1
|
Yoo R, Jore MM, Julien J. Targeting Bottlenecks in Malaria Transmission: Antibody-Epitope Descriptions Guide the Design of Next-Generation Biomedical Interventions. Immunol Rev 2025; 330:e70001. [PMID: 39907429 PMCID: PMC11796336 DOI: 10.1111/imr.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 01/08/2025] [Indexed: 02/06/2025]
Abstract
Malaria continues to pose a significant burden to global health. Thus, a strong need exists for the development of a diverse panel of intervention strategies and modalities to combat malaria and achieve elimination and eradication goals. Deploying interventions that target bottlenecks in the transmission life cycle of the causative agent of malaria, Plasmodium parasites, is an attractive strategy. The development of highly potent antibody-based biologics, including vaccines, can be greatly facilitated by an in-depth molecular understanding of antibody-epitope interactions. Here, we provide an overview of structurally characterized antibodies targeting lead vaccine candidates expressed during the bottlenecks of the Plasmodium life cycle which include the pre-erythrocytic and sexual stages. The repeat region of the circumsporozoite protein (CSP), domain 1 of Pfs230 and domains 1 and 3 of Pfs48/45 are critical Plasmodium regions targeted by the most potent antibodies at the two bottlenecks of transmission, with other promising targets emerging and requiring further characterization.
Collapse
Affiliation(s)
- Randy Yoo
- Program in Molecular MedicineThe Hospital for Sick Children Research InstituteTorontoOntarioCanada
- Department of BiochemistryUniversity of TorontoTorontoOntarioCanada
| | - Matthijs M. Jore
- Department of Medical MicrobiologyRadboudumcNijmegenThe Netherlands
| | - Jean‐Philippe Julien
- Program in Molecular MedicineThe Hospital for Sick Children Research InstituteTorontoOntarioCanada
- Department of BiochemistryUniversity of TorontoTorontoOntarioCanada
- Department of ImmunologyUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
2
|
Dacon C, Moskovitz R, Swearingen K, Pereira LDS, Flores-Garcia Y, Aleshnick M, Kanatani S, Flynn B, Molina-Cruz A, Wollenberg K, Traver M, Kirtley P, Purser L, Dillon M, Bonilla B, Franco A, Petros S, Kritzberg J, Tucker C, Paez GG, Gupta P, Shears MJ, Pazzi J, Edgar JM, Teng AA, Belmonte A, Oda K, Doumbo S, Krymskaya L, Skinner J, Li S, Ghosal S, Kayentao K, Ongoiba A, Vaughan A, Campo JJ, Traore B, Barillas-Mury C, Wijayalath W, Idris A, Crompton PD, Sinnis P, Wilder BK, Zavala F, Seder RA, Wilson IA, Tan J. Protective antibodies target cryptic epitope unmasked by cleavage of malaria sporozoite protein. Science 2025; 387:eadr0510. [PMID: 39745947 PMCID: PMC11804177 DOI: 10.1126/science.adr0510] [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: 06/14/2024] [Accepted: 10/31/2024] [Indexed: 01/04/2025]
Abstract
The most advanced monoclonal antibodies (mAbs) and vaccines against malaria target the central repeat region or closely related sequences within the Plasmodium falciparum circumsporozoite protein (PfCSP). Here, using an antigen-agnostic strategy to investigate human antibody responses to whole sporozoites, we identified a class of mAbs that target a cryptic PfCSP epitope that is only exposed after cleavage and subsequent pyroglutamylation (pGlu) of the newly formed N terminus. This pGlu-CSP epitope is not targeted by current anti-PfCSP mAbs and is not included in the licensed malaria vaccines. MAD21-101, the most potent mAb in this class, confers sterile protection against Pf infection in a human liver-chimeric mouse model. These findings reveal a site of vulnerability on the sporozoite surface that can be targeted by next-generation antimalarial interventions.
Collapse
Affiliation(s)
- Cherrelle Dacon
- Antibody Biology Unit, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Re’em Moskovitz
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | - Lais Da Silva Pereira
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yevel Flores-Garcia
- Department of Molecular Microbiology and Immunology and Johns Hopkins Malaria Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Maya Aleshnick
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Portland, OR 97006, USA
| | - Sachie Kanatani
- Department of Molecular Microbiology and Immunology and Johns Hopkins Malaria Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Barbara Flynn
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alvaro Molina-Cruz
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Kurt Wollenberg
- Bioinformatics & Computational Biosciences Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Maria Traver
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Payton Kirtley
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Portland, OR 97006, USA
| | - Lauren Purser
- Antibody Biology Unit, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Marlon Dillon
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Brian Bonilla
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Adriano Franco
- Antibody Biology Unit, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Samantha Petros
- Antibody Biology Unit, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Jake Kritzberg
- Antibody Biology Unit, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Courtney Tucker
- Antibody Biology Unit, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
- Department of Biology, The Catholic University of America, Washington, DC 20064, USA
| | - Gonzalo Gonzalez Paez
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Priya Gupta
- Seattle Children’s Research Institute, Seattle, WA 98109, USA
| | - Melanie J. Shears
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Joseph Pazzi
- Antigen Discovery, Incorporated, Irvine, CA 92618, USA
| | | | - Andy A. Teng
- Antigen Discovery, Incorporated, Irvine, CA 92618, USA
| | - Arnel Belmonte
- General Dynamics Information Technology, Inc, Falls Church, 3150 Fairview Park Drive, Falls Church, VA 22042, USA
- Agile Vaccines and Therapeutics Department, Naval Medical Research Command, Silver Spring, Maryland, MD 20910, USA
| | - Kyosuke Oda
- Agile Vaccines and Therapeutics Department, Naval Medical Research Command, Silver Spring, Maryland, MD 20910, USA
- Henry M Jackson Foundation for the Advancement of Military Medicine, Inc, 6720A Rockledge Dr, Bethesda, MD 20817, USA
| | - Safiatou Doumbo
- Mali International Center of Excellence in Research, University of Sciences, Technique and Technology of Bamako, BP 1805, Point G, Bamako, Mali
| | - Ludmila Krymskaya
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Jeff Skinner
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Shanping Li
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Suman Ghosal
- Bioinformatics & Computational Biosciences Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kassoum Kayentao
- Mali International Center of Excellence in Research, University of Sciences, Technique and Technology of Bamako, BP 1805, Point G, Bamako, Mali
| | - Aissata Ongoiba
- Mali International Center of Excellence in Research, University of Sciences, Technique and Technology of Bamako, BP 1805, Point G, Bamako, Mali
| | - Ashley Vaughan
- Seattle Children’s Research Institute, Seattle, WA 98109, USA
| | | | - Boubacar Traore
- Mali International Center of Excellence in Research, University of Sciences, Technique and Technology of Bamako, BP 1805, Point G, Bamako, Mali
| | - Carolina Barillas-Mury
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Wathsala Wijayalath
- Agile Vaccines and Therapeutics Department, Naval Medical Research Command, Silver Spring, Maryland, MD 20910, USA
- Henry M Jackson Foundation for the Advancement of Military Medicine, Inc, 6720A Rockledge Dr, Bethesda, MD 20817, USA
| | - Azza Idris
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, 20139, 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 20852, USA
| | - Photini Sinnis
- Department of Molecular Microbiology and Immunology and Johns Hopkins Malaria Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Brandon K. Wilder
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Portland, OR 97006, USA
| | - Fidel Zavala
- Department of Molecular Microbiology and Immunology and Johns Hopkins Malaria Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Robert A. Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ian A. Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Joshua Tan
- Antibody Biology Unit, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| |
Collapse
|
3
|
Guizetti J. Imaging malaria parasites across scales and time. J Microsc 2025. [PMID: 39749880 DOI: 10.1111/jmi.13384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025]
Abstract
The idea that disease is caused at the cellular level is so fundamental to us that we might forget the critical role microscopy played in generating and developing this insight. Visually identifying diseased or infected cells lays the foundation for any effort to curb human pathology. Since the discovery of the Plasmodium-infected red blood cells, which cause malaria, microscopy has undergone an impressive development now literally resolving individual molecules. This review explores the expansive field of light microscopy, focusing on its application to malaria research. Imaging technologies have transformed our understanding of biological systems, yet navigating the complex and ever-growing landscape of techniques can be daunting. This review offers a guide for researchers, especially those working on malaria, by providing historical context as well as practical advice on selecting the right imaging approach. The review advocates an integrated methodology that prioritises the research question while considering key factors like sample preparation, fluorophore choice, imaging modality, and data analysis. In addition to presenting seminal studies and innovative applications of microscopy, the review highlights a broad range of topics, from traditional techniques like white light microscopy to advanced methods such as superresolution microscopy and time-lapse imaging. It addresses the emerging challenges of microscopy, including phototoxicity and trade-offs in resolution and speed, and offers insights into future technologies that might impact malaria research. This review offers a mix of historical perspective, technological progress, and practical guidance that appeal to novice and advanced microscopists alike. It aims to inspire malaria researchers to explore imaging techniques that could enrich their studies, thus advancing the field through enhanced visual exploration of the parasite across scales and time.
Collapse
Affiliation(s)
- Julien Guizetti
- Centre for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
4
|
Schmid M, Beyeler R, Caldelari R, Rehmann R, Heussler V, Roques M. Generation of a genetically double-attenuated Plasmodium berghei parasite that fully arrests growth during late liver stage development. PLoS One 2024; 19:e0316164. [PMID: 39739824 DOI: 10.1371/journal.pone.0316164] [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: 07/24/2024] [Accepted: 12/06/2024] [Indexed: 01/02/2025] Open
Abstract
Malaria caused by Plasmodium parasites remains a large health burden. One approach to combat this disease involves vaccinating individuals with whole sporozoites that have been genetically modified to arrest their development at a specific stage in the liver by targeted gene deletion, resulting in a genetically attenuated parasite (GAP). Through a comprehensive phenotyping screen, we identified the hscb gene, encoding a putative iron-sulfur protein assembly chaperone, as crucial for liver stage development, making it a suitable candidate gene for GAP generation. Parasites lacking Plasmodium berghei HscB (PbHscB) exhibited normal sporozoite production in mosquitoes, but their liver stage development was severely impaired, characterized by slow growth and delayed expression of merozoite surface protein 1 (MSP1). In vivo experiments demonstrated that PbHscB-deficient parasites exhibited a delay in prepatency of 2-4 days, emphasizing the significance of PbHscB for exo-erythrocytic development. Although knockout of PbHscB alone allowed breakthrough infections, it is a potent candidate for a dual gene deletion strategy. PlasMei2, an RNA-binding protein, was previously found to be crucial for the completion of liver stage development. We generated a PbHscB-PbMei2-double attenuated parasite line, serving as a late liver stage-arresting replication-competent (LARC) GAP, providing a solid block of liver-to-blood stage transition.
Collapse
Affiliation(s)
- Melanie Schmid
- Institute of Cell Biology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Raphael Beyeler
- Institute of Cell Biology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Reto Caldelari
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Ruth Rehmann
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Volker Heussler
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Magali Roques
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| |
Collapse
|
5
|
Carruthers VB. Apicomplexan Pore-Forming Toxins. Annu Rev Microbiol 2024; 78:277-291. [PMID: 39088861 DOI: 10.1146/annurev-micro-041222-025939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2024]
Abstract
Pore-forming toxins (PFTs) are released by one cell to directly inflict damage on another cell. Hosts use PFTs, including members of the membrane attack complex/perforin protein family, to fight infections and cancer, while bacteria and parasites deploy PFTs to promote infection. Apicomplexan parasites secrete perforin-like proteins as PFTs to egress from infected cells and traverse tissue barriers. Other protozoa, along with helminth parasites, utilize saposin-like PFTs prospectively for nutrient acquisition during infection. This review discusses seminal and more recent advances in understanding how parasite PFTs promote infection and describes how they are regulated and fulfill their roles without causing parasite self-harm. Although exciting progress has been made in defining mechanisms of pore formation by PFTs, many open questions remain to be addressed to gain additional key insights into these remarkable determinants of parasitic infections.
Collapse
Affiliation(s)
- Vern B Carruthers
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA;
| |
Collapse
|
6
|
Hildebrandt F, Iturritza MU, Zwicker C, Vanneste B, Van Hul N, Semle E, Quin J, Pascini T, Saarenpää S, He M, Andersson ER, Scott CL, Vega-Rodriguez J, Lundeberg J, Ankarklev J. Host-pathogen interactions in the Plasmodium-infected mouse liver at spatial and single-cell resolution. Nat Commun 2024; 15:7105. [PMID: 39160174 PMCID: PMC11333755 DOI: 10.1038/s41467-024-51418-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 08/06/2024] [Indexed: 08/21/2024] Open
Abstract
Upon infecting its vertebrate host, the malaria parasite initially invades the liver where it undergoes massive replication, whilst remaining clinically silent. The coordination of host responses across the complex liver tissue during malaria infection remains unexplored. Here, we perform spatial transcriptomics in combination with single-nuclei RNA sequencing over multiple time points to delineate host-pathogen interactions across Plasmodium berghei-infected liver tissues. Our data reveals significant changes in spatial gene expression in the malaria-infected tissues. These include changes related to lipid metabolism in the proximity to sites of Plasmodium infection, distinct inflammation programs between lobular zones, and regions with enrichment of different inflammatory cells, which we term 'inflammatory hotspots'. We also observe significant upregulation of genes involved in inflammation in the control liver tissues of mice injected with mosquito salivary gland components. However, this response is considerably delayed compared to that observed in P. berghei-infected mice. Our study establishes a benchmark for investigating transcriptome changes during host-parasite interactions in tissues, it provides informative insights regarding in vivo study design linked to infection and offers a useful tool for the discovery and validation of de novo intervention strategies aimed at malaria liver stage infection.
