, Mabandla MV. Pre-infection administration of asiatic acid retards parasitaemia induction in Plasmodium berghei murine malaria infected Sprague-Dawley rats. Malar J 2016; 15:226. [PMID: 27098750 PMCID: PMC4839140 DOI: 10.1186/s12936-016-1278-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 04/12/2016] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Malaria prevention has remained a critical area in the absence of efficacious vaccines against malaria. Drugs currently used as chemotherapeutics are also used in chemoprophylaxis increasing possible drug resistance. Asiatic acid is a natural phytochemical with oxidant, antioxidant and anti-inflammatory properties with emerging anti-malarial potential. The influence of asiatic acid administration prior to Plasmodium berghei infection of Sprague-Dawley rats on parasitaemia induction is here reported. METHODS Sprague-Dawley rats (90-120 g) were administered with asiatic acid (10 mg/kg) 48 h before intraperitoneal infection with P. berghei. Parasitaemia induction and progression, food and water intake as well as weight were compared to 30 mg/kg chloroquine-treated and infected control rats during sub-chronic studies (21 days). RESULTS Asiatic acid pre-infection administration preserved food and water intake as well as increase in percentage weight gain of infected animals. In pre-infection treated animals, the pre-patent period was extended to day 6 from 72 h. Asiatic acid suppressed parasitaemia while oral chloroquine (30 mg/kg) did not influence malaria induction. CONCLUSIONS Per-oral, pre-infection, asiatic acid administration influenced parasitaemia patency and parasitaemia progression, food, water, and weight gain percentage. This may suggest possible chemoprophylaxis effects of asiatic acid in malaria.
Collapse
Affiliation(s)
- Greanious Alfred Mavondo
- Discipline of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu Natal, Westville Campus, Durban, 4000 South Africa
| | - Blessing Nkazimulo Mkhwananzi
- Discipline of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu Natal, Westville Campus, Durban, 4000 South Africa
| | - Musa Vuyisile Mabandla
- Discipline of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu Natal, Westville Campus, Durban, 4000 South Africa
| |
Collapse
|
34
|
Douglas RG, Amino R, Sinnis P, Frischknecht F. Active migration and passive transport of malaria parasites. Trends Parasitol 2015; 31:357-62. [PMID: 26001482 DOI: 10.1016/j.pt.2015.04.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 04/22/2015] [Accepted: 04/23/2015] [Indexed: 11/16/2022]
Abstract
Malaria parasites undergo a complex life cycle between their hosts and vectors. During this cycle the parasites invade different types of cells, migrate across barriers, and transfer from one host to another. Recent literature hints at a misunderstanding of the difference between active, parasite-driven migration and passive, circulation-driven movement of the parasite or parasite-infected cells in the various bodily fluids of mosquito and mammalian hosts. Because both active migration and passive transport could be targeted in different ways to interfere with the parasite, a distinction between the two ways the parasite uses to get from one location to another is essential. We discuss the two types of motion needed for parasite dissemination and elaborate on how they could be targeted by future vaccines or drugs.
Collapse
Affiliation(s)
- Ross G Douglas
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Rogerio Amino
- Unité de Biologie et Génétique du Paludisme, Département Parasites et Insectes Vecteurs, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Photini Sinnis
- Johns Hopkins Malaria Research Institute, Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, MD 21205, USA
| | - Freddy Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany.
