1
|
Ngotho P, Dantzler Press K, Peedell M, Muasya W, Omondi BR, Otoboh SE, Gomez J, Coronado L, Seydel KB, Kapulu M, Laufer M, Taylor T, Bousema T, Marti M. Reversible host cell surface remodelling limits immune recognition and maximizes survival of Plasmodium falciparum gametocytes. PLoS Pathog 2025; 21:e1013110. [PMID: 40354414 DOI: 10.1371/journal.ppat.1013110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 05/20/2025] [Accepted: 04/08/2025] [Indexed: 05/14/2025] Open
Abstract
Reducing malaria transmission has been a major pillar of control programmes and is considered crucial for achieving malaria elimination. Gametocytes, the transmissible forms of the P. falciparum parasite, arise during the blood stage of the parasite and develop through 5 morphologically distinct stages. Immature gametocytes (stage I-IV) sequester and develop in the extravascular niche of the bone marrow and possibly spleen. Only mature stage V gametocytes re-enter peripheral circulation to be taken up by mosquitoes for successful onward transmission. We have recently shown that immature, but not mature gametocytes are targets of host immune responses and identified putative target surface antigens. We hypothesize that these antigens play a role in gametocyte sequestration and contribute to acquired transmission-reducing immunity. Here we demonstrate that surface antigen expression, serum reactivity by human IgG, and opsonic phagocytosis by macrophages all show similar dynamics during gametocyte maturation, i.e., peaking in the immature stages and tapering off in mature gametocytes. Moreover, the switch in surface reactivity coincides with reversal in phosphatidylserine (PS) surface exposure, a marker for red blood cell age and clearance. PS is exposed on the surface of a proportion of immature gametocyte-infected RBCs (as well as in late asexual stages) but is removed from the surface in later gametocyte stages (IV-V). Using parasite reverse genetics and drug perturbations, we confirm that parasite protein export into the host cell and phospholipid scramblase activity are required for the observed surface modifications in asexual and sexual P. falciparum stages. Based on these findings we propose that the reversible surface remodelling allows (i) immature gametocyte sequestration in bone marrow followed by (ii) mature gametocyte release into peripheral circulation (and immune evasion due to loss of surface antigens), therefore contributing to mature gametocyte survival in vivo and onward transmission to mosquitoes. Importantly, blocking scramblase activity during gametocyte maturation results in efficient clearance of mature gametocytes, revealing a potential path for transmission blocking interventions. Our studies have important implications for our understanding of parasite biology and form a starting point for novel intervention strategies to simultaneously reduce parasite burden and transmission.
Collapse
Affiliation(s)
- Priscilla Ngotho
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, United Kingdom
- Institute of Parasitology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Kathleen Dantzler Press
- Department of Medicine, Stanford University School of Medicine, Stanford, California, United States of America
| | - Megan Peedell
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, United Kingdom
- Institute of Parasitology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - William Muasya
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, United Kingdom
| | - Brian Roy Omondi
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Stanley E Otoboh
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Jahiro Gomez
- Instituto de Investigaciones Científicas y Servicios de alta Tecnología de Panamá, Panamá City, Panamá
| | - Lorena Coronado
- Instituto de Investigaciones Científicas y Servicios de alta Tecnología de Panamá, Panamá City, Panamá
| | - Karl B Seydel
- Department of Osteopathic Medical Specialties, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan, United States of America
- Blantyre Malaria Project, Kamuzu University of Health Sciences, Blantyre, Malawi
| | | | - Miriam Laufer
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine Baltimore, Maryland, United States of America
| | - Terrie Taylor
- Department of Osteopathic Medical Specialties, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan, United States of America
- Blantyre Malaria Project, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Teun Bousema
- Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Matthias Marti
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, United Kingdom
- Institute of Parasitology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
2
|
Küster N, Roling L, Ouayoue A, Steeg K, Przyborski JM. A Systematic Targeted Genetic Screen Identifies Proteins Involved in Cytoadherence of the Malaria Parasite P. falciparum. Mol Microbiol 2025; 123:330-343. [PMID: 39829323 PMCID: PMC11976115 DOI: 10.1111/mmi.15337] [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: 07/24/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 01/22/2025]
Abstract
Immediately after invading their chosen host cell, the mature human erythrocyte, malaria parasites begin to export an array of proteins to this compartment, where they initiate processes that are prerequisite for parasite survival and propagation, including nutrient import and immune evasion. One consequence of these activities is the emergence of novel adhesive phenotypes that can lead directly to pathology in the human host. To identify parasite proteins involved in this process, we used modern genetic tools to target genes encoding 15 exported parasite proteins, selected by an in silico workflow. This resulted in four genetically modified parasite lines that were then characterised in detail. Of these lines, three could be shown to have aberrations in adhesion, and of these one appears to have a block in the transport and/or correct folding of the major surface adhesin PfEMP1 (Plasmodium falciparum erythrocyte membrane protein 1). Our data expand the known factors involved in this important process and once again highlight the complexity of this phenomenon.