Collapse
Affiliation(s)
- Franziska Hildebrandt
- Molecular Biosciences, the Wenner Gren Institute, Stockholm University, Svante Arrhenius Väg 20C, SE-106 91, Stockholm, Sweden.
| | - Miren Urrutia Iturritza
- Molecular Biosciences, the Wenner Gren Institute, Stockholm University, Svante Arrhenius Väg 20C, SE-106 91, Stockholm, Sweden
| | - Christian Zwicker
- Department of Biomedical Molecular Biology, Faculty of Sciences, Ghent University, Ghent, Belgium
- Laboratory of Myeloid Cell Biology in Tissue Damage and Inflammation, VIB-UGent Center for Inflammation Research, Technologiepark-Zwijnaarde 71, Ghent, 9052, Belgium
| | - Bavo Vanneste
- Department of Biomedical Molecular Biology, Faculty of Sciences, Ghent University, Ghent, Belgium
- Laboratory of Myeloid Cell Biology in Tissue Damage and Inflammation, VIB-UGent Center for Inflammation Research, Technologiepark-Zwijnaarde 71, Ghent, 9052, Belgium
- Laboratory of Myeloid Cell Biology in Tissue Homeostasis and Regeneration, VIB-UGent Center for Inflammation Research, Technologiepark-Zwijnaarde 71, Ghent, 9052, Belgium
| | - Noémi Van Hul
- Department of Cell and Molecular Biology, Karolinska Institutet Stockholm, SE-171 77, Solna, Sweden
| | - Elisa Semle
- Molecular Biosciences, the Wenner Gren Institute, Stockholm University, Svante Arrhenius Väg 20C, SE-106 91, Stockholm, Sweden
| | - Jaclyn Quin
- Molecular Biosciences, the Wenner Gren Institute, Stockholm University, Svante Arrhenius Väg 20C, SE-106 91, Stockholm, Sweden
| | - Tales Pascini
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rm 2E20A, Rockville, MD, 20852, USA
| | - Sami Saarenpää
- SciLifeLab, Department of Gene Technology, KTH Royal Institute of Technology, Tomtebodavägen 23a, SE-171 65, Solna, Sweden
| | - Mengxiao He
- SciLifeLab, Department of Gene Technology, KTH Royal Institute of Technology, Tomtebodavägen 23a, SE-171 65, Solna, Sweden
| | - Emma R Andersson
- Department of Cell and Molecular Biology, Karolinska Institutet Stockholm, SE-171 77, Solna, Sweden
| | - Charlotte L Scott
- Department of Biomedical Molecular Biology, Faculty of Sciences, Ghent University, Ghent, Belgium
- Laboratory of Myeloid Cell Biology in Tissue Damage and Inflammation, VIB-UGent Center for Inflammation Research, Technologiepark-Zwijnaarde 71, Ghent, 9052, Belgium
| | - Joel Vega-Rodriguez
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rm 2E20A, Rockville, MD, 20852, USA
| | - Joakim Lundeberg
- SciLifeLab, Department of Gene Technology, KTH Royal Institute of Technology, Tomtebodavägen 23a, SE-171 65, Solna, Sweden
| | - Johan Ankarklev
- Molecular Biosciences, the Wenner Gren Institute, Stockholm University, Svante Arrhenius Väg 20C, SE-106 91, Stockholm, Sweden.
| |
Collapse
|
7
|
Dans MG, Boulet C, Watson GM, Nguyen W, Dziekan JM, Evelyn C, Reaksudsan K, Mehra S, Razook Z, Geoghegan ND, Mlodzianoski MJ, Goodman CD, Ling DB, Jonsdottir TK, Tong J, Famodimu MT, Kristan M, Pollard H, Stewart LB, Brandner-Garrod L, Sutherland CJ, Delves MJ, McFadden GI, Barry AE, Crabb BS, de Koning-Ward TF, Rogers KL, Cowman AF, Tham WH, Sleebs BE, Gilson PR. Aryl amino acetamides prevent Plasmodium falciparum ring development via targeting the lipid-transfer protein PfSTART1. Nat Commun 2024; 15:5219. [PMID: 38890312 PMCID: PMC11189555 DOI: 10.1038/s41467-024-49491-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 06/06/2024] [Indexed: 06/20/2024] Open
Abstract
With resistance to most antimalarials increasing, it is imperative that new drugs are developed. We previously identified an aryl acetamide compound, MMV006833 (M-833), that inhibited the ring-stage development of newly invaded merozoites. Here, we select parasites resistant to M-833 and identify mutations in the START lipid transfer protein (PF3D7_0104200, PfSTART1). Introducing PfSTART1 mutations into wildtype parasites reproduces resistance to M-833 as well as to more potent analogues. PfSTART1 binding to the analogues is validated using organic solvent-based Proteome Integral Solubility Alteration (Solvent PISA) assays. Imaging of invading merozoites shows the inhibitors prevent the development of ring-stage parasites potentially by inhibiting the expansion of the encasing parasitophorous vacuole membrane. The PfSTART1-targeting compounds also block transmission to mosquitoes and with multiple stages of the parasite's lifecycle being affected, PfSTART1 represents a drug target with a new mechanism of action.
Collapse
Affiliation(s)
- Madeline G Dans
- Burnet Institute, Melbourne, VIC, 3004, Australia.
- Walter and Eliza Hall Institute, Parkville, VIC, 3052, Australia.
- Institute of Mental and Physical Health and Clinical Translation (IMPACT) and School of Medicine, Deakin University, Geelong, VIC, 3220, Australia.
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Coralie Boulet
- Burnet Institute, Melbourne, VIC, 3004, Australia
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, 1206, Switzerland
| | - Gabrielle M Watson
- Walter and Eliza Hall Institute, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - William Nguyen
- Walter and Eliza Hall Institute, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Jerzy M Dziekan
- Walter and Eliza Hall Institute, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Cindy Evelyn
- Walter and Eliza Hall Institute, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Kitsanapong Reaksudsan
- Walter and Eliza Hall Institute, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Somya Mehra
- Burnet Institute, Melbourne, VIC, 3004, Australia
- Institute of Mental and Physical Health and Clinical Translation (IMPACT) and School of Medicine, Deakin University, Geelong, VIC, 3220, Australia
| | - Zahra Razook
- Burnet Institute, Melbourne, VIC, 3004, Australia
- Institute of Mental and Physical Health and Clinical Translation (IMPACT) and School of Medicine, Deakin University, Geelong, VIC, 3220, Australia
| | - Niall D Geoghegan
- Walter and Eliza Hall Institute, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Michael J Mlodzianoski
- Walter and Eliza Hall Institute, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | | | | | - Thorey K Jonsdottir
- Burnet Institute, Melbourne, VIC, 3004, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Department of Molecular Biology, Umeå University, Umeå, 901 87, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå, Sweden
| | - Joshua Tong
- Walter and Eliza Hall Institute, Parkville, VIC, 3052, Australia
| | - Mufuliat Toyin Famodimu
- Department of Infection Biology, Faculty of Infectious Diseases, London School of Hygiene and Tropical Medicine, WC1E 7HT, London, UK
| | - Mojca Kristan
- Wellcome Trust Human Malaria Transmission Facility, Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Harry Pollard
- Wellcome Trust Human Malaria Transmission Facility, Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Lindsay B Stewart
- Wellcome Trust Human Malaria Transmission Facility, Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Luke Brandner-Garrod
- Wellcome Trust Human Malaria Transmission Facility, Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Colin J Sutherland
- Department of Infection Biology, Faculty of Infectious Diseases, London School of Hygiene and Tropical Medicine, WC1E 7HT, London, UK
- Wellcome Trust Human Malaria Transmission Facility, Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Michael J Delves
- Department of Infection Biology, Faculty of Infectious Diseases, London School of Hygiene and Tropical Medicine, WC1E 7HT, London, UK
| | - Geoffrey I McFadden
- School of Biosciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Alyssa E Barry
- Burnet Institute, Melbourne, VIC, 3004, Australia
- Institute of Mental and Physical Health and Clinical Translation (IMPACT) and School of Medicine, Deakin University, Geelong, VIC, 3220, Australia
| | - Brendan S Crabb
- Burnet Institute, Melbourne, VIC, 3004, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Monash University, 3800, Melbourne, VIC, Australia
| | - Tania F de Koning-Ward
- Institute of Mental and Physical Health and Clinical Translation (IMPACT) and School of Medicine, Deakin University, Geelong, VIC, 3220, Australia
| | - Kelly L Rogers
- Walter and Eliza Hall Institute, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Alan F Cowman
- Walter and Eliza Hall Institute, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Wai-Hong Tham
- Walter and Eliza Hall Institute, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Brad E Sleebs
- Walter and Eliza Hall Institute, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Paul R Gilson
- Burnet Institute, Melbourne, VIC, 3004, Australia.
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
8
|
van Schuijlenburg R, Azargoshasb S, de Korne CM, Sijtsma JC, Bezemer S, van der Ham AJ, Baalbergen E, Geurten F, de Bes-Roeleveld LM, Chevalley-Maurel SC, van Oosterom MN, van Leeuwen FWB, Franke-Fayard B, Roestenberg M. Ageing of Plasmodium falciparum malaria sporozoites alters their motility, infectivity and reduces immune activation in vitro. Malar J 2024; 23:111. [PMID: 38641838 PMCID: PMC11027264 DOI: 10.1186/s12936-024-04946-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/12/2024] [Indexed: 04/21/2024] Open
Abstract
BACKGROUND Sporozoites (SPZ), the infective form of Plasmodium falciparum malaria, can be inoculated into the human host skin by Anopheline mosquitoes. These SPZ migrate at approximately 1 µm/s to find a blood vessel and travel to the liver where they infect hepatocytes and multiply. In the skin they are still low in number (50-100 SPZ) and vulnerable to immune attack by antibodies and skin macrophages. This is why whole SPZ and SPZ proteins are used as the basis for most malaria vaccines currently deployed and undergoing late clinical testing. Mosquitoes typically inoculate SPZ into a human host between 14 and 25 days after their previous infective blood meal. However, it is unknown whether residing time within the mosquito affects SPZ condition, infectivity or immunogenicity. This study aimed to unravel how the age of P. falciparum SPZ in salivary glands (14, 17, or 20 days post blood meal) affects their infectivity and the ensuing immune responses. METHODS SPZ numbers, viability by live/dead staining, motility using dedicated sporozoite motility orienting and organizing tool software (SMOOT), and infectivity of HC-04.j7 liver cells at 14, 17 and 20 days after mosquito feeding have been investigated. In vitro co-culture assays with SPZ stimulated monocyte-derived macrophages (MoMɸ) and CD8+ T-cells, analysed by flow cytometry, were used to investigate immune responses. RESULTS SPZ age did not result in different SPZ numbers or viability. However, a markedly different motility pattern, whereby motility decreased from 89% at day 14 to 80% at day 17 and 71% at day 20 was observed (p ≤ 0.0001). Similarly, infectivity of day 20 SPZ dropped to ~ 50% compared with day 14 SPZ (p = 0.004). MoMɸ were better able to take up day 14 SPZ than day 20 SPZ (from 7.6% to 4.1%, p = 0.03) and displayed an increased expression of pro-inflammatory CD80, IL-6 (p = 0.005), regulatory markers PDL1 (p = 0.02), IL-10 (p = 0.009) and cytokines upon phagocytosis of younger SPZ. Interestingly, co-culture of these cells with CD8+ T-cells revealed a decreased expression of activation marker CD137 and cytokine IFNγ compared to their day 20 counterparts. These findings suggest that older (day 17-20) P. falciparum SPZ are less infectious and have decreased immune regulatory potential. CONCLUSION Overall, this data is a first step in enhancing the understanding of how mosquito residing time affects P. falciparum SPZ and could impact the understanding of the P. falciparum infectious reservoir and the potency of whole SPZ vaccines.
Collapse
Affiliation(s)
- Roos van Schuijlenburg
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, Netherlands
| | - Samaneh Azargoshasb
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Clarize M de Korne
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Jeroen C Sijtsma
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, Netherlands
| | - Sascha Bezemer
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, Netherlands
| | - Alwin J van der Ham
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, Netherlands
| | - Els Baalbergen
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, Netherlands
| | - Fiona Geurten
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, Netherlands
| | - Laura M de Bes-Roeleveld
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, Netherlands
| | - Severine C Chevalley-Maurel
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, Netherlands
| | - Matthias N van Oosterom
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Fijs W B van Leeuwen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Blandine Franke-Fayard
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, Netherlands
| | - Meta Roestenberg
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, Netherlands.
| |
Collapse
|
9
|
Jennison C, Armstrong JM, Dankwa DA, Hertoghs N, Kumar S, Abatiyow BA, Naung M, Minkah NK, Swearingen KE, Moritz R, Barry AE, Kappe SHI, Vaughan AM. Plasmodium GPI-anchored micronemal antigen is essential for parasite transmission through the mosquito host. Mol Microbiol 2024; 121:394-412. [PMID: 37314965 PMCID: PMC11076100 DOI: 10.1111/mmi.15078] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/03/2023] [Accepted: 05/08/2023] [Indexed: 06/16/2023]
Abstract
Plasmodium parasites, the eukaryotic pathogens that cause malaria, feature three distinct invasive forms tailored to the host environment they must navigate and invade for life cycle progression. One conserved feature of these invasive forms is the micronemes, apically oriented secretory organelles involved in egress, motility, adhesion, and invasion. Here we investigate the role of GPI-anchored micronemal antigen (GAMA), which shows a micronemal localization in all zoite forms of the rodent-infecting species Plasmodium berghei. ∆GAMA parasites are severely defective for invasion of the mosquito midgut. Once formed, oocysts develop normally, however, sporozoites are unable to egress and exhibit defective motility. Epitope-tagging of GAMA revealed tight temporal expression late during sporogony and showed that GAMA is shed during sporozoite gliding motility in a similar manner to circumsporozoite protein. Complementation of P. berghei knockout parasites with full-length P. falciparum GAMA partially restored infectivity to mosquitoes, indicating conservation of function across Plasmodium species. A suite of parasites with GAMA expressed under the promoters of CTRP, CAP380, and TRAP, further confirmed the involvement of GAMA in midgut infection, motility, and vertebrate infection. These data show GAMA's involvement in sporozoite motility, egress, and invasion, implicating GAMA as a regulator of microneme function.
Collapse
Affiliation(s)
- Charlie Jennison
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Washington, Seattle, USA
| | - Janna M. Armstrong
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Washington, Seattle, USA
| | - Dorender A. Dankwa
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Washington, Seattle, USA
| | - Nina Hertoghs
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Washington, Seattle, USA
| | - Sudhir Kumar
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Washington, Seattle, USA
| | - Biley A. Abatiyow
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Washington, Seattle, USA
| | - Myo Naung
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Victoria, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Victoria, Carlton, Australia
- Department of Global Health, University of Washington, Washington, Seattle, USA
| | - Nana K. Minkah
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Washington, Seattle, USA
| | - Kristian E. Swearingen
- Institute of Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Victoria, Geelong, Australia
| | - Robert Moritz
- Institute of Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Deakin University, Victoria, Geelong, Australia
| | - Alyssa E. Barry
- Department of Global Health, University of Washington, Washington, Seattle, USA
- Institute for Systems Biology, Washington, Seattle, USA
| | - Stefan H. I. Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Washington, Seattle, USA
- Burnet Institute, Victoria, Melbourne, Australia
- Department of Pediatrics, University of Washington, Washington, Seattle, USA
| | - Ashley M. Vaughan
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Washington, Seattle, USA
- Burnet Institute, Victoria, Melbourne, Australia
- Department of Pediatrics, University of Washington, Washington, Seattle, USA
| |
Collapse
|
10
|
Atchou K, Berger BM, Heussler V, Ochsenreiter T. Pre-gelation staining expansion microscopy for visualisation of the Plasmodium liver stage. J Cell Sci 2023; 136:jcs261377. [PMID: 37942994 PMCID: PMC10729816 DOI: 10.1242/jcs.261377] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 11/01/2023] [Indexed: 11/10/2023] Open
Abstract
Fluorescence and light microscopy are important tools in the history of natural science. However, the resolution of microscopes is limited by the diffraction of light. One possible method to circumvent this physical restriction is the recently developed expansion microscopy (ExM). However, the original ultrastructure ExM (U-ExM) protocol is very time-consuming, and some epitopes are lost during the process. In this study, we developed a shortened pre-gelation staining ExM (PS-ExM) protocol and tested it to investigate the Plasmodium liver stage. The protocol presented in this study allows expanding of pre-stained samples, which results in shorter incubation times, better preservation of some epitopes and the advantage that non-expanded controls can be performed alongside using the same staining protocol. The protocol applicability was accessed throughout the Plasmodium liver stage, showing isotropic five-fold expansion. Furthermore, we used PS-ExM to visualise parasite mitochondria as well as the association of lysosomes to the parasitophorous vacuole membrane (PVM) as an example of visualising host-pathogen interaction. We are convinced that this new tool will be helpful for a deeper understanding of the biology of the Plasmodium liver stage.