| |
Collapse
|
35
|
Matsuoka H, Tomita H, Hattori R, Arai M, Hirai M. Visualization of Malaria Parasites in the Skin Using the Luciferase Transgenic Parasite, Plasmodium berghei. Trop Med Health 2014; 43:53-61. [PMID: 25859153 PMCID: PMC4361344 DOI: 10.2149/tmh.2014-18] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 11/13/2014] [Indexed: 01/25/2023] Open
Abstract
We produced a transgenic rodent malaria parasite (Plasmodium berghei) that contained the luciferase gene under a promoter region of elongation factor-1α. These transgenic (TG) parasites expressed luciferase in all stages of their life cycle, as previously reported. However, we were the first to succeed in observing sporozoites as a mass in mouse skin following their deposition by the probing of infective mosquitoes. Our transgenic parasites may have emitted stronger bioluminescence than previous TG parasites. The estimated number of injected sporozoites by mosquitoes was between 34 and 775 (median 80). Since luciferase activity diminished immediately after the death of the parasites, luciferase activity could be an indicator of the existence of live parasites. Our results indicated that sporozoites survived at the probed site for more than 42 hours. We also detected sporozoites in the liver within 15 min of the intravenous injection. Bioluminescence was not observed in the lung, kidney or spleen. We confirmed the observation that the liver was the first organ in which malaria parasites entered and increased in number.
Collapse
Affiliation(s)
- Hiroyuki Matsuoka
- Division of Medical Zoology, Jichi Medical University , 3311-1 Yakushiji, Shimotsuke-shi 329-0498, Japan
| | - Hiroyuki Tomita
- Division of Medical Zoology, Jichi Medical University , 3311-1 Yakushiji, Shimotsuke-shi 329-0498, Japan
| | - Ryuta Hattori
- Division of Medical Zoology, Jichi Medical University , 3311-1 Yakushiji, Shimotsuke-shi 329-0498, Japan
| | - Meiji Arai
- Division of Medical Zoology, Jichi Medical University , 3311-1 Yakushiji, Shimotsuke-shi 329-0498, Japan ; Department of International Medical Zoology, Graduate School of Medicine, Kagawa University , Miki-cho 761-0793, Japan
| | - Makoto Hirai
- Division of Medical Zoology, Jichi Medical University , 3311-1 Yakushiji, Shimotsuke-shi 329-0498, Japan ; Department of Parasitology, School of Medicine, Juntendo University , Bunkyo-ku, Tokyo 113-8421, Japan
| |
Collapse
|
36
|
Zuzarte-Luis V, Mota MM, Vigário AM. Malaria infections: what and how can mice teach us. J Immunol Methods 2014; 410:113-22. [PMID: 24837740 DOI: 10.1016/j.jim.2014.05.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 04/24/2014] [Accepted: 05/01/2014] [Indexed: 01/07/2023]
Abstract
Malaria imposes a horrific public health burden - hundreds of millions of infections and millions of deaths - on large parts of the world. While this unacceptable health burden and its economic and social impact have made it a focal point of the international development agenda, it became consensual that malaria control or elimination will be difficult to attain prior to gain a better understanding of the complex interactions occurring between its main players: Plasmodium, the causative agent of disease, and its hosts. Practical and ethical limitations exist regarding the ability to carry out research with human subjects or with human samples. In this review, we highlight how rodent models of infection have contributed significantly during the past decades to a better understanding of the basic biology of the parasite, host response and pathogenesis.
Collapse
Affiliation(s)
- Vanessa Zuzarte-Luis
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Maria M Mota
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal.
| | - Ana M Vigário
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal; Unidade de Ciências Médicas, Centro de Competência de Ciências da Vida, Universidade da Madeira, Funchal, Portugal.