Collapse
Affiliation(s)
- Nina Küster
- Department of Biochemistry and Molecular BiologyJustus‐Liebig University GießenGießenGermany
| | - Lena Roling
- Department of Biochemistry and Molecular BiologyJustus‐Liebig University GießenGießenGermany
| | - Ardin Ouayoue
- Department of Biochemistry and Molecular BiologyJustus‐Liebig University GießenGießenGermany
| | - Katharina Steeg
- Department of Biochemistry and Molecular BiologyJustus‐Liebig University GießenGießenGermany
| | - Jude M. Przyborski
- Department of Biochemistry and Molecular BiologyJustus‐Liebig University GießenGießenGermany
| |
Collapse
|
3
|
Thiam F, Djoumoi D, Mbaye MN, Fall A, Diouara AAM, Diop M, Nguer CM, Mbengue B, Diop G, Kohli E, Dieye A. Secreted extracellular heat shock protein gp96 and inflammatory cytokines are markers of severe malaria outcome. Cell Stress Chaperones 2025; 30:48-56. [PMID: 39732362 PMCID: PMC11761890 DOI: 10.1016/j.cstres.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 12/23/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024] Open
Abstract
Malaria caused by Plasmodium spp., is a major public health issue in sub-Saharan Africa. The fight against malaria has stalled due to increasing resistance to treatments and insecticides. There is an urgent need to focus on new therapeutic targets to combat malaria effectively. This study aimed to measure the secreted heat shock protein gp96 levels in both malaria patients and controls. Indeed, gp96 plays a crucial role in parasite survival within the host and in establishing a successful infection. Therefore, gp96 could be a promising target for antimalarial drugs. In our study, we included 60 malaria patients, 30 with severe malaria (SM) and 30 with uncomplicated malaria (UM). Additionally, 28 controls were included. Using the ELISA method, we measured gp96 levels in the participants' blood samples. We then used the Mann-Whitney or analyse of variance tests to calculate descriptive statistics and determined the correlation between gp96 level and parasitemia using Spearman's rank correlation test. The study found that gp96 levels in the plasma significantly increased in malaria patients (23.86 ng/mL) compared to control (5.88 ng/mL), with a P < 0.0001. Interestingly, there was a significant difference between SM (27.56 ng/mL) and UM (13.9 ng/mL), with a P-value of 0.001. These findings are accompanied by significantly higher parasitemia and elevated proinflammatory cytokines such as IL-17A and IL-1β levels in SM patients compared to UM and controls. Furthermore, there was no significant positive correlation between gp96 levels and parasitemia/proinflammatory cytokines. Our research has revealed, for the first time, that individuals with SM have significantly higher levels of gp96 in the context of high parasitemia and proinflammatory cytokines. Our preliminary results will be taken further to evaluate gp96 as a valuable biomarker for the diagnosis of SM and a potential target for antimalarial drug discovery.
Collapse
Affiliation(s)
- Fatou Thiam
- Groupe de Recherche Biotechnologies Appliquees & Bioprocedes Environnementaux, Ecole Superieure Polytechnique, Universite Cheikh Anta Diop de Dakar, 5085 Dakar-Fann, Senegal.