Collapse
Affiliation(s)
- Kodzo Atchou
- Institute of Cell Biology, University of Bern, 3012 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Bianca Manuela Berger
- Institute of Cell Biology, University of Bern, 3012 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Volker Heussler
- Institute of Cell Biology, University of Bern, 3012 Bern, Switzerland
| | | |
Collapse
|
11
|
Nicholas J, Kolli SK, Subramani PA, De SL, Ogbondah MM, Barnes SJ, Ntumngia FB, Adams JH. Comparative analyses of functional antibody-mediated inhibition with anti-circumsporozoite monoclonal antibodies against transgenic Plasmodium berghei. Malar J 2023; 22:335. [PMID: 37936181 PMCID: PMC10629016 DOI: 10.1186/s12936-023-04765-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/24/2023] [Indexed: 11/09/2023] Open
Abstract
BACKGROUND Acquired functional inhibitory antibodies are one of several humoral immune mechanisms used to neutralize foreign pathogens. In vitro bioassays are useful tools for quantifying antibody-mediated inhibition and evaluating anti-parasite immune antibodies. However, a gap remains in understanding of how antibody-mediated inhibition in vitro translates to inhibition in vivo. In this study, two well-characterized transgenic Plasmodium berghei parasite lines, PbmCh-luc and Pb-PfCSP(r), and murine monoclonal antibodies (mAbs) specific to P. berghei and Plasmodium falciparum circumsporozoite protein (CSP), 3D11 and 2A10, respectively, were used to evaluate antibody-mediated inhibition of parasite development in both in vitro and in vivo functional assays. METHODS IC50 values of mAbs were determined using an established inhibition of liver-stage development assay (ILSDA). For the in vivo inhibition assay, mice were passively immunized by transfer of the mAbs and subsequently challenged with 5.0 × 103 sporozoites via tail vein injection. The infection burden in both assays was quantified by luminescence and qRT-PCR of P. berghei 18S rRNA normalized to host GAPDH. RESULTS The IC50 values quantified by relative luminescence of mAbs 3D11 and 2A10 were 0.396 µg/ml and 0.093 µg/ml, respectively, against transgenic lines in vitro. Using the highest (> 90%) inhibitory antibody concentrations in a passive transfer, an IC50 of 233.8 µg/ml and 181.5 µg/ml for mAbs 3D11 and 2A10, respectively, was observed in vivo. At 25 µg (250 µg/ml), the 2A10 antibody significantly inhibited liver burden in mice compared to control. Additionally, qRT-PCR of P. berghei 18S rRNA served as a secondary validation of liver burden quantification. CONCLUSIONS Results from both experimental models, ILSDA and in vivo challenge, demonstrated that increased concentrations of the homologous anti-CSP repeat mAbs increased parasite inhibition. However, differences in antibody IC50 values between parasite lines did not allow a direct correlation between the inhibition of sporozoite invasion in vitro by ILSDA and the inhibition of mouse liver stage burden. Further studies are needed to establish the conditions for confident predictions for the in vitro ILSDA to be a predictor of in vivo outcomes using this model system.
Collapse
Affiliation(s)
- Justin Nicholas
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Surendra Kumar Kolli
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - Pradeep Annamalai Subramani
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - Sai Lata De
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
- Department of Infectious Disease & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Madison M Ogbondah
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - Samantha J Barnes
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - Francis Babila Ntumngia
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - John H Adams
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA.
| |
Collapse
|
12
|
Rajneesh, Tiwari R, Singh VK, Kumar A, Gupta RP, Singh AK, Gautam V, Kumar R. Advancements and Challenges in Developing Malaria Vaccines: Targeting Multiple Stages of the Parasite Life Cycle. ACS Infect Dis 2023; 9:1795-1814. [PMID: 37708228 DOI: 10.1021/acsinfecdis.3c00332] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
Malaria, caused by Plasmodium species, remains a major global health concern, causing millions of deaths annually. While the introduction of the RTS,S vaccine has shown promise, there is a pressing need for more effective vaccines due to the emergence of drug-resistant parasites and insecticide-resistant vectors. However, the complex life cycle and genetic diversity of the parasite, technical obstacles, limited funding, and the impact of the 2019 pandemic have hindered progress in malaria vaccine development. This review focuses on advancements in malaria vaccine development, particularly the ongoing clinical trials targeting antigens from different stages of the Plasmodium life cycle. Additionally, we discuss the rationale, strategies, and challenges associated with vaccine design, aiming to enhance the immune response and protective efficacy of vaccine candidates. A cost-effective and multistage vaccine could hold the key to controlling and eradicating malaria.
Collapse
Affiliation(s)
- Rajneesh
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Rahul Tiwari
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Vishal K Singh
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Awnish Kumar
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Rohit P Gupta
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
- Department of Applied Microbiology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Akhilesh K Singh
- Faculty of Dental Science, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Vibhav Gautam
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Rajiv Kumar
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| |
Collapse
|
13
|
D'Ambrosio HK, Keeler AM, Derbyshire ER. Examination of Secondary Metabolite Biosynthesis in Apicomplexa. Chembiochem 2023; 24:e202300263. [PMID: 37171468 DOI: 10.1002/cbic.202300263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 05/13/2023]
Abstract
Natural product discovery has traditionally relied on the isolation of small molecules from producing species, but genome-sequencing technology and advances in molecular biology techniques have expanded efforts to a wider array of organisms. Protists represent an underexplored kingdom for specialized metabolite searches despite bioinformatic analysis that suggests they harbor distinct biologically active small molecules. Specifically, pathogenic apicomplexan parasites, responsible for billions of global infections, have been found to possess multiple biosynthetic gene clusters, which hints at their capacity to produce polyketide metabolites. Biochemical studies have revealed unique features of apicomplexan polyketide synthases, but to date, the identity and function of the polyketides synthesized by these megaenzymes remains unknown. Herein, we discuss the potential for specialized metabolite production in protists and the possible evolution of polyketide biosynthetic gene clusters in apicomplexan parasites. We then focus on a polyketide synthase from the apicomplexan Toxoplasma gondii to discuss the unique domain architecture and properties of these proteins when compared to previously characterized systems, and further speculate on the possible functions for polyketides in these pathogenic parasites.
Collapse
Affiliation(s)
- Hannah K D'Ambrosio
- Department of Chemistry, Duke University, 124 Science Drive, Durham, NC 27708, USA
| | - Aaron M Keeler
- Department of Chemistry, Duke University, 124 Science Drive, Durham, NC 27708, USA
| | - Emily R Derbyshire
- Department of Chemistry, Duke University, 124 Science Drive, Durham, NC 27708, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, 213 Research Drive, Durham, NC 27710, USA
| |
Collapse
|
14
|
Quaye IK, Aleksenko L, Paganotti GM, Peloewetse E, Haiyambo DH, Ntebela D, Oeuvray C, Greco B. Malaria Elimination in Africa: Rethinking Strategies for Plasmodium vivax and Lessons from Botswana. Trop Med Infect Dis 2023; 8:392. [PMID: 37624330 PMCID: PMC10458071 DOI: 10.3390/tropicalmed8080392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/21/2023] [Accepted: 07/22/2023] [Indexed: 08/26/2023] Open
Abstract
The global malaria community has picked up the theme of malaria elimination in more than 90% of the world's population in the next decade. Recent reports of Plasmodium vivax (P. vivax) in sub-Saharan Africa, including in Duffy-negative individuals, threaten the efforts aimed at achieving elimination. This is not only in view of strategies that are tailored only to P. falciparum elimination but also due to currently revealed biological characteristics of P. vivax concerning the relapse patterns of hypnozoites and conservation of large biomasses in cryptic sites in the bone marrow and spleen. A typical scenario was observed in Botswana between 2008 and 2018, which palpably projects how P. vivax could endanger malaria elimination efforts where the two parasites co-exist. The need for the global malaria community, national malaria programs (NMPs), funding agencies and relevant stakeholders to engage in a forum to discuss and recommend clear pathways for elimination of malaria, including P. vivax, in sub-Saharan Africa is warranted.
Collapse
Affiliation(s)
- Isaac K. Quaye
- Pan African Vivax and Ovale Network, Faculty of Engineering Computer and Allied Sciences, Regent University College of Science and Technology, #1 Regent Ave, McCarthy Hill, Mendskrom, Dansoman, Accra P.O. Box DS1636, Ghana
| | - Larysa Aleksenko
- Department of Health Sciences, School of Public Health, College of Health, Medicine and Life Sciences, Brunel University, Kingston Lane, Uxbridge, Middlesex, London UB8 3PH, UK;
| | - Giacomo M. Paganotti
- Botswana-University of Pennsylvania Partnership, Riverwalk, Gaborone P.O. Box 45498, Botswana;
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elias Peloewetse
- Department of Biological Sciences, Faculty of Sciences, University of Botswana, Gaborone Private Bag 00704, Botswana;
| | - Daniel H. Haiyambo
- Department of Human, Biological and Translational Medical Sciences, Faculty of Health Sciences and Veterinary Medicine, University of Namibia School of Medicine, Hage Geingob Campus, Windhoek Private Bag 13301, Namibia;
| | - Davies Ntebela
- National Malaria Program, Ministry of Health, Gaborone Private Bag 0038, Botswana;
| | - Claude Oeuvray
- Global Health Institute of Merck, Terre Bonne Building Z0, Route de Crassier 1, Eysin, 1266 Geneva, Switzerland; (C.O.); (B.G.)
| | - Beatrice Greco
- Global Health Institute of Merck, Terre Bonne Building Z0, Route de Crassier 1, Eysin, 1266 Geneva, Switzerland; (C.O.); (B.G.)
| | - the PAVON Consortium
- PAVON, Regent University College of Science and Technology, #1 Regent Avenue, McCarthy Hiil, Mendskrom, Dansoman, Accra P.O. Box DS1636, Ghana
| |
Collapse
|
15
|
Ahmed MA, Baruah P, Saif A, Han JH, Al-Zharani M, Wazid SW, Alkahtani S, Patgiri SJ, Al-Eissa MS, Quan FS. In Silico Analysis Reveals High Levels of Genetic Diversity of Plasmodium knowlesi Cell Traversal Protein for Ookinetes and Sporozoites ( PkCelTOS) in Clinical Samples. Trop Med Infect Dis 2023; 8:380. [PMID: 37624318 PMCID: PMC10458480 DOI: 10.3390/tropicalmed8080380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 07/05/2023] [Accepted: 07/20/2023] [Indexed: 08/26/2023] Open
Abstract
The cell-traversal protein for ookinetes and sporozoites (CelTOS), expressed on the surface of ookinetes and sporozoitesin Plasmodium species, is a promising malaria vaccine candidate. CelTOS is essential for parasite invasion into mosquito midgut and human hepatocytes, thereby contributing to malaria transmission and disease pathogenesis. This study explores the genetic diversity, polymorphisms, haplotypes, natural selection, phylogenetic analysis, and epitope prediction in the full-length Plasmodium knowlesi CelTOS gene in clinical samples from Sarawak, Malaysian Borneo, and long-term laboratory strains from Peninsular Malaysia and the Philippines. Our analysis revealed a high level of genetic variation in the PkCelTOS gene, with a nucleotide diversity of π ~ 0.021, which was skewed towards the 3' end of the gene. This level of diversity is double that observed in PfCelTOS and 20 times that observed in PvCelTOS from worldwide clinical samples. Tests of natural selection revealed evidence for positive selection within clinical samples. Phylogenetic analysis of the amino acid sequence of PkCelTOS revealed the presence of two distinct groups, although no geographical clustering was observed. Epitope prediction analysis identified two potential epitopes (96AQLKATA102 and 124TIKPPRIKED133) using the IEDB server and one epitope (125IKPPRIKED133) by Bcepred server on the C' terminal region of PkCelTOS protein. Both the servers predicted a common epitope region of nine amino acid length (IKPPRIKED) peptide, which can be studied in the future as a potential candidate for vaccine development. These findings shed light on the genetic diversity, polymorphism, haplotypes, and natural selection within PkCelTOS in clinical samples and provide insights about its future prospects as a potential candidate for P. knowlesi malaria vaccine development.
Collapse
Affiliation(s)
- Md Atique Ahmed
- ICMR-Regional Medical Research Centre, NE Region, Dibrugarh 786010, Assam, India (S.J.P.)
| | - Pratisthita Baruah
- ICMR-Regional Medical Research Centre, NE Region, Dibrugarh 786010, Assam, India (S.J.P.)
| | - Ahmed Saif
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, King Khalid University, Abha 61321, Saudi Arabia
| | - Jin-Hee Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Mohammed Al-Zharani
- Biology Department, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSU), Riyadh 11623, Saudi Arabia (M.S.A.-E.)
| | - Syeda Wasfeea Wazid
- Arogyo Society of Health, Welfare and Support (ASHWAS), Guwahati 785640, Assam, India;
| | - Saad Alkahtani
- Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Saurav J. Patgiri
- ICMR-Regional Medical Research Centre, NE Region, Dibrugarh 786010, Assam, India (S.J.P.)
| | - Mohammed S. Al-Eissa
- Biology Department, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSU), Riyadh 11623, Saudi Arabia (M.S.A.-E.)
| | - Fu-Shi Quan
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Core Research Institute (CRI), Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Medical Zoology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
16
|
Bhattacharjee S, Ghosh D, Saha R, Sarkar R, Kumar S, Khokhar M, Pandey RK. Mechanism of Immune Evasion in Mosquito-Borne Diseases. Pathogens 2023; 12:635. [PMID: 37242305 PMCID: PMC10222277 DOI: 10.3390/pathogens12050635] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
In recent decades, mosquito-borne illnesses have emerged as a major health burden in many tropical regions. These diseases, such as malaria, dengue fever, chikungunya, yellow fever, Zika virus infection, Rift Valley fever, Japanese encephalitis, and West Nile virus infection, are transmitted through the bite of infected mosquitoes. These pathogens have been shown to interfere with the host's immune system through adaptive and innate immune mechanisms, as well as the human circulatory system. Crucial immune checkpoints such as antigen presentation, T cell activation, differentiation, and proinflammatory response play a vital role in the host cell's response to pathogenic infection. Furthermore, these immune evasions have the potential to stimulate the human immune system, resulting in other associated non-communicable diseases. This review aims to advance our understanding of mosquito-borne diseases and the immune evasion mechanisms by associated pathogens. Moreover, it highlights the adverse outcomes of mosquito-borne disease.
Collapse
Affiliation(s)
| | - Debanjan Ghosh
- Department of Biotechnology, Pondicherry University, Puducherry 605014, India
| | - Rounak Saha
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry 605014, India
| | - Rima Sarkar
- DBT Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
| | - Saurav Kumar
- DBT Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
| | - Manoj Khokhar
- Department of Biochemistry, AIIMS, Jodhpur 342005, India
| | - Rajan Kumar Pandey
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 77 Solna, Sweden
| |
Collapse
|
17
|
Santos BMD, Przyborski JM, Garcia CRS. Changes in K + Concentration as a Signaling Mechanism in the Apicomplexa Parasites Plasmodium and Toxoplasma. Int J Mol Sci 2023; 24:ijms24087276. [PMID: 37108438 PMCID: PMC10138558 DOI: 10.3390/ijms24087276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/04/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
During their life cycle, apicomplexan parasites pass through different microenvironments and encounter a range of ion concentrations. The discovery that the GPCR-like SR25 in Plasmodium falciparum is activated by a shift in potassium concentration indicates that the parasite can take advantage of its development by sensing different ionic concentrations in the external milieu. This pathway involves the activation of phospholipase C and an increase in cytosolic calcium. In the present report, we summarize the information available in the literature regarding the role of potassium ions during parasite development. A deeper understanding of the mechanisms that allow the parasite to cope with ionic potassium changes contributes to our knowledge about the cell cycle of Plasmodium spp.