| |
Collapse
|
37
|
Cator LJ, Lynch PA, Thomas MB, Read AF. Alterations in mosquito behaviour by malaria parasites: potential impact on force of infection. Malar J 2014; 13:164. [PMID: 24885783 PMCID: PMC4113138 DOI: 10.1186/1475-2875-13-164] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 04/21/2014] [Indexed: 11/13/2022] Open
Abstract
Background A variety of studies have reported that malaria parasites alter the behaviour of mosquitoes. These behavioural alterations likely increase transmission because they reduce the risk of vector death during parasite development and increase biting after parasites become infectious. Methods A mathematical model is used to investigate the potential impact of these behavioural alterations on the lifetime number of infectious bites delivered. The model is used to explore the importance of assumptions about the magnitude and distribution of mortality as well as the importance of extrinsic incubation period and gonotrophic cycle length. Additionally, the model is applied to four datasets taken from actual transmission settings. Results The impact of behavioural changes on the relative number of lifetime bites is highly dependent on assumptions about the distribution of mortality over the mosquito-feeding cycle. Even using fairly conservative estimates of these parameters and field collected data, the model outputs suggest that altered feeding could easily cause a doubling in the force of infection. Conclusions Infection-induced behavioural alterations have their greatest impact on the lifetime number of infectious bites in environments with high feeding-related adult mortality and many pre-infectious feeding cycles. Interventions that increase feeding-associated mortality are predicted to amplify the relative fitness benefits and hence enhance the strength of selection for behavioural alteration.
Collapse
Affiliation(s)
- Lauren J Cator
- Department of Life Sciences, Imperial College London, Ascot, UK.
| | | | | | | |
Collapse
|
38
|
Yamamoto DS, Yokomine T, Sumitani M, Yagi K, Matsuoka H, Yoshida S. Visualization and live imaging analysis of a mosquito saliva protein in host animal skin using a transgenic mosquito with a secreted luciferase reporter system. INSECT MOLECULAR BIOLOGY 2013; 22:685-693. [PMID: 24118655 DOI: 10.1111/imb.12055] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Mosquitoes inject saliva into a vertebrate host during blood feeding. The analysis of mosquito saliva in host skin is important for the elucidation of the inflammatory responses to mosquito bites, the development of antithrombotic drugs, and the transmission-blocking of vector-borne diseases. We produced transgenic Anopheles stephensi mosquitoes expressing the secretory luciferase protein (MetLuc) fused to a saliva protein (AAPP) in the salivary glands. The transgene product (AAPP-MetLuc) of transgenic mosquitoes exhibited both luciferase activity as a MetLuc and binding activity to collagen as an AAPP. The detection of luminescence in the skin of mice bitten by transgenic mosquitoes showed that AAPP-MetLuc was injected into the skin as a component of saliva via blood feeding. AAPP-MetLuc remained at the mosquito bite site in host skin with luciferase activity for at least 4 h after blood feeding. AAPP was also suspected of remaining at the site of injury caused by the mosquito bite and blocking platelet aggregation by binding to collagen. These results demonstrated the establishment of visualization and time-lapse analysis of mosquito saliva in living vertebrate host skin. This technique may facilitate the analysis of mosquito saliva after its injection into host skin, and the development of new drugs and disease control strategies.
Collapse
Affiliation(s)
- D S Yamamoto
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, Shimotsuke, Japan
| | | | | | | | | | | |
Collapse
|
39
|
Abstract
Malaria, which is caused by Plasmodium spp., starts with an asymptomatic phase, during which sporozoites, the parasite form that is injected into the skin by a mosquito, develop into merozoites, the form that infects erythrocytes. This pre-erythrocytic phase is still the most enigmatic in the parasite life cycle, but has long been recognized as an attractive vaccination target. In this Review, we present what has been learned in recent years about the natural history of the pre-erythrocytic stages, mainly using intravital imaging in rodents. We also consider how this new knowledge is in turn changing our understanding of the immune response mounted by the host against the pre-erythrocytic forms.