| | - Djibaba Djoumoi
- Groupe de Recherche Biotechnologies Appliquees & Bioprocedes Environnementaux, Ecole Superieure Polytechnique, Universite Cheikh Anta Diop de Dakar, 5085 Dakar-Fann, Senegal
| | - Mame Ndew Mbaye
- Groupe de Recherche Biotechnologies Appliquees & Bioprocedes Environnementaux, Ecole Superieure Polytechnique, Universite Cheikh Anta Diop de Dakar, 5085 Dakar-Fann, Senegal
| | - Aminata Fall
- Unite postulante de Biologie Genetique, Genomique et Bio-informatique (G2B), Departement de Biologie animale, Faculté des Sciences et Techniques, Universite Cheikh Anta DIOP, Avenue Cheikh Anta DIOP, BP: 5005, Dakar, Senegal
| | - Abou Abdallah Malick Diouara
- Groupe de Recherche Biotechnologies Appliquees & Bioprocedes Environnementaux, Ecole Superieure Polytechnique, Universite Cheikh Anta Diop de Dakar, 5085 Dakar-Fann, Senegal
| | - Mamadou Diop
- Groupe de Recherche Biotechnologies Appliquees & Bioprocedes Environnementaux, Ecole Superieure Polytechnique, Universite Cheikh Anta Diop de Dakar, 5085 Dakar-Fann, Senegal
| | - Cheikh Momar Nguer
- Groupe de Recherche Biotechnologies Appliquees & Bioprocedes Environnementaux, Ecole Superieure Polytechnique, Universite Cheikh Anta Diop de Dakar, 5085 Dakar-Fann, Senegal
| | - Babacar Mbengue
- Service d'Immunologie, Faculté de Médecine, de Pharmacie et d'Odontostomatologie, Université Cheikh Anta DIOP, Avenue Cheikh Anta DIOP, BP: 5005, Dakar, Senegal
| | - Gora Diop
- Unite postulante de Biologie Genetique, Genomique et Bio-informatique (G2B), Departement de Biologie animale, Faculté des Sciences et Techniques, Universite Cheikh Anta DIOP, Avenue Cheikh Anta DIOP, BP: 5005, Dakar, Senegal
| | - Evelyne Kohli
- Université de Bourgogne Franche-Comté, LNC UMR 866 Dijon, France
| | - Alioune Dieye
- Service d'Immunologie, Faculté de Médecine, de Pharmacie et d'Odontostomatologie, Université Cheikh Anta DIOP, Avenue Cheikh Anta DIOP, BP: 5005, Dakar, Senegal
| |
Collapse
|
4
|
Catacalos-Goad C, Chakrabarti M, Salem DH, Camporeale C, Somalraju S, Tegowski M, Singh R, Reid RW, Janies DA, Meyer KD, Janga SC, Hunt AG, Chakrabarti K. Nucleotide-resolution Mapping of RNA N6-Methyladenosine (m6A) modifications and comprehensive analysis of global polyadenylation events in mRNA 3' end processing in malaria pathogen Plasmodium falciparum. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.07.631827. [PMID: 39829786 PMCID: PMC11741415 DOI: 10.1101/2025.01.07.631827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Plasmodium falciparum is an obligate human parasite of the phylum Apicomplexa and is the causative agent of the most lethal form of human malaria. Although N6-methyladenosine modification is thought to be one of the major post-transcriptional regulatory mechanisms for stage-specific gene expression in apicomplexan parasites, the precise base position of m6A in mRNAs or noncoding RNAs in these parasites remains unknown. Here, we report global nucleotide-resolution mapping of m6A residues in P. falciparum using DART-seq technology, which quantitatively displayed a stage-specific, dynamic distribution pattern with enrichment near mRNA 3' ends. In this process we identified 894, 788, and 1,762 m6A-modified genes in Ring, Trophozoite and Schizont stages respectively, with an average of 5-7 m6A sites per-transcript at the individual gene level. Notably, several genes involved in malaria pathophysiology, such as KAHRP, ETRAMPs, SERA and stress response genes, such as members of Heat Shock Protein (HSP) family are highly enriched in m6A and therefore could be regulated by this RNA modification. Since we observed preferential methylation at the 3' ends of P. falciparum transcripts and because malaria polyadenylation specificity factor PfCPSF30 harbors an m6A reader 'YTH' domain, we reasoned that m6A might play an important role in 3'-end processing of malaria mRNAs. To investigate this, we used two complementary high-throughput RNA 3'-end mapping approaches, which provided an initial framework to explore potential roles of m6A in the regulation of alternative polyadenylation (APA) during malaria development in human hosts.