Collapse
Affiliation(s)
- Benedito M Dos Santos
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Jude M Przyborski
- Department of Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus-Liebig University, 35390 Gießen, Germany
| | - Célia R S Garcia
- Department of Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus-Liebig University, 35390 Gießen, Germany
| |
Collapse
|
18
|
Plasmodium 6-cysteine proteins determine the commitment of sporozoites to liver-infection. Parasitol Int 2023; 93:102700. [PMID: 36403748 DOI: 10.1016/j.parint.2022.102700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/11/2022] [Accepted: 11/13/2022] [Indexed: 11/19/2022]
Abstract
Plasmodium sporozoites travel a long way from the site where they are released by a mosquito bite to the liver, where they infect hepatocytes and develop into erythrocyte-invasive forms. The success of this infection depends on the ability of the sporozoites to correctly recognize the hepatocyte as a target and change their behavior from migration to infection. However, how this change is accomplished remains incompletely understood. In this paper, we report that 6-cysteine protein family members expressed in sporozoites including B9 are responsible for this ability. Experiments on parasites using double knockouts of B9 and SPECT2, which is essential for sporozoite to migrate through the hepatocyte, showed that the parasites lacked the capacity to stop migration. This finding suggests that interactions between these parasite proteins and hepatocyte-specific cell surface ligands mediate correct recognition of hepatocytes by sporozoites, which is an essential step in malaria transmission to humans.
Collapse
|
19
|
Abstract
INTRODUCTION : Eradication of malaria remains one of the main aims of medicine. Despite progress in malaria treatment, mortality rate remains high, especially in the poorest parts of the world. Therefore, prevention through vaccines is fundamental and recent approval of the first effective vaccine reinforced this assumption. However, since the parasite cycle is complex, being composed of three stages, different types of vaccine targeting stage-specific antigens shall be developed. Moreover, the beneficial effect on vaccinated subjects can be tuned using compositions targeting different disease stages. AREA COVERED : We analysed the malaria vaccine patent landscape describing the most significant patents published after 2016, classified according to the different parasite stages targeted focusing on selected protein antigens or epitopes. We searched "malaria vaccine" on Patentscope and Espacenet. EXPERT OPINION : Pre-erythrocytic vaccines were boosted by RTS,S approval, but its partial efficacy, limited to sporozoites, calls for compositions active against other disease stages. In particular, multi-antigens vaccines could be more effective than single-stage ones, as they would activate an immune response more similar to that acquired in endemic regions. Furthermore, vaccine storage is another factor to be taken into account given the climate of the areas where malaria is widespread. More advanced technologies can lead to more effective and safer vaccines.
Collapse
Affiliation(s)
- Michael Quagliata
- Interdepartmental Research Unit of Peptide and Protein Chemistry and Biology, Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 13, I-50019 Sesto Fiorentino, Italy
| | - Anna Maria Papini
- Interdepartmental Research Unit of Peptide and Protein Chemistry and Biology, Department of Chemistry "Ugo Schiff", University of Florence, Via della Lastruccia 13, I-50019 Sesto Fiorentino, Italy
| | - Paolo Rovero
- Interdepartmental Research Unit of Peptide and Protein Chemistry and Biology, Department of NeuroFarBa, University of Florence, Via Ugo Schiff 6, I-50019 Sesto Fiorentino, Italy
| |
Collapse
|
20
|
Romano PS, Akematsu T, Besteiro S, Bindschedler A, Carruthers VB, Chahine Z, Coppens I, Descoteaux A, Alberto Duque TL, He CY, Heussler V, Le Roch KG, Li FJ, de Menezes JPB, Menna-Barreto RFS, Mottram JC, Schmuckli-Maurer J, Turk B, Tavares Veras PS, Salassa BN, Vanrell MC. Autophagy in protists and their hosts: When, how and why? AUTOPHAGY REPORTS 2023; 2:2149211. [PMID: 37064813 PMCID: PMC10104450 DOI: 10.1080/27694127.2022.2149211] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 11/15/2022] [Indexed: 03/12/2023]
Abstract
Pathogenic protists are a group of organisms responsible for causing a variety of human diseases including malaria, sleeping sickness, Chagas disease, leishmaniasis, and toxoplasmosis, among others. These diseases, which affect more than one billion people globally, mainly the poorest populations, are characterized by severe chronic stages and the lack of effective antiparasitic treatment. Parasitic protists display complex life-cycles and go through different cellular transformations in order to adapt to the different hosts they live in. Autophagy, a highly conserved cellular degradation process, has emerged as a key mechanism required for these differentiation processes, as well as other functions that are crucial to parasite fitness. In contrast to yeasts and mammals, protist autophagy is characterized by a modest number of conserved autophagy-related proteins (ATGs) that, even though, can drive the autophagosome formation and degradation. In addition, during their intracellular cycle, the interaction of these pathogens with the host autophagy system plays a crucial role resulting in a beneficial or harmful effect that is important for the outcome of the infection. In this review, we summarize the current state of knowledge on autophagy and other related mechanisms in pathogenic protists and their hosts. We sought to emphasize when, how, and why this process takes place, and the effects it may have on the parasitic cycle. A better understanding of the significance of autophagy for the protist life-cycle will potentially be helpful to design novel anti-parasitic strategies.
Collapse
Affiliation(s)
- Patricia Silvia Romano
- Laboratorio de Biología de Trypanosoma cruzi y de la célula hospedadora. Instituto de Histología y Embriología de Mendoza. Universidad Nacional de Cuyo. (IHEM-CONICET-UNCUYO). Facultad de Ciencias Médicas. Universidad Nacional de Cuyo. Av. Libertador 80 (5500), Mendoza, Argentina
| | - Takahiko Akematsu
- Department of Biosciences, College of Humanities and Sciences, Nihon University, Tokyo, Japan
| | | | | | - Vern B Carruthers
- Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Zeinab Chahine
- Department of Molecular, Cell and Systems Biology, University of California Riverside, CA, USA
| | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology. Department of Molecular Microbiology and Immunology. Johns Hopkins Malaria Research Institute. Johns Hopkins University Bloomberg School of Public Health. Baltimore 21205, MD, USA
| | - Albert Descoteaux
- Centre Armand-Frappier Santé Biotechnologie, Institut national de la recherche scientifique, Laval, QC
| | - Thabata Lopes Alberto Duque
- Autophagy Inflammation and Metabolism Center, University of New Mexico Health Sciences Center, Albuquerque, NM, USA; Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Cynthia Y He
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Volker Heussler
- Institute of Cell Biology.University of Bern. Baltzerstr. 4 3012 Bern
| | - Karine G Le Roch
- Department of Molecular, Cell and Systems Biology, University of California Riverside, CA, USA
| | - Feng-Jun Li
- Department of Biological Sciences, National University of Singapore, Singapore
| | | | | | - Jeremy C Mottram
- York Biomedical Research Institute, Department of Biology, University of York, York, UK
| | | | - Boris Turk
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, SI-1000 Ljubljana, Slovenia
| | - Patricia Sampaio Tavares Veras
- Laboratory of Host-Parasite Interaction and Epidemiology, Gonçalo Moniz Institute, Fiocruz-Bahia
- National Institute of Science and Technology of Tropical Diseases - National Council for Scientific Research and Development (CNPq)
| | - Betiana Nebai Salassa
- Laboratorio de Biología de Trypanosoma cruzi y de la célula hospedadora. Instituto de Histología y Embriología de Mendoza. Universidad Nacional de Cuyo. (IHEM-CONICET-UNCUYO). Facultad de Ciencias Médicas. Universidad Nacional de Cuyo. Av. Libertador 80 (5500), Mendoza, Argentina
| | - María Cristina Vanrell
- Laboratorio de Biología de Trypanosoma cruzi y de la célula hospedadora. Instituto de Histología y Embriología de Mendoza. Universidad Nacional de Cuyo. (IHEM-CONICET-UNCUYO). Facultad de Ciencias Médicas. Universidad Nacional de Cuyo. Av. Libertador 80 (5500), Mendoza, Argentina
| |
Collapse
|
21
|
Loubens M, Marinach C, Paquereau CE, Hamada S, Hoareau-Coudert B, Akbar D, Franetich JF, Silvie O. The claudin-like apicomplexan microneme protein is required for gliding motility and infectivity of Plasmodium sporozoites. PLoS Pathog 2023; 19:e1011261. [PMID: 36928686 PMCID: PMC10047546 DOI: 10.1371/journal.ppat.1011261] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 03/28/2023] [Accepted: 03/02/2023] [Indexed: 03/18/2023] Open
Abstract
Invasion of host cells by apicomplexan parasites such as Toxoplasma and Plasmodium spp requires the sequential secretion of the parasite apical organelles, the micronemes and the rhoptries. The claudin-like apicomplexan microneme protein (CLAMP) is a conserved protein that plays an essential role during invasion by Toxoplasma gondii tachyzoites and in Plasmodium falciparum asexual blood stages. CLAMP is also expressed in Plasmodium sporozoites, the mosquito-transmitted forms of the malaria parasite, but its role in this stage is still unknown. CLAMP is essential for Plasmodium blood stage growth and is refractory to conventional gene deletion. To circumvent this obstacle and study the function of CLAMP in sporozoites, we used a conditional genome editing strategy based on the dimerisable Cre recombinase in the rodent malaria model parasite P. berghei. We successfully deleted clamp gene in P. berghei transmission stages and analyzed the functional consequences on sporozoite infectivity. In mosquitoes, sporozoite development and egress from oocysts was not affected in conditional mutants. However, invasion of the mosquito salivary glands was dramatically reduced upon deletion of clamp gene. In addition, CLAMP-deficient sporozoites were impaired in cell traversal and productive invasion of mammalian hepatocytes. This severe phenotype was associated with major defects in gliding motility and with reduced shedding of the sporozoite adhesin TRAP. Expansion microscopy revealed partial colocalization of CLAMP and TRAP in a subset of micronemes, and a distinct accumulation of CLAMP at the apical tip of sporozoites. Collectively, these results demonstrate that CLAMP is essential across invasive stages of the malaria parasite, and support a role of the protein upstream of host cell invasion, possibly by regulating the secretion or function of adhesins in Plasmodium sporozoites.
Collapse
Affiliation(s)
- Manon Loubens
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Carine Marinach
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Clara-Eva Paquereau
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Soumia Hamada
- Sorbonne Université, INSERM, UMS PASS, Plateforme Post-génomique de la Pitié Salpêtrière (P3S), Paris, France
| | - Bénédicte Hoareau-Coudert
- Sorbonne Université, INSERM, UMS PASS, Plateforme de cytométrie de la Pitié-Salpêtrière (CyPS), Paris, France
| | - David Akbar
- Sorbonne Université, INSERM, CNRS, Hôpital de la Pitié Salpêtrière, Paris Brain Institute, ICM Quant Cell imaging Core Facility, Paris, France
| | - Jean-François Franetich
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Olivier Silvie
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| |
Collapse
|
22
|
Schepis A, Kumar S, Kappe SHI. Malaria parasites harness Rho GTPase signaling and host cell membrane ruffling for productive invasion of hepatocytes. Cell Rep 2023; 42:111927. [PMID: 36640315 DOI: 10.1016/j.celrep.2022.111927] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 09/06/2022] [Accepted: 12/14/2022] [Indexed: 12/31/2022] Open
Abstract
Plasmodium sporozoites are the motile forms of the malaria parasites that infect hepatocytes. The initial invasion of hepatocytes is thought to be actively driven by sporozoites, but host cell processes might also play a role. Sporozoite invasion triggers a host plasma membrane invagination that forms a vacuole around the intracellular parasite, which is critical for subsequent intracellular parasite replication. Using fast live confocal microscopy, we observed that the initial interactions between sporozoites and hepatocytes induce plasma membrane ruffles and filopodia extensions. Importantly, we find that these host cell processes facilitate invasion and that Rho GTPase signaling, which regulates membrane ruffling and filopodia extension, is critical for productive infection. Interestingly, sporozoite cell traversal stimulates these processes, suggesting that it increases hepatocyte susceptibility to productive infection. Our study identifies host cell signaling events involved in plasma membrane dynamics as a critical host component of successful malaria parasite infection of hepatocytes.
Collapse
Affiliation(s)
- Antonino Schepis
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Seattle, WA 98109, USA
| | - Sudhir Kumar
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Seattle, WA 98109, USA
| | - Stefan H I Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, 307 Westlake Avenue North, Seattle, WA 98109, USA; Department of Pediatrics, University of Washington, Seattle, WA 98105, USA; Department of Global Health, University of Washington, Seattle, WA 98105, USA.
| |
Collapse
|
23
|
Hirako IC, Antunes MM, Rezende RM, Hojo-Souza NS, Figueiredo MM, Dias T, Nakaya H, Menezes GB, Gazzinelli RT. Uptake of Plasmodium chabaudi hemozoin drives Kupffer cell death and fuels superinfections. Sci Rep 2022; 12:19805. [PMID: 36396745 PMCID: PMC9671901 DOI: 10.1038/s41598-022-23858-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 11/07/2022] [Indexed: 11/18/2022] Open
Abstract
Kupffer cells (KCs) are self-maintained tissue-resident macrophages that line liver sinusoids and play an important role on host defense. It has been demonstrated that upon infection or intense liver inflammation, KCs might be severely depleted and replaced by immature monocytic cells; however, the mechanisms of cell death and the alterations on liver immunity against infections deserves further investigation. We explored the impact of acute Plasmodium infection on KC biology and on the hepatic immune response against secondary infections. Similar to patients, infection with Plasmodium chabaudi induced acute liver damage as determined by serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) elevation. This was associated with accumulation of hemozoin, increased of proinflammatory response and impaired bacterial and viral clearance, which led to pathogen spread to other organs. In line with this, mice infected with Plasmodium had enhanced mortality during secondary infections, which was associated with increased production of mitochondrial superoxide, lipid peroxidation and increased free iron within KCs-hallmarks of cell death by ferroptosis. Therefore, we revealed that accumulation of iron with KCs, triggered by uptake of circulating hemozoin, is a novel mechanism of macrophage depletion and liver inflammation during malaria, providing novel insights on host susceptibility to secondary infections. Malaria can cause severe liver damage, along with depletion of liver macrophages, which can predispose individuals to secondary infections and enhance the chances of death.
Collapse
Affiliation(s)
- Isabella C Hirako
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, MG, Brazil
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, 364 Plantation Street, Lazare Research Building, 3rd Floor, Worcester, MA, USA
| | - Maísa Mota Antunes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rafael Machado Rezende
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | - Thomaz Dias
- Escola de Ciências Farmacêuticas - Universidade de São Paulo, São Paulo, SP, Brazil
| | - Helder Nakaya
- Escola de Ciências Farmacêuticas - Universidade de São Paulo, São Paulo, SP, Brazil
| | - Gustavo Batista Menezes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ricardo Tostes Gazzinelli
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, MG, Brazil.