Collapse
|
40
|
Vanderberg JP. Imaging mosquito transmission of Plasmodium sporozoites into the mammalian host: immunological implications. Parasitol Int 2013; 63:150-64. [PMID: 24060541 DOI: 10.1016/j.parint.2013.09.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Accepted: 09/11/2013] [Indexed: 10/26/2022]
Abstract
The malaria infection is initiated in mammals by injection of the sporozoite stage of the parasite through the bite of Plasmodium-infected, female Anopheles mosquitoes. Sporozoites are injected into extravascular portions of the skin while the mosquito is probing for a blood source. Sporozoite gliding motility allows them to locate and penetrate blood vessels of the dermis or subcutaneous tissues; once in the blood, they reach the liver, within which they continue their development. Some of the injected parasites invade dermal lymph vessels and travel to the proximal draining lymphatic node, where they interact with host immunocytes. The host responds to viable or attenuated sporozoites with antibodies directed against the immunodominant circumsporozoite protein (CSP), as well as against other sporozoite proteins. These CSP antibodies can inhibit the numbers of sporozoites injected by mosquitoes and the motility of those injected into the skin. This first phase of the immune response is followed by cell-mediated immunity involving CD8 T-cells directed against the developing liver stage of the parasite. This review discusses the early history of imaging studies, and focuses on the role that imaging has played in enabling a better understanding of both the induction and effector functions of the immune responses against sporozoites.
Collapse
Affiliation(s)
- Jerome P Vanderberg
- Division of Medical Parasitology, Department of Microbiology, New York University School of Medicine, 341 E 25th Street, New York, NY 10010, USA.
| |
Collapse
|
41
|
The skin: where malaria infection and the host immune response begin. Semin Immunopathol 2012; 34:787-92. [PMID: 23053392 DOI: 10.1007/s00281-012-0345-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 09/13/2012] [Indexed: 10/27/2022]
Abstract
Infection by malaria parasites begins with the inoculation of sporozoites into the skin of the host. The early events following sporozoite deposition in the dermis are critical for both the establishment of malaria infection and for the induction of protective immune responses. The initial sporozoite inoculum is generally low, and only a small percentage of these sporozoites successfully reach the liver and grow to the next life cycle stage, making this a significant bottleneck for the parasite. Recent studies highlight the importance of sporozoite motility and host cell traversal in dermal exit. Importantly, protective immune responses against sporozoites and liver stages of Plasmodium are induced by dendritic cells in the lymph node draining the skin inoculation site. The cellular, molecular, and immunological events that occur in the skin and associated lymph nodes are the topic of this review.
Collapse
|
42
|
Stone W, Bousema T, Jones S, Gesase S, Hashim R, Gosling R, Carneiro I, Chandramohan D, Theander T, Ronca R, Modiano D, Arcà B, Drakeley C. IgG responses to Anopheles gambiae salivary antigen gSG6 detect variation in exposure to malaria vectors and disease risk. PLoS One 2012; 7:e40170. [PMID: 22768250 PMCID: PMC3387013 DOI: 10.1371/journal.pone.0040170] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2012] [Accepted: 06/05/2012] [Indexed: 01/05/2023] Open
Abstract
Assessment of exposure to malaria vectors is important to our understanding of spatial and temporal variations in disease transmission and facilitates the targeting and evaluation of control efforts. Recently, an immunogenic Anopheles gambiae salivary protein (gSG6) was identified and proposed as the basis of an immuno-assay determining exposure to Afrotropical malaria vectors. In the present study, IgG responses to gSG6 and 6 malaria antigens (CSP, AMA-1, MSP-1, MSP-3, GLURP R1, and GLURP R2) were compared to Anopheles exposure and malaria incidence in a cohort of children from Korogwe district, Tanzania, an area of moderate and heterogeneous malaria transmission. Anti-gSG6 responses above the threshold for seropositivity were detected in 15% (96/636) of the children, and were positively associated with geographical variations in Anopheles exposure (OR 1.25, CI 1.01–1.54, p = 0.04). Additionally, IgG responses to gSG6 in individual children showed a strong positive association with household level mosquito exposure. IgG levels for all antigens except AMA-1 were associated with the frequency of malaria episodes following sampling. gSG6 seropositivity was strongly positively associated with subsequent malaria incidence (test for trend p = 0.004), comparable to malaria antigens MSP-1 and GLURP R2. Our results show that the gSG6 assay is sensitive to micro-epidemiological variations in exposure to Anopheles mosquitoes, and provides a correlate of malaria risk that is unrelated to immune protection. While the technique requires further evaluation in a range of malaria endemic settings, our findings suggest that the gSG6 assay may have a role in the evaluation and planning of targeted and preventative anti-malaria interventions.