Collapse
Affiliation(s)
- Cassandra Catacalos-Goad
- Department of Biological Sciences, University of North Carolina, Charlotte, NC, USA, United States of America
| | - Manohar Chakrabarti
- School of Integrative Biological and Chemical Sciences, University of Texas Rio Grande Valley, Edinburg, TX
| | - Doaa Hassan Salem
- Department of Biomedical Engineering and Informatics, Luddy School of Informatics, Computing and Engineering, Indiana University Indianapolis (IUI), Indianapolis, Indiana, United States of America
| | - Carli Camporeale
- Department of Biological Sciences, University of North Carolina, Charlotte, NC, USA, United States of America
| | - Sahiti Somalraju
- Department of Biomedical Engineering and Informatics, Luddy School of Informatics, Computing and Engineering, Indiana University Indianapolis (IUI), Indianapolis, Indiana, United States of America
| | - Matthew Tegowski
- Department of Biochemistry, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Ruchi Singh
- Department of Biological Sciences, University of North Carolina, Charlotte, NC, USA, United States of America
| | - Robert W Reid
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, North Carolina, United States of America
| | - Daniel A Janies
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, North Carolina, United States of America
| | - Kate D Meyer
- Department of Biochemistry, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Sarath Chandra Janga
- Department of Biomedical Engineering and Informatics, Luddy School of Informatics, Computing and Engineering, Indiana University Indianapolis (IUI), Indianapolis, Indiana, United States of America
| | - Arthur G Hunt
- Department of Plant and Soil Sciences, University of Kentucky, Lexington, KY, USA, United States of America
| | - Kausik Chakrabarti
- Department of Biological Sciences, University of North Carolina, Charlotte, NC, USA, United States of America
| |
Collapse
|
5
|
Allweier J, Bartels M, Torabi H, Tauler MDPM, Metwally NG, Roeder T, Gutsmann T, Bruchhaus I. Cytoadhesion of Plasmodium falciparum-Infected Red Blood Cells Changes the Expression of Cytokine-, Histone- and Antiviral Protein-Encoding Genes in Brain Endothelial Cells. Mol Microbiol 2024; 122:948-967. [PMID: 39630601 DOI: 10.1111/mmi.15331] [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: 10/24/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024]
Abstract
Malaria remains a significant global health problem, mainly due to Plasmodium falciparum, which is responsible for most fatal infections. Infected red blood cells (iRBCs) evade spleen clearance by adhering to endothelial cells (ECs), triggering capillary blockage, inflammation, endothelial dysfunction and altered vascular permeability, prompting an endothelial transcriptional response. The iRBCIT4var04/HBEC-5i model, where iRBCs present IT4var04 (VAR2CSA) on their surface, was used to analyze the effects of iRBC binding on ECs at different temperature (37°C vs. 40°C). Binding of non-infected RBCs (niRBCs) and fever alone altered the expression of hundreds of genes in ECs. Comparing the expression profile of HBEC-5i cells cultured either in the presence of iRBCs or in the presence of niRBCs revealed significant upregulation of genes linked to immune response, nucleosome assembly, NF-kappa B signaling, angiogenesis, and antiviral immune response/interferon-alpha/beta signaling. Raising the temperature to 40°C, simulating fever, led to further upregulation of many genes, particularly those involved in cytokine production and angiogenesis. In summary, the presence of iRBCs stimulates ECs, activating several immunological pathways and affecting antiviral (-parasitic) mechanisms and angiogenesis. Our data uncovered the induction of the interferon-alpha/beta signaling pathway in ECs in response to iRBCs.
Collapse
Affiliation(s)
- Johannes Allweier
- Research Group Host-Parasite Interaction, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Division of Biophysics, Research Center Borstel, Leibniz-Center for Medicine and Biosciences, Borstel, Germany
| | - Michael Bartels
- Research Group Host-Parasite Interaction, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Hanifeh Torabi
- Research Group Host-Parasite Interaction, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | | | - Nahla Galal Metwally
- Research Group Host-Parasite Interaction, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Thomas Roeder
- Department Molecular Physiology, Zoology, Kiel University, Kiel, Germany
- DZL, German Center for Lung Research, ARCN, Airway Research Center North, Kiel, Germany
| | - Thomas Gutsmann
- Division of Biophysics, Research Center Borstel, Leibniz-Center for Medicine and Biosciences, Borstel, Germany
| | - Iris Bruchhaus
- Research Group Host-Parasite Interaction, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Department of Biology, University of Hamburg, Hamburg, Germany
| |
Collapse
|
6
|
Tang R, Fan Y, Lu B, Jiang Q, Cheng X, Zhang Z, Shen L, Shang X. The RNA m 5C methyltransferase NSUN1 modulates human malaria gene expression during intraerythrocytic development. Front Cell Infect Microbiol 2024; 14:1474229. [PMID: 39435184 PMCID: PMC11491294 DOI: 10.3389/fcimb.2024.1474229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 08/29/2024] [Indexed: 10/23/2024] Open
Abstract
Introduction Plasmodium falciparum is the most damaging malaria pathogen and brings a heavy burden to global health. Host switching and morphological changes in P. falciparum are dependent on an effective gene expression regulatory system. C5 methylation of cytosines is a common RNA modification in eukaryotes, and the NSUN family are essential m5C modification executors. Currently, little is known about this family in Plasmodium spp. In this study, we focus on exploring the function of PfNSUN1 protein. Methods An efficient CRISPR/Cas9 gene editing technique was applied to construct the PfNSUN1 knockdown strain. The knockdown efficiency was confirmed by growth curves and western blot experiments. The knockdown transcriptome data was acquired to find differentially expressed genes, and target genes of PfNSUN1 protein were identified by RNA immunoprecipitation and high-throughput sequencing experiments. Results The efficiency of PfNSUN1 protein down-regulated was about 34%. RNA-seq data revealed that differentially expressed genes were mainly down-regulated. And there were 224, 278, 556 genes that were down-regulated with more than 2-fold changes and p-adj<0.05 at ring, trophozoite and schizont stages, respectively. PfNSUN1 protein was significantly enriched on 154 target genes, including 28S ribosomal RNA and pfap2-g5 transcription factor. Discussion PfNSUN1 is a crucial RNA post-transcriptional modification protein in P. falciparum. It plays a pivotal role in regulating gene expression and parasite growth by targeting 28S ribosomal RNA and pfap2-g5 transcription factor.