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, 364 Plantation Street, Lazare Research Building, 3rd Floor, Worcester, MA, USA.
- Departamento de Bioquímica E Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
24
|
Kumari J, Sah RK, Mohaideen. S NM, Ahmad S, Pati S, Singh S. Studying the Rationale of Fire Ant Sting Therapy Usage by the Tribal Natives of Bastar Revealed Ant Venom-Derived Peptides with Promising Anti-Malarial Activity. Toxins (Basel) 2022; 14:toxins14110789. [PMID: 36422964 PMCID: PMC9697016 DOI: 10.3390/toxins14110789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/20/2022] [Accepted: 10/22/2022] [Indexed: 11/16/2022] Open
Abstract
Prevailing drug resistance in malaria imposes the major roadblock for the existing interventions necessitating the timely need to search for alternative therapies. Ants in Solenopsis spp, termed 'Fire ants', are well known for their aggressive behavior, which leads to the release of toxic venom. Notably, the tribal natives of the malaria-laden densely forested Bastar region, Chhattisgarh, India, use fire ant sting-based therapy to cure malaria-like high fever. Inspired by this, we have collected the fire ants from the forest of Bastar and extracted peptide and alkaloid fractions from ant venom using HPLC and analyzed them by LC/MS-based applications. Evaluation of the anti-malarial efficacy of these peptide fractions demonstrated a significant reduction in the growth of Plasmodium falciparum (Pf 3D7) in vitro, whereas the alkaloid fraction showed a negligible effect. in vitro hemolytic activity confirmed the venom peptide fraction to be non-hemolytic. Additionally, the venom peptide fraction is purely non-toxic to HepG2 cells. Anti-malarial efficiency of the same in Plasmodium berghei ANKA infected mice models showed a drastic reduction in parasitemia representing promising anti-malarial activity. Overall, our study has unraveled the scientific rationale underlying fire ant sting therapy used as a tribal naturotherapy for curing malaria-like fever, thus, introducing a way forward to develop nature-inspired anti-malarial chemotherapeutics.
Collapse
Affiliation(s)
- Jyoti Kumari
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institute of Eminence, Delhi 201314, India
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India
| | - Raj Kumar Sah
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India
| | | | - Shakeel Ahmad
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland
| | - Soumya Pati
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institute of Eminence, Delhi 201314, India
| | - Shailja Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India
- Correspondence:
| |
Collapse
|
25
|
Vijayan K, Arang N, Wei L, Morrison R, Geiger R, Parks KR, Lewis AJ, Mast FD, Douglass AN, Kain HS, Aitchison JD, Johnson JS, Aderem A, Kaushansky A. A genome-wide CRISPR-Cas9 screen identifies CENPJ as a host regulator of altered microtubule organization during Plasmodium liver infection. Cell Chem Biol 2022; 29:1419-1433.e5. [PMID: 35738280 PMCID: PMC9481707 DOI: 10.1016/j.chembiol.2022.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 02/03/2022] [Accepted: 06/01/2022] [Indexed: 11/30/2022]
Abstract
Prior to initiating symptomatic malaria, a single Plasmodium sporozoite infects a hepatocyte and develops into thousands of merozoites, in part by scavenging host resources, likely delivered by vesicles. Here, we demonstrate that host microtubules (MTs) dynamically reorganize around the developing liver stage (LS) parasite to facilitate vesicular transport to the parasite. Using a genome-wide CRISPR-Cas9 screen, we identified host regulators of cytoskeleton organization, vesicle trafficking, and ER/Golgi stress that regulate LS development. Foci of γ-tubulin localized to the parasite periphery; depletion of centromere protein J (CENPJ), a novel regulator identified in the screen, exacerbated this re-localization and increased infection. We demonstrate that the Golgi acts as a non-centrosomal MT organizing center (ncMTOC) by positioning γ-tubulin and stimulating MT nucleation at parasite periphery. Together, these data support a model where the Plasmodium LS recruits host Golgi to form MT-mediated conduits along which host organelles are recruited to PVM and support parasite development.
Collapse
Affiliation(s)
- Kamalakannan Vijayan
- Center for Infectious Disease Research, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA
| | - Nadia Arang
- Center for Infectious Disease Research, Seattle, WA, USA
| | - Ling Wei
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Robert Morrison
- Center for Infectious Disease Research, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA
| | - Rechel Geiger
- MSTP Program, University of Washington, Seattle, WA, USA
| | - K Rachael Parks
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Adam J Lewis
- Center for Infectious Disease Research, Seattle, WA, USA
| | - Fred D Mast
- Center for Infectious Disease Research, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA
| | - Alyse N Douglass
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Heather S Kain
- Center for Infectious Disease Research, Seattle, WA, USA
| | - John D Aitchison
- Center for Infectious Disease Research, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA; Department of Biochemistry, University of Washington, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA
| | | | - Alan Aderem
- Center for Infectious Disease Research, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Alexis Kaushansky
- Center for Infectious Disease Research, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA; Department of Global Health, University of Washington, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA; Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.
| |
Collapse
|
26
|
Valenciano AL, Gomez-Lorenzo MG, Vega-Rodríguez J, Adams JH, Roth A. In vitro models for human malaria: targeting the liver stage. Trends Parasitol 2022; 38:758-774. [PMID: 35780012 PMCID: PMC9378454 DOI: 10.1016/j.pt.2022.05.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/27/2022] [Accepted: 05/27/2022] [Indexed: 11/16/2022]
Abstract
The Plasmodium liver stage represents a vulnerable therapeutic target to prevent disease progression as the parasite resides in the liver before clinical representation caused by intraerythrocytic development. However, most antimalarial drugs target the blood stage of the parasite's life cycle, and the few drugs that target the liver stage are lethal to patients with a glucose-6-phosphate dehydrogenase deficiency. Furthermore, implementation of in vitro liver models to study and develop novel therapeutics against the liver stage of human Plasmodium species remains challenging. In this review, we focus on the progression of in vitro liver models developed for human Plasmodium spp. parasites, provide a brief review on important assay requirements, and lastly present recommendations to improve models to enhance the discovery process of novel preclinical therapeutics.
Collapse
Affiliation(s)
- Ana Lisa Valenciano
- Center for Global Health and Infectious Diseases, College of Public Health, University of South Florida, Tampa, FL 33612, USA; Global Health Medicines R&D, GlaxoSmithKline, Severo Ochoa 2, Tres Cantos 28760, Madrid, Spain
| | - Maria G Gomez-Lorenzo
- Global Health Medicines R&D, GlaxoSmithKline, Severo Ochoa 2, Tres Cantos 28760, Madrid, Spain
| | - Joel Vega-Rodríguez
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - John H Adams
- Center for Global Health and Infectious Diseases, College of Public Health, University of South Florida, Tampa, FL 33612, USA
| | - Alison Roth
- Department of Drug Discovery, Experimental Therapeutics Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA.
| |
Collapse
|
27
|
Mughal H, Bell EC, Mughal K, Derbyshire ER, Freundlich JS. Random Forest Model Predictions Afford Dual-Stage Antimalarial Agents. ACS Infect Dis 2022; 8:1553-1562. [PMID: 35894649 PMCID: PMC9987178 DOI: 10.1021/acsinfecdis.2c00189] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The need for novel antimalarials is apparent given the continuing disease burden worldwide, despite significant drug discovery advances from the bench to the bedside. In particular, small-molecule agents with potent efficacy against both the liver and blood stages of Plasmodium parasite infection are critical for clinical settings as they would simultaneously prevent and treat malaria with a reduced selection pressure for resistance. While experimental screens for such dual-stage inhibitors have been conducted, the time and cost of these efforts limit their scope. Here, we have focused on leveraging machine learning approaches to discover novel antimalarials with such properties. A random forest modeling approach was taken to predict small molecules with in vitro efficacy versus liver-stage Plasmodium berghei parasites and a lack of human liver cell cytotoxicity. Empirical validation of the model was achieved with the realization of hits with liver-stage efficacy after prospective scoring of a commercial diversity library and consideration of structural diversity. A subset of these hits also demonstrated promising blood-stage Plasmodium falciparum efficacy. These 18 validated dual-stage antimalarials represent novel starting points for drug discovery and mechanism of action studies with significant potential for seeding a new generation of therapies.
Collapse
Affiliation(s)
- Haseeb Mughal
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers University – New Jersey Medical School, 185 South Orange Ave, Newark, NJ, 07103
| | - Elise C. Bell
- Department of Chemistry, Duke University, 124 Science Drive, Durham, NC 27708, USA
| | - Khadija Mughal
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers University – New Jersey Medical School, 185 South Orange Ave, Newark, NJ, 07103
| | - Emily R. Derbyshire
- Department of Chemistry, Duke University, 124 Science Drive, Durham, NC 27708, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, 213 Research Drive, Durham, NC 27710, USA
| | - Joel S. Freundlich
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers University – New Jersey Medical School, 185 South Orange Ave, Newark, NJ, 07103
- Department of Medicine, Center for Emerging and Re-emerging Pathogens, Rutgers University – New Jersey Medical School, Newark, NJ, 07103
| |
Collapse
|
28
|
Schroeder EA, Chirgwin ME, Derbyshire ER. Plasmodium's fight for survival: escaping elimination while acquiring nutrients. Trends Parasitol 2022; 38:544-557. [PMID: 35534377 PMCID: PMC9187605 DOI: 10.1016/j.pt.2022.04.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/10/2022] [Accepted: 04/10/2022] [Indexed: 01/08/2023]
Abstract
Plasmodium parasites extensively alter their host hepatocyte to evade host detection and support an unprecedented replication rate. Host cell manipulation includes association with the host early and late endomembrane systems, where Plasmodium accesses nutrients while suppressing cellular immune processes. Early endomembrane organelles provide an opportunity to sequester an abundance of lipids and proteins, but the association with late endomembrane organelles also risks autophagy-mediated elimination. While not all parasites survive, those that do benefit from a plethora of nutrients provided through this pathway. In this review, we discuss recent advances in our understanding of how Plasmodium parasites balance the need for host nutrients while avoiding elimination during the liver stage.
Collapse
Affiliation(s)
- Erin A Schroeder
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | | | - Emily R Derbyshire
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA; Department of Chemistry, Duke University, Durham, NC, USA.
| |
Collapse
|
29
|
Fernandes P, Loubens M, Le Borgne R, Marinach C, Ardin B, Briquet S, Vincensini L, Hamada S, Hoareau-Coudert B, Verbavatz JM, Weiner A, Silvie O. The AMA1-RON complex drives Plasmodium sporozoite invasion in the mosquito and mammalian hosts. PLoS Pathog 2022; 18:e1010643. [PMID: 35731833 PMCID: PMC9255738 DOI: 10.1371/journal.ppat.1010643] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 07/05/2022] [Accepted: 06/02/2022] [Indexed: 11/22/2022] Open
Abstract
Plasmodium sporozoites that are transmitted by blood-feeding female Anopheles mosquitoes invade hepatocytes for an initial round of intracellular replication, leading to the release of merozoites that invade and multiply within red blood cells. Sporozoites and merozoites share a number of proteins that are expressed by both stages, including the Apical Membrane Antigen 1 (AMA1) and the Rhoptry Neck Proteins (RONs). Although AMA1 and RONs are essential for merozoite invasion of erythrocytes during asexual blood stage replication of the parasite, their function in sporozoites was still unclear. Here we show that AMA1 interacts with RONs in mature sporozoites. By using DiCre-mediated conditional gene deletion in P. berghei, we demonstrate that loss of AMA1, RON2 or RON4 in sporozoites impairs colonization of the mosquito salivary glands and invasion of mammalian hepatocytes, without affecting transcellular parasite migration. Three-dimensional electron microscopy data showed that sporozoites enter salivary gland cells through a ring-like structure and by forming a transient vacuole. The absence of a functional AMA1-RON complex led to an altered morphology of the entry junction, associated with epithelial cell damage. Our data establish that AMA1 and RONs facilitate host cell invasion across Plasmodium invasive stages, and suggest that sporozoites use the AMA1-RON complex to efficiently and safely enter the mosquito salivary glands to ensure successful parasite transmission. These results open up the possibility of targeting the AMA1-RON complex for transmission-blocking antimalarial strategies. Malaria is caused by Plasmodium parasites, which are transmitted by mosquitoes. Infectious stages of the parasite known as sporozoites colonize the mosquito salivary glands and are injected into the host when the insect probes the skin for blood feeding. Sporozoites rapidly migrate to the host liver, invade hepatocytes and differentiate into the next invasive forms, the merozoites, which invade and replicate inside red blood cells. Merozoites invade cells through a specialized structure, known as the moving junction, formed by proteins called AMA1 and RONs. The role of these proteins in sporozoites remains unclear. Here we used conditional genome editing in a rodent malaria model to generate AMA1- and RON-deficient sporozoites. Phenotypic analysis of the mutants revealed that sporozoites use the AMA1-RON complex twice, first in the mosquito to safely enter the salivary glands and ensure successful parasite transmission, then in the mammalian host liver to establish a replicative niche. Our data establish that AMA1 and RONs facilitate host cell invasion across Plasmodium invasive stages, and might represent potential targets for transmission-blocking antimalarial strategies.
Collapse
Affiliation(s)
- Priyanka Fernandes
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, Paris, France
| | - Manon Loubens
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, Paris, France
| | - Rémi Le Borgne
- Institut Jacques Monod, Université Paris Cité, CNRS, UMR 7592, Paris, France
| | - Carine Marinach
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, Paris, France
| | - Béatrice Ardin
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, Paris, France
| | - Sylvie Briquet
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, Paris, France
| | - Laetitia Vincensini
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, Paris, France
| | - Soumia Hamada
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, Paris, France
- Sorbonne Université, INSERM, UMS PASS, Plateforme Post-génomique de la Pitié Salpêtrière (P3S), Paris, France
| | - Bénédicte Hoareau-Coudert
- Sorbonne Université, INSERM, UMS PASS, Plateforme de cytométrie de la Pitié-Salpêtrière (CyPS), Paris, France
| | - Jean-Marc Verbavatz
- Institut Jacques Monod, Université Paris Cité, CNRS, UMR 7592, Paris, France
| | - Allon Weiner
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, Paris, France
| | - Olivier Silvie
- Sorbonne Université, INSERM, CNRS, Centre d’Immunologie et des Maladies Infectieuses, Paris, France
- * E-mail:
| |
Collapse
|
30
|
Introini V, Govendir MA, Rayner JC, Cicuta P, Bernabeu M. Biophysical Tools and Concepts Enable Understanding of Asexual Blood Stage Malaria. Front Cell Infect Microbiol 2022; 12:908241. [PMID: 35711656 PMCID: PMC9192966 DOI: 10.3389/fcimb.2022.908241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 04/27/2022] [Indexed: 12/02/2022] Open
Abstract
Forces and mechanical properties of cells and tissues set constraints on biological functions, and are key determinants of human physiology. Changes in cell mechanics may arise from disease, or directly contribute to pathogenesis. Malaria gives many striking examples. Plasmodium parasites, the causative agents of malaria, are single-celled organisms that cannot survive outside their hosts; thus, thost-pathogen interactions are fundamental for parasite’s biological success and to the host response to infection. These interactions are often combinations of biochemical and mechanical factors, but most research focuses on the molecular side. However, Plasmodium infection of human red blood cells leads to changes in their mechanical properties, which has a crucial impact on disease pathogenesis because of the interaction of infected red blood cells with other human tissues through various adhesion mechanisms, which can be probed and modelled with biophysical techniques. Recently, natural polymorphisms affecting red blood cell biomechanics have also been shown to protect human populations, highlighting the potential of understanding biomechanical factors to inform future vaccines and drug development. Here we review biophysical techniques that have revealed new aspects of Plasmodium falciparum invasion of red blood cells and cytoadhesion of infected cells to the host vasculature. These mechanisms occur differently across Plasmodium species and are linked to malaria pathogenesis. We highlight promising techniques from the fields of bioengineering, immunomechanics, and soft matter physics that could be beneficial for studying malaria. Some approaches might also be applied to other phases of the malaria lifecycle and to apicomplexan infections with complex host-pathogen interactions.