Collapse
Affiliation(s)
- Will Stone
- Department of Immunity and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
- Department of Medical Microbiology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Teun Bousema
- Department of Immunity and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
- Department of Medical Microbiology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Sophie Jones
- Department of Immunity and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Samwel Gesase
- National Institute for Medical Research, Tanga, Tanzania
| | | | - Roly Gosling
- Global Health Group, University of California San Francisco (UCSF), San Francisco, California, United States of America
| | - Ilona Carneiro
- Department of Immunity and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Daniel Chandramohan
- Department of Immunity and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Thor Theander
- Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Raffaele Ronca
- Department of Structural and Functional Biology, University “Federico II”, Naples, Italy
| | - David Modiano
- Parasitology Section, Department of Public Health and Infectious Diseases, University “La Sapienza”, Rome, Italy
| | - Bruno Arcà
- Department of Structural and Functional Biology, University “Federico II”, Naples, Italy
- Parasitology Section, Department of Public Health and Infectious Diseases, University “La Sapienza”, Rome, Italy
| | - Chris Drakeley
- Department of Immunity and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
- * E-mail:
| |
Collapse
|
43
|
Guilbride DL, Guilbride PD, Gawlinski P. Malaria's deadly secret: a skin stage. Trends Parasitol 2012; 28:142-50. [DOI: 10.1016/j.pt.2012.01.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Revised: 01/04/2012] [Accepted: 01/05/2012] [Indexed: 11/28/2022]
|
44
|
Plasmodium vinckei: infectivity of arteether-sensitive and arteether-resistant parasites in different strains of mice. Parasitol Res 2011; 109:1143-9. [PMID: 21479576 DOI: 10.1007/s00436-011-2358-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Accepted: 03/16/2011] [Indexed: 10/18/2022]
Abstract
Malaria is one of the most lethal parasitic infections in the world. The lethality of the parasite depends on the rate of multiplication of the parasite within host erythrocytes. Different strains of the malaria parasite often respond in a different way to the same strain of mice or vice versa. In the present study, we investigated the course of infection of the arteether-sensitive and arteether-resistant Plasmodium vinckei parasites in Swiss albino AKR (inbred) and AJ (outbred) mice. The higher parasite burden and mortality were observed in the sensitive parasite-infected mice, whereas the infection with the resistant parasite was non-lethal. Resistant parasite-infected mice developed a moderate level of parasitemia that decreased gradually throughout the infection. The microscopic examination suggests that the resistant parasite invades reticulocytes more efficiently than normocytes, regardless of the mouse strain examined. Since the reticulocytes are rare in blood circulation, it limits the increase in parasite proliferations, while arteether-sensitive parasites can invade both mature normocytes and reticulocytes, resulting in the mortality of the mice. However, treatment with phenylhydrazine in Swiss mice results in reticulocytosis, which transforms the non-lethal resistant parasites to produce lethal infections. Our findings demonstrate that the characteristic response during infections with the arteether-resistant strain is dependent on the availability of reticulocytes in peripheral blood circulation. We can use this model for identifying the interaction between host and artemisinin derivative-resistant parasites.