Collapse
Affiliation(s)
- Ruoyu Tang
- Department of Parasitology, Xiangya School of Medicine, Central South University, Changsha, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Tongji Hospital, Clinical Center for Brain and Spinal Cord Research, School of Medicine, Tongji University, Shanghai, China
| | - Yanting Fan
- Department of Parasitology, School of Medicine, Northwest University, Xi’an, Shanxi, China
- Laboratory of Molecular Parasitology, The Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Clinical Center for Brain and Spinal Cord Research, School of Medicine, Tongji University, Shanghai, China
| | - BinBin Lu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Tongji Hospital, Clinical Center for Brain and Spinal Cord Research, School of Medicine, Tongji University, Shanghai, China
| | - Qunfeng Jiang
- Department of General Manager Office, Hunan Xingchen Biotechnology Company, Yongzhou, China
| | - Xinyu Cheng
- Department of Parasitology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Zuping Zhang
- Department of Parasitology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Li Shen
- Laboratory of Molecular Parasitology, The Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Clinical Center for Brain and Spinal Cord Research, School of Medicine, Tongji University, Shanghai, China
| | - Xiaomin Shang
- Department of Parasitology, Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
7
|
Charneau S, de Oliveira LS, Zenonos Z, Hopp CS, Bastos IMD, Loew D, Lombard B, Pandolfo Silveira A, de Carvalho Nardeli Basílio Lobo G, Bao SN, Grellier P, Rayner JC. APEX2-based proximity proteomic analysis identifies candidate interactors for Plasmodium falciparum knob-associated histidine-rich protein in infected erythrocytes. Sci Rep 2024; 14:11242. [PMID: 38755230 PMCID: PMC11099048 DOI: 10.1038/s41598-024-61295-w] [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: 12/19/2023] [Accepted: 05/03/2024] [Indexed: 05/18/2024] Open
Abstract
The interaction of Plasmodium falciparum-infected red blood cells (iRBCs) with the vascular endothelium plays a crucial role in malaria pathology and disease. KAHRP is an exported P. falciparum protein involved in iRBC remodelling, which is essential for the formation of protrusions or "knobs" on the iRBC surface. These knobs and the proteins that are concentrated within them allow the parasites to escape the immune response and host spleen clearance by mediating cytoadherence of the iRBC to the endothelial wall, but this also slows down blood circulation, leading in some cases to severe cerebral and placental complications. In this work, we have applied genetic and biochemical tools to identify proteins that interact with P. falciparum KAHRP using enhanced ascorbate peroxidase 2 (APEX2) proximity-dependent biotinylation and label-free shotgun proteomics. A total of 30 potential KAHRP-interacting candidates were identified, based on the assigned fragmented biotinylated ions. Several identified proteins have been previously reported to be part of the Maurer's clefts and knobs, where KAHRP resides. This study may contribute to a broader understanding of P. falciparum protein trafficking and knob architecture and shows for the first time the feasibility of using APEX2-proximity labelling in iRBCs.