Collapse
Affiliation(s)
- Viola Introini
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
- Cavendish Laboratory, University of Cambridge, Cambridge, United Kingdom
- *Correspondence: Viola Introini,
| | - Matt A. Govendir
- European Molecular Biology Laboratory (EMBL) Barcelona, Barcelona, Spain
| | - Julian C. Rayner
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Pietro Cicuta
- Cavendish Laboratory, University of Cambridge, Cambridge, United Kingdom
| | - Maria Bernabeu
- European Molecular Biology Laboratory (EMBL) Barcelona, Barcelona, Spain
| |
Collapse
|
31
|
Matteucci KC, Correa AAS, Costa DL. Recent Advances in Host-Directed Therapies for Tuberculosis and Malaria. Front Cell Infect Microbiol 2022; 12:905278. [PMID: 35669122 PMCID: PMC9163498 DOI: 10.3389/fcimb.2022.905278] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 04/21/2022] [Indexed: 11/30/2022] Open
Abstract
Tuberculosis (TB), caused by the bacterium Mycobacterium tuberculosis, and malaria, caused by parasites from the Plasmodium genus, are two of the major causes of death due to infectious diseases in the world. Both diseases are treatable with drugs that have microbicidal properties against each of the etiologic agents. However, problems related to treatment compliance by patients and emergence of drug resistant microorganisms have been a major problem for combating TB and malaria. This factor is further complicated by the absence of highly effective vaccines that can prevent the infection with either M. tuberculosis or Plasmodium. However, certain host biological processes have been found to play a role in the promotion of infection or in the pathogenesis of each disease. These processes can be targeted by host-directed therapies (HDTs), which can be administered in conjunction with the standard drug treatments for each pathogen, aiming to accelerate their elimination or to minimize detrimental side effects resulting from exacerbated inflammation. In this review we discuss potential new targets for the development of HDTs revealed by recent advances in the knowledge of host-pathogen interaction biology, and present an overview of strategies that have been tested in vivo, either in experimental models or in patients.
Collapse
Affiliation(s)
- Kely C. Matteucci
- Plataforma de Medicina Translacional Fundação Oswaldo Cruz/Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - André A. S. Correa
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Diego L. Costa
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
- *Correspondence: Diego L. Costa,
| |
Collapse
|
32
|
Mancio-Silva L, Gural N, Real E, Wadsworth MH, Butty VL, March S, Nerurkar N, Hughes TK, Roobsoong W, Fleming HE, Whittaker CA, Levine SS, Sattabongkot J, Shalek AK, Bhatia SN. A single-cell liver atlas of Plasmodium vivax infection. Cell Host Microbe 2022; 30:1048-1060.e5. [PMID: 35443155 DOI: 10.1016/j.chom.2022.03.034] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 01/31/2022] [Accepted: 03/25/2022] [Indexed: 12/15/2022]
Abstract
Malaria-causing Plasmodium vivax parasites can linger in the human liver for weeks to years and reactivate to cause recurrent blood-stage infection. Although they are an important target for malaria eradication, little is known about the molecular features of replicative and non-replicative intracellular liver-stage parasites and their host cell dependence. Here, we leverage a bioengineered human microliver platform to culture patient-derived P. vivax parasites for transcriptional profiling. Coupling enrichment strategies with bulk and single-cell analyses, we capture both parasite and host transcripts in individual hepatocytes throughout the course of infection. We define host- and state-dependent transcriptional signatures and identify unappreciated populations of replicative and non-replicative parasites that share features with sexual transmissive forms. We find that infection suppresses the transcription of key hepatocyte function genes and elicits an anti-parasite innate immune response. Our work provides a foundation for understanding host-parasite interactions and reveals insights into the biology of P. vivax dormancy and transmission.
Collapse
Affiliation(s)
- Liliana Mancio-Silva
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; Institut Pasteur, Université Paris Cité, Inserm U1201, CNRS EMR9195, Unité de Biologie des Interactions Hôte-Parasite, 75015 Paris, France.
| | - Nil Gural
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Eliana Real
- Institut Pasteur, Université Paris Cité, Inserm U1201, CNRS EMR9195, Unité de Biologie des Interactions Hôte-Parasite, 75015 Paris, France
| | - Marc H Wadsworth
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; Department of Chemistry, MIT, Cambridge, MA 02139, USA
| | - Vincent L Butty
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; BioMicro Center, MIT, Cambridge, MA 02139, USA
| | - Sandra March
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Niketa Nerurkar
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Travis K Hughes
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; Department of Chemistry, MIT, Cambridge, MA 02139, USA
| | - Wanlapa Roobsoong
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine Mahidol University, Bangkok 10400, Thailand
| | - Heather E Fleming
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Charlie A Whittaker
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; BioMicro Center, MIT, Cambridge, MA 02139, USA
| | - Stuart S Levine
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; BioMicro Center, MIT, Cambridge, MA 02139, USA
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine Mahidol University, Bangkok 10400, Thailand
| | - Alex K Shalek
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; Department of Chemistry, MIT, Cambridge, MA 02139, USA; Ragon Institute of Massachusetts General Hospital, MIT, and Harvard, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA.
| | - Sangeeta N Bhatia
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; The Wyss Institute for Biologically Inspired Engineering Harvard University Boston, MA 02215, USA.
| |
Collapse
|
33
|
Paoletta MS, Wilkowsky SE. Thrombospondin Related Anonymous Protein Superfamily in Vector-Borne Apicomplexans: The Parasite’s Toolkit for Cell Invasion. Front Cell Infect Microbiol 2022; 12:831592. [PMID: 35463644 PMCID: PMC9019593 DOI: 10.3389/fcimb.2022.831592] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/07/2022] [Indexed: 11/23/2022] Open
Abstract
Apicomplexan parasites transmitted by vectors, including Babesia spp. and Plasmodium spp., cause severe disease in both humans and animals. These parasites have a complex life cycle during which they migrate, invade, and replicate in contrasting hosts such as the mammal and the invertebrate vector. The interaction of parasites with the host cell is mediated by adhesive proteins which play a key role in the different cellular processes regarding successful progression of the life cycle. Thrombospondin related anonymous protein (TRAP) is a superfamily of adhesins that are involved in motility, invasion and egress of the parasite. These proteins are stored and released from apical organelles and have either one or two types of adhesive domains, namely thrombospondin type 1 repeat and von Willebrand factor type A, that upon secretion are located in the extracellular portion of the molecule. Proteins from the TRAP superfamily have been intensively studied in Plasmodium species and to a lesser extent in Babesia spp., where they have proven to be functionally relevant throughout the entire parasite’s journey both in the arthropod vector and in the mammalian host. In recent years new findings provided answers to the role of TRAP proteins and in some cases the function of these adhesins during the parasite’s life cycle was redefined. In this review we will discuss the current knowledge of the diverse roles of the TRAP superfamily in vector-borne parasites from Class Aconoidasida. We will focus on the varied approaches that allowed the understanding of protein function and the relevance of TRAP- superfamily throughout the entire parasite’s cell cycle.
Collapse
|
34
|
Leleu I, Alloo J, Cazenave PA, Roland J, Pied S. Autophagy Pathways in the Genesis of Plasmodium-Derived Microvesicles: A Double-Edged Sword? Life (Basel) 2022; 12:life12030415. [PMID: 35330166 PMCID: PMC8955828 DOI: 10.3390/life12030415] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/02/2022] [Accepted: 03/08/2022] [Indexed: 11/16/2022] Open
Abstract
Malaria, caused by Plasmodium species (spp.), is a deadly parasitic disease that results in approximately 400,000 deaths per year globally. Autophagy pathways play a fundamental role in the developmental stages of the parasite within the mammalian host. They are also involved in the production of Plasmodium-derived extracellular vesicles (EVs), which play an important role in the infection process, either by providing nutrients for parasite growth or by contributing to the immunopathophysiology of the disease. For example, during the hepatic stage, Plasmodium-derived EVs contribute to parasite virulence by modulating the host immune response. EVs help in evading the different autophagy mechanisms deployed by the host for parasite clearance. During cerebral malaria, on the other hand, parasite-derived EVs promote an astrocyte-mediated inflammatory response, through the induction of a non-conventional host autophagy pathway. In this review, we will discuss the cross-talk between Plasmodium-derived microvesicles and autophagy, and how it influences the outcome of infection.
Collapse
|
35
|
Arias CF, Acosta FJ, Fernandez-Arias C. Killing the competition: a theoretical framework for liver-stage malaria. Open Biol 2022; 12:210341. [PMID: 35350863 PMCID: PMC8965401 DOI: 10.1098/rsob.210341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The first stage of malaria infections takes place inside the host's hepatocytes. Remarkably, Plasmodium parasites do not infect hepatocytes immediately after reaching the liver. Instead, they migrate through several hepatocytes before infecting their definitive host cells, thus increasing their chances of immune destruction. Considering that malaria can proceed normally without cell traversal, this is indeed a puzzling behaviour. In fact, the role of hepatocyte traversal remains unknown to date, implying that the current understanding of malaria is incomplete. In this work, we hypothesize that the parasites traverse hepatocytes to actively trigger an immune response in the host. This behaviour would be part of a strategy of superinfection exclusion aimed to reduce intraspecific competition during the blood stage of the infection. Based on this hypothesis, we formulate a comprehensive theory of liver-stage malaria that integrates all the available knowledge about the infection. The interest of this new paradigm is not merely theoretical. It highlights major issues in the current empirical approach to the study of Plasmodium and suggests new strategies to fight malaria.
Collapse
Affiliation(s)
- Clemente F. Arias
- Centro de Investigaciones Biológicas (CSIC), Madrid, Spain,Grupo Interdisciplinar de Sistemas Complejos de Madrid, Spain
| | | | - Cristina Fernandez-Arias
- Departamento de Inmunología, Universidad Complutense de Madrid, Spain,Instituto de Medicina Molecular, Universidade de Lisboa, Portugal
| |
Collapse
|
36
|
van der Boor SC, van Gemert GJ, Hanssen AEJ, van Waardenburg YM, McCall MBB, Bousema T, de Wilt JHW, Sauerwein RW, Yang ASP. Mid-Liver Stage Arrest of Plasmodium falciparum Schizonts in Primary Porcine Hepatocytes. Front Cell Infect Microbiol 2022; 12:834850. [PMID: 35252038 PMCID: PMC8892583 DOI: 10.3389/fcimb.2022.834850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
During co-evolution Plasmodium parasites and vertebrates went through a process of selection resulting in defined and preferred parasite-host combinations. As such, Plasmodium falciparum (Pf) sporozoites can infect human hepatocytes while seemingly incompatible with host cellular machinery of other species. The compatibility between parasite invasion ligands and their respective human hepatocyte receptors plays a key role in Pf host selectivity. However, it is unclear whether the ability of Pf sporozoites to mature in cross-species infection also plays a role in host tropism. Here we used fresh hepatocytes isolated from porcine livers to study permissiveness to Pf sporozoite invasion and development. We monitored intra-hepatic development via immunofluorescence using anti-HSP70, MSP1, EXP1, and EXP2 antibodies. Our data shows that Pf sporozoites can invade non-human hepatocytes and undergo partial maturation with a significant decrease in schizont numbers between day three and day five. A possible explanation is that Pf sporozoites fail to form a parasitophorous vacuolar membrane (PVM) during invasion. Indeed, the observed aberrant EXP1 and EXP2 staining supports the presence of an atypical PVM. Functions of the PVM include the transport of nutrients, export of waste, and offering a protective barrier against intracellular host effectors. Therefore, an atypical PVM likely results in deficiencies that may detrimentally impact parasite development at multiple levels. In summary, despite successful invasion of porcine hepatocytes, Pf development arrests at mid-stage, possibly due to an inability to mobilize critical nutrients across the PVM. These findings underscore the potential of a porcine liver model for understanding the importance of host factors required for Pf mid-liver stage development.
Collapse
Affiliation(s)
- Saskia C. van der Boor
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Geert-Jan van Gemert
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Alex E. J. Hanssen
- Animal Research Facility, Radboud University Medical Center, Nijmegen, Netherlands
| | - Youri M. van Waardenburg
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Matthew B. B. McCall
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Teun Bousema
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Robert W. Sauerwein
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
- TropIQ Health Sciences, Nijmegen, Netherlands
| | - Annie S. P. Yang
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
37
|
Portugal S, Rodriguez A, Prudêncio M. Maria M. Mota: Bringing Plasmodium Liver Infection to the Centre Stage of Malaria Research. Front Cell Infect Microbiol 2022; 12:851484. [PMID: 35211424 PMCID: PMC8860983 DOI: 10.3389/fcimb.2022.851484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
| | - Ana Rodriguez
- Department of Microbiology, New York University School of Medicine, New York City, NY, United States
| | - Miguel Prudêncio
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- *Correspondence: Miguel Prudêncio,
| |
Collapse
|
38
|
Glennon EKK, Tongogara T, Primavera VI, Reeder SM, Wei L, Kaushansky A. Elucidating Spatially-Resolved Changes in Host Signaling During Plasmodium Liver-Stage Infection. Front Cell Infect Microbiol 2022; 11:804186. [PMID: 35111697 PMCID: PMC8801743 DOI: 10.3389/fcimb.2021.804186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/21/2021] [Indexed: 11/22/2022] Open
Abstract
Upon transmission to the human host, Plasmodium sporozoites exit the skin, are taken up by the blood stream, and then travel to the liver where they infect and significantly modify a single hepatocyte. Low infection rates within the liver have made proteomic studies of infected hepatocytes challenging, particularly in vivo, and existing studies have been largely unable to consider how protein and phosphoprotein differences are altered at different spatial locations within the heterogeneous liver. Using digital spatial profiling, we characterized changes in host signaling during Plasmodium yoelii infection in vivo without disrupting the liver tissue. Moreover, we measured alterations in protein expression around infected hepatocytes and identified a subset of CD163+ Kupffer cells that migrate towards infected cells during infection. These data offer the first insight into the heterogeneous microenvironment that surrounds the infected hepatocyte and provide insights into how the parasite may alter its milieu to influence its survival and modulate immunity.