Collapse
|
45
|
Rennenberg A, Lehmann C, Heitmann A, Witt T, Hansen G, Nagarajan K, Deschermeier C, Turk V, Hilgenfeld R, Heussler VT. Exoerythrocytic Plasmodium parasites secrete a cysteine protease inhibitor involved in sporozoite invasion and capable of blocking cell death of host hepatocytes. PLoS Pathog 2010; 6:e1000825. [PMID: 20361051 PMCID: PMC2845656 DOI: 10.1371/journal.ppat.1000825] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2009] [Accepted: 02/18/2010] [Indexed: 11/18/2022] Open
Abstract
Plasmodium parasites must control cysteine protease activity that is critical for hepatocyte invasion by sporozoites, liver stage development, host cell survival and merozoite liberation. Here we show that exoerythrocytic P. berghei parasites express a potent cysteine protease inhibitor (PbICP, P. berghei inhibitor of cysteine proteases). We provide evidence that it has an important function in sporozoite invasion and is capable of blocking hepatocyte cell death. Pre-incubation with specific anti-PbICP antiserum significantly decreased the ability of sporozoites to infect hepatocytes and expression of PbICP in mammalian cells protects them against peroxide- and camptothecin-induced cell death. PbICP is secreted by sporozoites prior to and after hepatocyte invasion, localizes to the parasitophorous vacuole as well as to the parasite cytoplasm in the schizont stage and is released into the host cell cytoplasm at the end of the liver stage. Like its homolog falstatin/PfICP in P. falciparum, PbICP consists of a classical N-terminal signal peptide, a long N-terminal extension region and a chagasin-like C-terminal domain. In exoerythrocytic parasites, PbICP is posttranslationally processed, leading to liberation of the C-terminal chagasin-like domain. Biochemical analysis has revealed that both full-length PbICP and the truncated C-terminal domain are very potent inhibitors of cathepsin L-like host and parasite cysteine proteases. The results presented in this study suggest that the inhibitor plays an important role in sporozoite invasion of host cells and in parasite survival during liver stage development by inhibiting host cell proteases involved in programmed cell death.
Collapse
Affiliation(s)
- Annika Rennenberg
- Bernhard Nocht Institute for Tropical Medicine, Department of Molecular Parasitology, Hamburg, Germany
| | - Christine Lehmann
- Bernhard Nocht Institute for Tropical Medicine, Department of Molecular Parasitology, Hamburg, Germany
| | - Anna Heitmann
- Bernhard Nocht Institute for Tropical Medicine, Department of Molecular Parasitology, Hamburg, Germany
| | - Tina Witt
- Bernhard Nocht Institute for Tropical Medicine, Department of Molecular Parasitology, Hamburg, Germany
| | - Guido Hansen
- Institute of Biochemistry, Center for Structural and Cell Biology in Medicine, University of Lübeck, Lübeck, Germany
| | - Krishna Nagarajan
- Institute of Biochemistry, Center for Structural and Cell Biology in Medicine, University of Lübeck, Lübeck, Germany
| | - Christina Deschermeier
- Bernhard Nocht Institute for Tropical Medicine, Department of Molecular Parasitology, Hamburg, Germany
| | - Vito Turk
- Josef Stefan Institute, Department of Biochemistry, Molecular and Structural Biology, Ljubljana, Slovenia
| | - Rolf Hilgenfeld
- Institute of Biochemistry, Center for Structural and Cell Biology in Medicine, University of Lübeck, Lübeck, Germany
| | - Volker T. Heussler
- Bernhard Nocht Institute for Tropical Medicine, Department of Molecular Parasitology, Hamburg, Germany
- * E-mail:
| |
Collapse
|
46
|
Conteh S, Chattopadhyay R, Anderson C, Hoffman SL. Plasmodium yoelii-infected A. stephensi inefficiently transmit malaria compared to intravenous route. PLoS One 2010; 5:e8947. [PMID: 20126610 PMCID: PMC2812485 DOI: 10.1371/journal.pone.0008947] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2009] [Accepted: 01/11/2010] [Indexed: 11/19/2022] Open