Collapse
Affiliation(s)
- Sébastien Charneau
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasília, Brasília, 70910-900, Brazil.
| | - Lucas Silva de Oliveira
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasília, Brasília, 70910-900, Brazil
- UMR 7245 MCAM Molecules of Communication and Adaptation of Microorganisms, Muséum National d'Histoire Naturelle, CNRS, 75231, Paris Cedex 05, France
| | - Zenon Zenonos
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK
- Biologics Engineering, Oncology R&D, AstraZenecaGranta Park, Cambridge, UK
| | - Christine S Hopp
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK
- Protozoa Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Izabela M D Bastos
- Laboratory of Host Pathogen Interaction, Department of Cell Biology, Institute of Biology, University of Brasília, Brasília, 70910-900, Brazil
| | - Damarys Loew
- Institut Curie, Centre de Recherche, PSL Research University, CurieCoreTech Mass Spectrometry Proteomics, 26 rue d'Ulm, 75248, Paris Cedex 05, France
| | - Bérangère Lombard
- Institut Curie, Centre de Recherche, PSL Research University, CurieCoreTech Mass Spectrometry Proteomics, 26 rue d'Ulm, 75248, Paris Cedex 05, France
| | - Ariane Pandolfo Silveira
- Laboratory of Microscopy and Microanalysis, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, 70910-900, Brazil
| | | | - Sônia Nair Bao
- Laboratory of Microscopy and Microanalysis, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, 70910-900, Brazil
| | - Philippe Grellier
- UMR 7245 MCAM Molecules of Communication and Adaptation of Microorganisms, Muséum National d'Histoire Naturelle, CNRS, 75231, Paris Cedex 05, France
| | - Julian C Rayner
- Cambridge Institute for Medical Research, University of Cambridge, Hills Road, Cambridge, CB2 0XY, UK
| |
Collapse
|
8
|
Hoo R, Ruiz-Morales ER, Kelava I, Rawat M, Mazzeo CI, Tuck E, Sancho-Serra C, Chelaghma S, Predeus AV, Murray S, Fernandez-Antoran D, Waller RF, Álvarez-Errico D, Lee MCS, Vento-Tormo R. Acute response to pathogens in the early human placenta at single-cell resolution. Cell Syst 2024; 15:425-444.e9. [PMID: 38703772 DOI: 10.1016/j.cels.2024.04.002] [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: 05/19/2023] [Revised: 12/01/2023] [Accepted: 04/16/2024] [Indexed: 05/06/2024]
Abstract
The placenta is a selective maternal-fetal barrier that provides nourishment and protection from infections. However, certain pathogens can attach to and even cross the placenta, causing pregnancy complications with potential lifelong impacts on the child's health. Here, we profiled at the single-cell level the placental responses to three pathogens associated with intrauterine complications-Plasmodium falciparum, Listeria monocytogenes, and Toxoplasma gondii. We found that upon exposure to the pathogens, all placental lineages trigger inflammatory responses that may compromise placental function. Additionally, we characterized the responses of fetal macrophages known as Hofbauer cells (HBCs) to each pathogen and propose that they are the probable niche for T. gondii. Finally, we revealed how P. falciparum adapts to the placental microenvironment by modulating protein export into the host erythrocyte and nutrient uptake pathways. Altogether, we have defined the cellular networks and signaling pathways mediating acute placental inflammatory responses that could contribute to pregnancy complications.
Collapse
Affiliation(s)
- Regina Hoo
- Wellcome Sanger Institute, Cambridge, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | | | - Iva Kelava
- Wellcome Sanger Institute, Cambridge, UK
| | - Mukul Rawat
- Wellcome Sanger Institute, Cambridge, UK; Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, University of Dundee, Dundee, UK
| | | | | | | | - Sara Chelaghma
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | | | - David Fernandez-Antoran
- Wellcome/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK; Department of Pathology, University of Cambridge, Cambridge, UK
| | - Ross F Waller
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | - Marcus C S Lee
- Wellcome Sanger Institute, Cambridge, UK; Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, University of Dundee, Dundee, UK.
| | - Roser Vento-Tormo
- Wellcome Sanger Institute, Cambridge, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| |
Collapse
|
9
|
Simião GM, Parreira KS, Klein SG, Ferreira FB, Freitas FDS, Silva EFD, Silva NM, Silva MVD, Lima WR. Involvement of Inflammatory Cytokines, Renal NaPi-IIa Cotransporter, and TRAIL Induced-Apoptosis in Experimental Malaria-Associated Acute Kidney Injury. Pathogens 2024; 13:376. [PMID: 38787228 DOI: 10.3390/pathogens13050376] [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: 02/09/2024] [Revised: 04/24/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024] Open
Abstract
The murine model of experimental cerebral malaria (ECM) induced by Plasmodium berghei ANKA was used to investigate the relationship among pro-inflammatory cytokines, alterations in renal function biomarkers, and the induction of the TRAIL apoptosis pathway during malaria-associated acute kidney injury (AKI). Renal function was evaluated through the measurement of plasma creatinine and blood urea nitrogen (BUN). The mRNA expression of several cytokines and NaPi-IIa was quantified. Kidney sections were examined and cytokine levels were assessed using cytometric bead array (CBA) assays. The presence of glomerular IgG deposits and apoptosis-related proteins were investigated using in situ immunofluorescence assays and quantitative real-time PCR, respectively. NaPi-IIa downregulation in the kidneys provided novel insights into the pathogenesis of hypophosphatemia during CM. Histopathological analysis revealed characteristic features of severe malaria-associated nephritis, including glomerular collapse and tubular alterations. Pro-inflammatory cytokines, such as TNF-α, IL-1β, and IL-6, were upregulated. The TRAIL apoptosis pathway was significantly activated, implicating its role in renal apoptosis. The observed alterations in renal biomarkers and the downregulation of NaPi-IIa shed light on potential mechanisms contributing to renal dysfunction in ECM. The intricate balance between pro- and anti-inflammatory cytokines, along with the activation of the TRAIL apoptosis pathway, highlights the complexity of malaria-associated AKI and provides new therapeutic targets.