Collapse
Affiliation(s)
- Elizabeth K. K. Glennon
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Tinotenda Tongogara
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
- Grinnell College, Grinnell, IA, United States
| | - Veronica I. Primavera
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Sophia M. Reeder
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Ling Wei
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Alexis Kaushansky
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
- Department of Pediatrics, University of Washington, Seattle, WA, United States
- Brotman Baty Institute for Precision Medicine, Seattle, WA, United States
- *Correspondence: Alexis Kaushansky,
| |
Collapse
|
39
|
Cha SJ, Kim MS, Na CH, Jacobs-Lorena M. Plasmodium sporozoite phospholipid scramblase interacts with mammalian carbamoyl-phosphate synthetase 1 to infect hepatocytes. Nat Commun 2021; 12:6773. [PMID: 34799567 PMCID: PMC8604956 DOI: 10.1038/s41467-021-27109-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 11/04/2021] [Indexed: 11/14/2022] Open
Abstract
After inoculation by the bite of an infected mosquito, Plasmodium sporozoites enter the blood stream and infect the liver, where each infected cell produces thousands of merozoites. These in turn, infect red blood cells and cause malaria symptoms. To initiate a productive infection, sporozoites must exit the circulation by traversing the blood lining of the liver vessels after which they infect hepatocytes with unique specificity. We screened a phage display library for peptides that structurally mimic (mimotope) a sporozoite ligand for hepatocyte recognition. We identified HP1 (hepatocyte-binding peptide 1) that mimics a ~50 kDa sporozoite ligand (identified as phospholipid scramblase). Further, we show that HP1 interacts with a ~160 kDa hepatocyte membrane putative receptor (identified as carbamoyl-phosphate synthetase 1). Importantly, immunization of mice with the HP1 peptide partially protects them from infection by the rodent parasite P. berghei. Moreover, an antibody to the HP1 mimotope inhibits human parasite P. falciparum infection of human hepatocytes in culture. The sporozoite ligand for hepatocyte invasion is a potential novel pre-erythrocytic vaccine candidate. After transmission of Plasmodium sporozoites from infected mosquitoes, parasites first infect hepatocytes. Here, Cha et al. identify a sporozoite ligand (phospholipid scramblase) and the hepatocytic receptor (carbamoyl-phosphate synthetase 1) as relevant for hepatocyte invasion and show that an antibody to hepatocyte-binding peptide 1 (HP1), which structurally mimics the sporozoite ligand, partially protects mice from infection.
Collapse
Affiliation(s)
- Sung-Jae Cha
- Johns Hopkins Bloomberg School of Public Health, Department of Molecular Microbiology and Immunology and Malaria Research Institute, 615N. Wolfe St., Baltimore, MD, 21205, USA.
| | - Min-Sik Kim
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Chan Hyun Na
- Department of Neurology, Institute for Cell Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Marcelo Jacobs-Lorena
- Johns Hopkins Bloomberg School of Public Health, Department of Molecular Microbiology and Immunology and Malaria Research Institute, 615N. Wolfe St., Baltimore, MD, 21205, USA.
| |
Collapse
|
40
|
Poole J, Hartley-Tassell LE, Day CJ, Stanisic DI, Groves PL, Chakravarty S, Lee Sim BK, Hoffman SL, Tiralongo J, Bovin N, Doolan DL, Jennings MP. Identification of the Glycan Binding Profile of Human and Rodent Plasmodium Sporozoites. ACS Infect Dis 2021; 7:2383-2389. [PMID: 34170120 DOI: 10.1021/acsinfecdis.1c00084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The transmission of Plasmodium spp. sporozoites to the mammalian host is the first step in the initiation of the mosquito-borne disease known as malaria. The exact route of transmission from the bloodstream to the liver is still not clearly elucidated, and identification of the host glycan structures bound by the sporozoites may inform as to which host cells are involved. Here, we provide a comprehensive analysis of the glycan structures that sporozoites from the human pathogen, P. falciparum, and the rodent pathogen, P. yoelii, recognize and bind. Glycan array analysis was used to profile the glycans bound by the sporozoites, and the binding affinities of these sporozoite-glycan interactions were then determined by surface plasmon resonance. Data showed that the different Plasmodium spp. bind different classes of glycans. P. falciparum was observed to bind to glycans with terminal N-acetylgalactosamine (GalNAc) or Galactose (Gal) linked to a GalNAc, and the highest-affinity observed was with the GalNAc monosaccharide (12.5 nM). P. yoelii bound glycosaminoglycans, mannosyl glycans, Gal linked to N-acetylglucosamine structures, and the αGal epitope. The highest-affinity interaction for P. yoelii was with the αGal epitope (31.4 nM). This is the first study to identify the key host glycan structures recognized by human and rodent Plasmodium spp. sporozoites. An understanding of how Plasmodium sporozoites interact with the specific glycan structures identified here may provide further insight into this infectious disease that could help direct the design of an effective therapeutic.
Collapse
Affiliation(s)
- Jessica Poole
- Institute for Glycomics, Griffith University, Southport 4222, Queensland Australia
| | | | - Christopher J. Day
- Institute for Glycomics, Griffith University, Southport 4222, Queensland Australia
| | - Danielle I. Stanisic
- Institute for Glycomics, Griffith University, Southport 4222, Queensland Australia
| | - Penny L. Groves
- QIMR Berghofer Medical Research Institute, Herston 4029, Queensland Australia
| | | | - B. Kim Lee Sim
- Sanaria Inc, Rockville, Maryland 20852, United States of America
| | | | - Joe Tiralongo
- Institute for Glycomics, Griffith University, Southport 4222, Queensland Australia
| | - Nicolai Bovin
- Shemyakin Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - Denise L. Doolan
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns 4878, Queensland Australia
| | - Michael P. Jennings
- Institute for Glycomics, Griffith University, Southport 4222, Queensland Australia
| |
Collapse
|
41
|
Vijayan K, Visweswaran GRR, Chandrasekaran R, Trakhimets O, Brown SL, Watson A, Zuck M, Dambrauskas N, Raappana A, Carbonetti S, Kelnhofer-Millevolte L, Glennon EKK, Postiglione R, Sather DN, Kaushansky A. Antibody interference by a non-neutralizing antibody abrogates humoral protection against Plasmodium yoelii liver stage. Cell Rep 2021; 36:109489. [PMID: 34348141 DOI: 10.1016/j.celrep.2021.109489] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 05/14/2021] [Accepted: 07/14/2021] [Indexed: 01/23/2023] Open
Abstract
Both subunit and attenuated whole-sporozoite vaccination strategies against Plasmodium infection have shown promising initial results in malaria-naive westerners but less efficacy in malaria-exposed individuals in endemic areas. Here, we demonstrate proof of concept by using a rodent malaria model in which non-neutralizing antibodies (nNAbs) can directly interfere with protective anti-circumsporozoite protein (CSP) humoral responses. We characterize a monoclonal antibody, RAM1, against Plasmodium yoelii sporozoite major surface antigen CSP. Unlike the canonical PyCSP repeat domain binding and neutralizing antibody (NAb) 2F6, RAM1 does not inhibit sporozoite traversal or entry of hepatocytes in vitro or infection in vivo. Although 2F6 and RAM1 bind non-overlapping regions of the CSP-repeat domain, pre-treatment with RAM1 abrogates the capacity of NAb to block sporozoite traversal and invasion in vitro. Importantly, RAM1 reduces the efficacy of the polyclonal humoral response against PyCSP in vivo. Collectively, our data provide a proof of concept that nNAbs can alter the efficacy of malaria vaccination.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Meghan Zuck
- Seattle Children's Research Institute, Seattle, WA, USA
| | | | | | | | | | | | | | - D Noah Sather
- Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA; Department of Global Health, University of Washington, Seattle, WA, USA.
| | - Alexis Kaushansky
- Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA; Department of Global Health, University of Washington, Seattle, WA, USA; Brotman Baty Research Institute, Seattle, WA, USA; Institute for Stem Cell and Regenerative Medicine, Seattle, WA, USA.
| |
Collapse
|
42
|
Chulanetra M, Chaicumpa W. Revisiting the Mechanisms of Immune Evasion Employed by Human Parasites. Front Cell Infect Microbiol 2021; 11:702125. [PMID: 34395313 PMCID: PMC8358743 DOI: 10.3389/fcimb.2021.702125] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/25/2021] [Indexed: 12/14/2022] Open
Abstract
For the establishment of a successful infection, i.e., long-term parasitism and a complete life cycle, parasites use various diverse mechanisms and factors, which they may be inherently bestowed with, or may acquire from the natural vector biting the host at the infection prelude, or may take over from the infecting host, to outmaneuver, evade, overcome, and/or suppress the host immunity, both innately and adaptively. This narrative review summarizes the up-to-date strategies exploited by a number of representative human parasites (protozoa and helminths) to counteract the target host immune defense. The revisited information should be useful for designing diagnostics and therapeutics as well as vaccines against the respective parasitic infections.
Collapse
Affiliation(s)
- Monrat Chulanetra
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Wanpen Chaicumpa
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
43
|
Digitale JC, Callaway PC, Martin M, Nelson G, Viard M, Rek J, Arinaitwe E, Dorsey G, Kamya M, Carrington M, Rodriguez-Barraquer I, Feeney ME. Association of Inhibitory Killer Cell Immunoglobulin-like Receptor Ligands With Higher Plasmodium falciparum Parasite Prevalence. J Infect Dis 2021; 224:175-183. [PMID: 33165540 PMCID: PMC8491837 DOI: 10.1093/infdis/jiaa698] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/05/2020] [Indexed: 01/01/2023] Open
Abstract
Killer cell immunoglobulin-like receptors (KIRs) and their HLA ligands influence the outcome of many infectious diseases. We analyzed the relationship of compound KIR-HLA genotypes with risk of Plasmodium falciparum infection in a longitudinal cohort of 890 Ugandan individuals. We found that presence of HLA-C2 and HLA-Bw4, ligands for inhibitory KIR2DL1 and KIR3DL1, respectively, increased the likelihood of P. falciparum parasitemia in an additive manner. Individuals homozygous for HLA-C2, which mediates strong inhibition via KIR2DL1, had the highest odds of parasitemia, HLA-C1/C2 heterozygotes had intermediate odds, and individuals homozygous for HLA-C1, which mediates weaker inhibition through KIR2DL2/3, had the lowest odds of parasitemia. In addition, higher surface expression of HLA-C, the ligand for inhibitory KIR2DL1/2/3, was associated with a higher likelihood of parasitemia. Together these data indicate that stronger KIR-mediated inhibition confers a higher risk of P. falciparum parasitemia and suggest that KIR-expressing effector cells play a role in mediating antiparasite immunity.
Collapse
Affiliation(s)
- Jean C Digitale
- Department of Medicine, University of California, San
Francisco, San Francisco, California, USA
- Department of Epidemiology and Biostatistics, University
of California, San Francisco, San Francisco, California, USA
| | - Perri C Callaway
- Department of Medicine, University of California, San
Francisco, San Francisco, California, USA
- Infectious Disease and Immunity Graduate Group, University
of California, Berkeley, Berkeley, California, USA
| | - Maureen Martin
- Basic Science Program, Frederick National Laboratory for
Cancer Research in the Laboratory of Integrative Cancer Immunology, National
Cancer Institute, Bethesda, Maryland, USA
| | - George Nelson
- Advanced Biomedical Computational Science, Frederick
National Laboratory for Cancer Research, Frederick, Maryland,
USA
| | - Mathias Viard
- Basic Science Program, Frederick National Laboratory for
Cancer Research in the Laboratory of Integrative Cancer Immunology, National
Cancer Institute, Bethesda, Maryland, USA
| | - John Rek
- Infectious Diseases Research Collaboration,
Kampala, Uganda
| | - Emmanuel Arinaitwe
- Infectious Diseases Research Collaboration,
Kampala, Uganda
- London School of Hygiene and Tropical
Medicine, London, United
Kingdom
| | - Grant Dorsey
- Department of Medicine, University of California, San
Francisco, San Francisco, California, USA
| | - Moses Kamya
- Infectious Diseases Research Collaboration,
Kampala, Uganda
- Department of Medicine, Makerere University,
Kampala, Uganda
| | - Mary Carrington
- Basic Science Program, Frederick National Laboratory for
Cancer Research in the Laboratory of Integrative Cancer Immunology, National
Cancer Institute, Bethesda, Maryland, USA
- Ragon Institute of MGH MIT and Harvard,
Cambridge, Massachusetts, USA
| | | | - Margaret E Feeney
- Department of Medicine, University of California, San
Francisco, San Francisco, California, USA
- Department of Pediatrics, University of California San
Francisco, San Francisco, California, USA
| |
Collapse
|
44
|
Vijayan K, Wei L, Glennon EKK, Mattocks C, Bourgeois N, Staker B, Kaushansky A. Host-targeted Interventions as an Exciting Opportunity to Combat Malaria. Chem Rev 2021; 121:10452-10468. [PMID: 34197083 DOI: 10.1021/acs.chemrev.1c00062] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Terminal and benign diseases alike in adults, children, pregnant women, and others are successfully treated by pharmacological inhibitors that target human enzymes. Despite extensive global efforts to fight malaria, the disease continues to be a massive worldwide health burden, and new interventional strategies are needed. Current drugs and vector control strategies have contributed to the reduction in malaria deaths over the past 10 years, but progress toward eradication has waned in recent years. Resistance to antimalarial drugs is a substantial and growing problem. Moreover, targeting dormant forms of the malaria parasite Plasmodium vivax is only possible with two approved drugs, which are both contraindicated for individuals with glucose-6-phosphate dehydrogenase deficiency and in pregnant women. Plasmodium parasites are obligate intracellular parasites and thus have specific and absolute requirements of their hosts. Growing evidence has described these host necessities, paving the way for opportunities to pharmacologically target host factors to eliminate Plasmodium infection. Here, we describe progress in malaria research and adjacent fields and discuss key challenges that remain in implementing host-directed therapy against malaria.
Collapse
Affiliation(s)
| | - Ling Wei
- Seattle Children's Research Institute, Seattle, Washington 98109, United States
| | | | - Christa Mattocks
- Department of Global Health, University of Washington, Seattle, Washington 98195, United States
| | - Natasha Bourgeois
- Seattle Children's Research Institute, Seattle, Washington 98109, United States.,Department of Global Health, University of Washington, Seattle, Washington 98195, United States
| | - Bart Staker
- Seattle Children's Research Institute, Seattle, Washington 98109, United States
| | - Alexis Kaushansky
- Seattle Children's Research Institute, Seattle, Washington 98109, United States.,Department of Global Health, University of Washington, Seattle, Washington 98195, United States.,Department of Pediatrics, University of Washington, Seattle, Washington 98105, United States.,Brotman Baty Institute for Precision Medicine, Seattle, Washington 98195, United States
| |
Collapse
|
45
|
Amanzougaghene N, Tajeri S, Yalaoui S, Lorthiois A, Soulard V, Gego A, Rametti A, Risco-Castillo V, Moreno A, Tefit M, van Gemert GJ, Sauerwein RW, Vaillant JC, Ravassard P, Pérignon JL, Froissard P, Mazier D, Franetich JF. The Host Protein Aquaporin-9 is Required for Efficient Plasmodium falciparum Sporozoite Entry into Human Hepatocytes. Front Cell Infect Microbiol 2021; 11:704662. [PMID: 34268141 PMCID: PMC8276244 DOI: 10.3389/fcimb.2021.704662] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/02/2021] [Indexed: 12/11/2022] Open
Abstract
Hepatocyte invasion by Plasmodium sporozoites represents a promising target for innovative antimalarial therapy, but the molecular events mediating this process are still largely uncharacterized. We previously showed that Plasmodium falciparum sporozoite entry into hepatocytes strictly requires CD81. However, CD81-overexpressing human hepatoma cells remain refractory to P. falciparum infection, suggesting the existence of additional host factors necessary for sporozoite entry. Here, through differential transcriptomic analysis of human hepatocytes and hepatoma HepG2-CD81 cells, the transmembrane protein Aquaporin-9 (AQP9) was found to be among the most downregulated genes in hepatoma cells. RNA silencing showed that sporozoite invasion of hepatocytes requires AQP9 expression. AQP9 overexpression in hepatocytes increased their permissiveness to P. falciparum. Moreover, chemical disruption with the AQP9 inhibitor phloretin markedly inhibited hepatocyte infection. Our findings identify AQP9 as a novel host factor required for P. falciparum sporozoite hepatocyte-entry and indicate that AQP9 could be a potential therapeutic target.