Abstract
It was recently reported that when mosquitoes infected with P. berghei sporozoites feed on mice, they deposit approximately 100–300 sporozoites in the dermis. When we inoculate P. yoelii (Py) sporozoites intravenously (IV) into BALB/c mice, the 50% infectious dose (ID50) is often less than 3 sporozoites, indicating that essentially all Py sporozoites in salivary glands are infectious. Thus, it should only take the bite of one infected mosquito to infect 100% of mice. In human subjects, it takes the bite of at least 5 P. falciparum-infected mosquitoes to achieve 100% blood stage infection. Exposure to 1–2 infected mosquitoes only leads to blood stage infection in approximately 50% of subjects. If mosquitoes carrying Py sporozoites inoculate 100–300 sporozoites per bite, and 1 to 2 mosquito bites achieve 50% blood stage infection rates, then this would suggest that the majority of sporozoites inoculated by mosquitoes into the dermis are not responsible for a productive infection, or that a significant number of sporozoite-infected mosquitoes do not inoculate any sporozoites. The objective of this study was to determine if this is the case. We therefore studied the infectivity to mice of the bites of 1, 2, 4, or 5–8 Py-infected mosquitoes. The bite of one Py sporozoite-infected mosquito caused blood stage infection in 41.4% (12/29) of mice, two bites infected 66.7% (22/33), four bites infected 75% (18/24), and five to eight bites infected 100% (21/21). These findings demonstrate that inoculation of sporozoites by mosquito bite is much less efficient than IV inoculation of Py sporozoites by needle and syringe. Such data may have implications for determining the best route and dose of administration to humans of our attenuated P. falciparum sporozoite vaccine, the scientific basis of which is immunity by bites from irradiated infected mosquitoes, and suggest that the challenge is to develop a method of administration that approximates IV inoculation, not one that mimics mosquito bite.
Collapse
Affiliation(s)
- Solomon Conteh
- Sanaria Inc., Rockville, Maryland, United States of America
| | | | | | | |
Collapse
|
47
|
Hegge S, Kudryashev M, Smith A, Frischknecht F. Automated classification of Plasmodium sporozoite movement patterns reveals a shift towards productive motility during salivary gland infection. Biotechnol J 2009; 4:903-13. [PMID: 19455538 DOI: 10.1002/biot.200900007] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The invasive stages of malaria and other apicomplexan parasites use a unique motility machinery based on actin, myosin and a number of parasite-specific proteins to invade host cells and tissues. The crucial importance of this motility machinery at several stages of the life cycle of these parasites makes the individual components potential drug targets. The different stages of the malaria parasite exhibit strikingly diverse movement patterns, likely reflecting the varied needs to achieve successful invasion. Here, we describe a Tool for Automated Sporozoite Tracking (ToAST) that allows the rapid simultaneous analysis of several hundred motile Plasmodium sporozoites, the stage of the malaria parasite transmitted by the mosquito. ToAST reliably categorizes different modes of sporozoite movement and can be used for both tracking changes in movement patterns and comparing overall movement parameters, such as average speed or the persistence of sporozoites undergoing a certain type of movement. This allows the comparison of potentially small differences between distinct parasite populations and will enable screening of drug libraries to find inhibitors of sporozoite motility. Using ToAST, we find that isolated sporozoites change their movement patterns towards productive motility during the first week after infection of mosquito salivary glands.