Collapse
Affiliation(s)
- Gustavo Martins Simião
- Faculty of Health Sciences, Federal University of Rondonopolis, Rondonópolis 78736-900, MT, Brazil
| | | | - Sandra Gabriela Klein
- Laboratory of Biotechnology in Experimental Models, Federal University of Uberlandia, Uberlândia 38410-337, MG, Brazil
| | - Flávia Batista Ferreira
- Laboratory of Biotechnology in Experimental Models, Federal University of Uberlandia, Uberlândia 38410-337, MG, Brazil
- Institute of Biomedical Sciences, Federal University of Uberlandia, Uberlândia 38405-318, MG, Brazil
| | | | | | - Neide Maria Silva
- Institute of Biomedical Sciences, Federal University of Uberlandia, Uberlândia 38405-318, MG, Brazil
| | - Murilo Vieira da Silva
- Laboratory of Biotechnology in Experimental Models, Federal University of Uberlandia, Uberlândia 38410-337, MG, Brazil
| | - Wânia Rezende Lima
- Faculty of Health Sciences, Federal University of Rondonopolis, Rondonópolis 78736-900, MT, Brazil
- Institute of Biotechnology, Federal University of Catalao, Catalão 75706-881, GO, Brazil
- Laboratory of Biotechnology in Experimental Models, Federal University of Uberlandia, Uberlândia 38410-337, MG, Brazil
| |
Collapse
|
10
|
Ngotho P, Press KD, Peedell M, Muasya W, Omondi BR, Otoboh SE, Seydel KB, Kapulu M, Laufer M, Taylor T, Bousema T, Marti M. Reversible host cell surface remodelling limits immune recognition and maximizes transmission of Plasmodium falciparum gametocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.30.591837. [PMID: 38746342 PMCID: PMC11092622 DOI: 10.1101/2024.04.30.591837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Reducing malaria transmission has been a major pillar of control programmes and is considered crucial for achieving malaria elimination. Gametocytes, the transmissible forms of the P. falciparum parasite, arise during the blood stage of the parasite and develop through 5 morphologically distinct stages. Immature gametocytes (stage I-IV) sequester and develop in the extravascular niche of the bone marrow and possibly spleen. Only mature stage V gametocytes re-enter peripheral circulation to be taken up by mosquitoes for successful onward transmission. We have recently shown that immature, but not mature gametocytes are targets of host immune responses and identified putative target surface antigens. We hypothesize that these antigens play a role in gametocyte sequestration and contribute to acquired transmission-reducing immunity. Here we demonstrate that surface antigen expression, serum reactivity by human IgG, and opsonic phagocytosis by macrophages all show similar dynamics during gametocyte maturation, i.e., on in immature and off in mature gametocytes. Moreover, the switch in surface reactivity coincides with reversal in phosphatidylserine (PS) surface exposure, a marker for red blood cell age and clearance. PS is exposed on the surface of immature gametocytes (as well as in late asexual stages) but is removed from the surface in later gametocyte stages (IV-V). Using parasite reverse genetics and drug perturbations, we confirm that parasite protein export into the host cell and phospholipid scramblase activity are required for the observed surface modifications in asexual and sexual P. falciparum stages. These findings suggest that the dynamic surface remodelling allows (i) immature gametocyte sequestration in bone marrow and (ii) mature gametocyte release into peripheral circulation and immune evasion, therefore contributing to mature gametocyte survival in vivo and onward transmission to mosquitoes. Importantly, blocking scramblase activity during gametocyte maturation results in efficient clearance of mature gametocytes, revealing a potential path for transmission blocking interventions. Our studies have important implications for our understanding of parasite biology and form a starting point for novel intervention strategies to simultaneously reduce parasite burden and transmission.