Collapse
Affiliation(s)
- Nadia Amanzougaghene
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Shahin Tajeri
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Samir Yalaoui
- Université Pierre et Marie Curie-Paris 6, UMR S945, Paris, France.,INSERM, U945, Paris, France
| | - Audrey Lorthiois
- Université Pierre et Marie Curie-Paris 6, UMR S945, Paris, France.,INSERM, U945, Paris, France
| | - Valérie Soulard
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Audrey Gego
- Université Pierre et Marie Curie-Paris 6, UMR S945, Paris, France.,INSERM, U945, Paris, France
| | - Armelle Rametti
- Université Pierre et Marie Curie-Paris 6, UMR S945, Paris, France.,INSERM, U945, Paris, France
| | | | - Alicia Moreno
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Maurel Tefit
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Geert-Jan van Gemert
- Department of Medical Microbiology, Radboud University Nijmegen Medical Centre, MMB-NCMLS, Nijmegen, Netherlands
| | - Robert W Sauerwein
- Department of Medical Microbiology, Radboud University Nijmegen Medical Centre, MMB-NCMLS, Nijmegen, Netherlands
| | - Jean-Christophe Vaillant
- AP-HP, Service de Chirurgie Digestive, Hépato-Bilio-Pancréatique et Transplantation Hépatique, Centre Hospitalo-Universitaire Pitié-Salpêtrière, Paris, France
| | - Philippe Ravassard
- CR-ICM - LGN CNRS UMR-7991, IFR des Neurosciences, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Jean-Louis Pérignon
- Université Pierre et Marie Curie-Paris 6, UMR S945, Paris, France.,INSERM, U945, Paris, France
| | - Patrick Froissard
- Université Pierre et Marie Curie-Paris 6, UMR S945, Paris, France.,INSERM, U945, Paris, France
| | - Dominique Mazier
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Jean-François Franetich
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| |
Collapse
|
46
|
Steel RWJ, Vigdorovich V, Dambrauskas N, Wilder BK, Arredondo SA, Goswami D, Kumar S, Carbonetti S, Swearingen KE, Nguyen T, Betz W, Camargo N, Fisher BS, Soden J, Thomas H, Freeth J, Moritz RL, Noah Sather D, Kappe SHI. Platelet derived growth factor receptor β (PDGFRβ) is a host receptor for the human malaria parasite adhesin TRAP. Sci Rep 2021; 11:11328. [PMID: 34059712 PMCID: PMC8166973 DOI: 10.1038/s41598-021-90722-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/13/2021] [Indexed: 02/04/2023] Open
Abstract
Following their inoculation by the bite of an infected Anopheles mosquito, the malaria parasite sporozoite forms travel from the bite site in the skin into the bloodstream, which transports them to the liver. The thrombospondin-related anonymous protein (TRAP) is a type 1 transmembrane protein that is released from secretory organelles and relocalized on the sporozoite plasma membrane. TRAP is required for sporozoite motility and host infection, and its extracellular portion contains adhesive domains that are predicted to engage host receptors. Here, we identified the human platelet-derived growth factor receptor β (hPDGFRβ) as one such protein receptor. Deletion constructs showed that the von Willebrand factor type A and thrombospondin repeat domains of TRAP are both required for optimal binding to hPDGFRβ-expressing cells. We also demonstrate that this interaction is conserved in the human-infective parasite Plasmodium vivax, but not the rodent-infective parasite Plasmodium yoelii. We observed expression of hPDGFRβ mainly in cells associated with the vasculature suggesting that TRAP:hPDGFRβ interaction may play a role in the recognition of blood vessels by invading sporozoites.
Collapse
Affiliation(s)
- Ryan W J Steel
- Seattle Children's Research Institute, Seattle, WA, USA
- Infectious Diseases and Immune Defence Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | | | | | - Brandon K Wilder
- Seattle Children's Research Institute, Seattle, WA, USA
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR, 97006, USA
| | | | | | - Sudhir Kumar
- Seattle Children's Research Institute, Seattle, WA, USA
| | | | | | - Thao Nguyen
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Will Betz
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Nelly Camargo
- Seattle Children's Research Institute, Seattle, WA, USA
| | | | - Jo Soden
- Retrogenix Ltd, Chinley, High Peak, SK23 6FJ, UK
| | - Helen Thomas
- Retrogenix Ltd, Chinley, High Peak, SK23 6FJ, UK
| | - Jim Freeth
- Retrogenix Ltd, Chinley, High Peak, SK23 6FJ, UK
| | | | - D Noah Sather
- Seattle Children's Research Institute, Seattle, WA, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, USA.
- Department of Global Health, University of Washington, Seattle, WA, USA.
| | - Stefan H I Kappe
- Seattle Children's Research Institute, Seattle, WA, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, USA.
- Department of Global Health, University of Washington, Seattle, WA, USA.
| |
Collapse
|
47
|
Sekar V, Rivero A, Pigeault R, Gandon S, Drews A, Ahren D, Hellgren O. Gene regulation of the avian malaria parasite Plasmodium relictum, during the different stages within the mosquito vector. Genomics 2021; 113:2327-2337. [PMID: 34023365 DOI: 10.1016/j.ygeno.2021.05.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/26/2021] [Accepted: 05/18/2021] [Indexed: 10/21/2022]
Abstract
The malaria parasite Plasmodium relictum is one of the most widespread species of avian malaria. As in the case of its human counterparts, bird Plasmodium undergoes a complex life cycle infecting two hosts: the arthropod vector and the vertebrate host. In this study, we examined transcriptomes of P. relictum (SGS1) during crucial timepoints within its vector, Culex pipiens quinquefasciatus. Differential gene-expression analyses identified genes linked to the parasites life-stages at: i) a few minutes after the blood meal is ingested, ii) during peak oocyst production phase, iii) during peak sporozoite phase and iv) during the late-stages of the infection. A large amount of genes coding for functions linked to host-immune invasion and multifunctional genes was active throughout the infection cycle. One gene associated with a conserved Plasmodium membrane protein with unknown function was upregulated throughout the parasite development in the vector, suggesting an important role in the successful completion of the sporogonic cycle. Gene expression analysis further identified genes, with unknown functions to be significantly differentially expressed during the infection in the vector as well as upregulation of reticulocyte-binding proteins, which raises the possibility of the multifunctionality of these RBPs. We establish the existence of highly stage-specific pathways being overexpressed during the infection. This first study of gene-expression of a non-human Plasmodium species in its vector provides a comprehensive insight into the molecular mechanisms of the common avian malaria parasite P. relictum and provides essential information on the evolutionary diversity in gene regulation of the Plasmodium's vector stages.
Collapse
Affiliation(s)
| | - Ana Rivero
- MIVEGEC (CNRS - Université de Montpellier - IRD), 34394 Montpellier, France; CREES (Centre de Recherche en Ecologie et Evolution de la Santé), 34394 Montpellier, France
| | - Romain Pigeault
- Department of Biology, Lund University, Sweden; Department of Ecology and Evolution, CH-1015 Lausanne, Switzerland
| | - Sylvain Gandon
- CEFE (CNRS - Université de Montpellier - Université Paul-Valéry - EPHE - IRD), Montpellier, France
| | - Anna Drews
- MEMEG, Department of Biology, Lund University, Sweden
| | - Dag Ahren
- National Bioinformatics Infrastructure Sweden (NBIS), SciLifeLab, Department of Biology, Lund, Sweden
| | - Olof Hellgren
- MEMEG, Department of Biology, Lund University, Sweden.
| |
Collapse
|
48
|
Loubens M, Vincensini L, Fernandes P, Briquet S, Marinach C, Silvie O. Plasmodium sporozoites on the move: Switching from cell traversal to productive invasion of hepatocytes. Mol Microbiol 2021; 115:870-881. [PMID: 33191548 PMCID: PMC8247013 DOI: 10.1111/mmi.14645] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 12/18/2022]
Abstract
Parasites of the genus Plasmodium, the etiological agent of malaria, are transmitted through the bite of anopheline mosquitoes, which deposit sporozoites into the host skin. Sporozoites migrate through the dermis, enter the bloodstream, and rapidly traffic to the liver. They cross the liver sinusoidal barrier and traverse several hepatocytes before switching to productive invasion of a final one for replication inside a parasitophorous vacuole. Cell traversal and productive invasion are functionally independent processes that require proteins secreted from specialized secretory organelles known as micronemes. In this review, we summarize the current understanding of how sporozoites traverse through cells and productively invade hepatocytes, and discuss the role of environmental sensing in switching from a migratory to an invasive state. We propose that timely controlled secretion of distinct microneme subsets could play a key role in successful migration and infection of hepatocytes. A better understanding of these essential biological features of the Plasmodium sporozoite may contribute to the development of new strategies to fight against the very first and asymptomatic stage of malaria.
Collapse
Affiliation(s)
- Manon Loubens
- Centre d’Immunologie et des Maladies InfectieusesSorbonne Université, INSERM, CNRS, CIMI‐ParisParisFrance
| | - Laetitia Vincensini
- Centre d’Immunologie et des Maladies InfectieusesSorbonne Université, INSERM, CNRS, CIMI‐ParisParisFrance
| | - Priyanka Fernandes
- Centre d’Immunologie et des Maladies InfectieusesSorbonne Université, INSERM, CNRS, CIMI‐ParisParisFrance
| | - Sylvie Briquet
- Centre d’Immunologie et des Maladies InfectieusesSorbonne Université, INSERM, CNRS, CIMI‐ParisParisFrance
| | - Carine Marinach
- Centre d’Immunologie et des Maladies InfectieusesSorbonne Université, INSERM, CNRS, CIMI‐ParisParisFrance
| | - Olivier Silvie
- Centre d’Immunologie et des Maladies InfectieusesSorbonne Université, INSERM, CNRS, CIMI‐ParisParisFrance
| |
Collapse
|
49
|
Sena-dos-Santos C, Braga-da-Silva C, Marques D, Azevedo dos Santos Pinheiro J, Ribeiro-dos-Santos Â, Cavalcante GC. Unraveling Cell Death Pathways during Malaria Infection: What Do We Know So Far? Cells 2021; 10:479. [PMID: 33672278 PMCID: PMC7926694 DOI: 10.3390/cells10020479] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/07/2020] [Accepted: 12/11/2020] [Indexed: 12/15/2022] Open
Abstract
Malaria is a parasitic disease (caused by different Plasmodium species) that affects millions of people worldwide. The lack of effective malaria drugs and a vaccine contributes to this disease, continuing to cause major public health and socioeconomic problems, especially in low-income countries. Cell death is implicated in malaria immune responses by eliminating infected cells, but it can also provoke an intense inflammatory response and lead to severe malaria outcomes. The study of the pathophysiological role of cell death in malaria in mammalians is key to understanding the parasite-host interactions and design prophylactic and therapeutic strategies for malaria. In this work, we review malaria-triggered cell death pathways (apoptosis, autophagy, necrosis, pyroptosis, NETosis, and ferroptosis) and we discuss their potential role in the development of new approaches for human malaria therapies.
Collapse
Affiliation(s)
- Camille Sena-dos-Santos
- Programa de Pós-Graduação em Genética e Biologia Molecular, Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66.075-110, Brazil; (C.S.-d.-S.); (C.B.-d.-S.); (D.M.); (J.A.d.S.P.); (Â.R.-d.-S.)
| | - Cíntia Braga-da-Silva
- Programa de Pós-Graduação em Genética e Biologia Molecular, Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66.075-110, Brazil; (C.S.-d.-S.); (C.B.-d.-S.); (D.M.); (J.A.d.S.P.); (Â.R.-d.-S.)
| | - Diego Marques
- Programa de Pós-Graduação em Genética e Biologia Molecular, Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66.075-110, Brazil; (C.S.-d.-S.); (C.B.-d.-S.); (D.M.); (J.A.d.S.P.); (Â.R.-d.-S.)
| | - Jhully Azevedo dos Santos Pinheiro
- Programa de Pós-Graduação em Genética e Biologia Molecular, Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66.075-110, Brazil; (C.S.-d.-S.); (C.B.-d.-S.); (D.M.); (J.A.d.S.P.); (Â.R.-d.-S.)
| | - Ândrea Ribeiro-dos-Santos
- Programa de Pós-Graduação em Genética e Biologia Molecular, Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66.075-110, Brazil; (C.S.-d.-S.); (C.B.-d.-S.); (D.M.); (J.A.d.S.P.); (Â.R.-d.-S.)
- Programa de Pós-Graduação em Oncologia e Ciências Médicas, Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém 66.075-110, Brazil
| | - Giovanna C. Cavalcante
- Programa de Pós-Graduação em Genética e Biologia Molecular, Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66.075-110, Brazil; (C.S.-d.-S.); (C.B.-d.-S.); (D.M.); (J.A.d.S.P.); (Â.R.-d.-S.)
| |
Collapse
|
50
|
Arredondo SA, Schepis A, Reynolds L, Kappe SHI. Secretory Organelle Function in the Plasmodium Sporozoite. Trends Parasitol 2021; 37:651-663. [PMID: 33589364 DOI: 10.1016/j.pt.2021.01.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/20/2021] [Accepted: 01/20/2021] [Indexed: 12/14/2022]
Abstract
Plasmodium sporozoites exhibit a complex infection biology in the mosquito and mammalian hosts. The sporozoite apical secretory organelles, the micronemes and rhoptries, store protein mediators of parasite/host/vector interactions and must secrete them in a temporally and spatially well orchestrated manner. Micronemal proteins are critical for sporozoite motility throughout its journey from the mosquito midgut oocyst to the mammalian liver, and also for cell traversal (CT) and hepatocyte invasion. Rhoptry proteins, until recently thought to be only important for hepatocyte invasion, appear to also play an unexpected role in motility and in the interaction with mosquito tissue. Therefore, navigating the different microenvironments with secretion likely requires the sporozoite to have a more complex system of secretory organelles than previously appreciated.
Collapse
Affiliation(s)
- Silvia A Arredondo
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Antonino Schepis
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Laura Reynolds
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Stefan H I Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA; Department of Pediatrics, University of Washington, Seattle, WA 98105, USA; Department of Global Health, University of Washington, Seattle, WA 98105, USA.
| |
Collapse
|