Collapse
Affiliation(s)
- Stephan Hegge
- Department of Parasitology, Hygiene Institute, University of Heidelberg Medical School, Heidelberg, Germany
| | | | | | | |
Collapse
|
48
|
Kinetics of mosquito-injected Plasmodium sporozoites in mice: fewer sporozoites are injected into sporozoite-immunized mice. PLoS Pathog 2009; 5:e1000399. [PMID: 19390607 PMCID: PMC2667259 DOI: 10.1371/journal.ppat.1000399] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2008] [Accepted: 03/26/2009] [Indexed: 11/19/2022] Open
Abstract
Malaria is initiated when the mosquito introduces sporozoites into the skin of a mammalian host. To successfully continue the infection, sporozoites must invade blood vessels in the dermis and be transported to the liver. A significant number of sporozoites, however, may enter lymphatic vessels in the skin or remain in the skin long after the mosquito bite. We have used fluorescence microscopy of Plasmodium berghei sporozoites expressing a fluorescent protein to evaluate the kinetics of sporozoite disappearance from the skin. Sporozoites injected into immunized mice were rapidly immobilized, did not appear to invade dermal blood vessels and became morphologically degraded within several hours. Strikingly, mosquitoes introduced significantly fewer sporozoites into immunized than into non-immunized mice, presumably by formation of an immune complex between soluble sporozoite antigens in the mosquito saliva and homologous host antibodies at the proboscis tip. These results indicate that protective antibodies directed against sporozoites may function both by reducing the numbers of sporozoites injected into immunized hosts and by inhibiting the movement of injected sporozoites into dermal blood vessels. Malaria is initiated by a mosquito injecting malaria sporozoites into the skin. To successfully continue the infection, sporozoites must then invade blood vessels in skin for transportation to the liver. However, the majority of these injected sporozoites are unable to reach the blood. The numbers of sporozoites that successfully invade the blood may influence the characteristics of the subsequent clinical malaria infection. We studied this by microscopy with fluorescent sporozoites of the rodent malaria parasite Plasmodium berghei injected into mice by mosquitoes. Sporozoites introduced into mice that have been immunized against sporozoites become immobilized and cannot reach the blood; those that remain at the bite site become degraded within several hours. Strikingly, mosquitoes introduce significantly fewer sporozoites into skin of immunized mice. These findings indicate that antibodies directed against sporozoites seem to function both by reducing the numbers of sporozoites injected into immunized hosts in the first place and then by inhibiting the movement of the injected sporozoites into the bloodstream.
Collapse
|
49
|
Abstract
Plasmodium sporozoites are the product of a complex developmental process in the mosquito vector and are destined to infect the mammalian liver. Attention has been drawn to the mosquito stages and pre-erythrocytic stages owing to recognition that these are bottlenecks in the parasite life cycle and that intervention at these stages can block transmission and prevent infection. Parasite progression in the Anopheles mosquito, sporozoite transmission to the mammalian host by mosquito bite, and subsequent infection of the liver are characterized by extensive migration of invasive stages, cell invasion, and developmental changes. Preparation for the liver phase in the mammalian host begins in the mosquito with an extensive reprogramming of the sporozoite to support efficient infection and survival. Here, we discuss what is known about the molecular and cellular basis of the developmental progression of parasites and their interactions with host tissues in the mosquito and during the early phase of mammalian infection.
Collapse
Affiliation(s)
- Ahmed S I Aly
- Seattle Biomedical Research Institute, Seattle, Washington 98109, USA.
| | | | | |
Collapse
|
50
|
Sinnis P, Zavala F. The skin stage of malaria infection: biology and relevance to the malaria vaccine effort. Future Microbiol 2008; 3:275-8. [PMID: 18505393 DOI: 10.2217/17460913.3.3.275] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Plasmodium sporozoites, the infective stage of the malaria parasite, are injected into the mammalian host by mosquitoes and travel to the liver where they invade hepatocytes. Recent studies demonstrating that sporozoites are inoculated into the skin, remain there for hours before exiting and that 20% of the inoculum goes to the lymph node draining the inoculation site, suggest that there is a 'skin stage' to malaria infection that may set the stage for subsequent host responses to the parasite. Here, we present an overview of what is currently known about sporozoite-host interactions at the inoculation site and the draining lymph node, and discuss the impact of the skin stage of malaria on immunity to pre-erythrocytic stages and malaria vaccine design.
Collapse
Affiliation(s)
- Photini Sinnis
- Department of Medical Parasitology, New York University School of Medicine, 341 East 25th Street, New York, NY 10010, USA.
| | | |
Collapse
|