Collapse
Affiliation(s)
- Priscilla Ngotho
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, United Kingdom
- Institute of Parasitology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | | | - Megan Peedell
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, United Kingdom
- Institute of Parasitology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - William Muasya
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, United Kingdom
| | - Brian Roy Omondi
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Stanley E. Otoboh
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Karl B. Seydel
- Department of Osteopathic Medical Specialties, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan, United States
- Blantyre Malaria Project, Kamuzu University of Health Sciences, Blantyre, Malawi
| | | | - Miriam Laufer
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine Baltimore, MD, United States
| | - Terrie Taylor
- Department of Osteopathic Medical Specialties, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan, United States
- Blantyre Malaria Project, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Teun Bousema
- Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Matthias Marti
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, United Kingdom
- Institute of Parasitology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
11
|
Fréville A, Ressurreição M, van Ooij C. Identification of a non-exported Plasmepsin V substrate that functions in the parasitophorous vacuole of malaria parasites. mBio 2024; 15:e0122323. [PMID: 38078758 PMCID: PMC10790765 DOI: 10.1128/mbio.01223-23] [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: 05/12/2023] [Accepted: 10/26/2023] [Indexed: 01/17/2024] Open
Abstract
IMPORTANCE In the manuscript, the authors investigate the role of the protease Plasmepsin V in the parasite-host interaction. Whereas processing by Plasmepsin V was previously thought to target a protein for export into the host cell, the authors now show that there are proteins cleaved by this protease that are not exported but instead function at the host-parasite interface. This changes the view of this protease, which turns out to have a much broader role than anticipated. The result shows that the protease may have a function much more similar to that of related organisms. The authors also investigate the requirements for protein export by analyzing exported and non-exported proteins and find commonalities between the proteins of each set that further our understanding of the requirements for protein export.
Collapse
Affiliation(s)
- Aline Fréville
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Margarida Ressurreição
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Christiaan van Ooij
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| |
Collapse
|
12
|
Dallmann J, Freitag J, Jung C, Khinvasara K, Merz L, Peters D, Schork M, Beck J. CIMT 2023: report on the 20th Annual Meeting of the Association for Cancer Immunotherapy. IMMUNO-ONCOLOGY TECHNOLOGY 2023; 20:100397. [PMID: 37876518 PMCID: PMC10590812 DOI: 10.1016/j.iotech.2023.100397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
The Association for Cancer Immunotherapy (CIMT) celebrated the 20th anniversary of the CIMT Annual Meeting. CIMT2023 was held 3-5 May 2023 in Mainz, Germany. 1051 academic and clinical professionals from over 30 countries attended the meeting and discussed the latest advances in cancer immunology and immunotherapy research. This report summarizes the highlights of CIMT2023.
Collapse
Affiliation(s)
- J. Dallmann
- Immunotherapies & Preclinical Research, BioNTech SE, Mainz
| | - J. Freitag
- TRON-Translational Oncology, University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - C. Jung
- BioNTech Cell & Gene Therapies GmbH, Mainz
| | - K. Khinvasara
- TRON-Translational Oncology, University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - L. Merz
- TRON-Translational Oncology, University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - D. Peters
- Immunotherapies & Preclinical Research, BioNTech SE, Mainz
| | - M. Schork
- TRON-Translational Oncology, University Medical Center of the Johannes Gutenberg University Mainz gGmbH, Mainz, Germany
| | - J.D. Beck
- Immunotherapies & Preclinical Research, BioNTech SE, Mainz
| |
Collapse
|
13
|
Vallintine T, van Ooij C. Distribution of malaria parasite-derived phosphatidylcholine in the infected erythrocyte. mSphere 2023; 8:e0013123. [PMID: 37606582 PMCID: PMC10597409 DOI: 10.1128/msphere.00131-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 07/05/2023] [Indexed: 08/23/2023] Open
Abstract
Malaria parasites modify their host erythrocyte in multiple ways, leading to changes in the deformability, adhesiveness, and permeability of the host erythrocyte. Most of these changes are mediated by proteins exported from the parasite to the host erythrocyte, where these proteins interact with the host cell cytoskeleton or form complexes in the plasma membrane of the infected erythrocyte. In addition, malaria parasites induce the formation of membranous compartments-the parasitophorous vacuole, the tubovesicular network (TVN), the Maurer's clefts and small vesicles-within the infected erythrocyte, a cell that is normally devoid of internal membranes. After infection, changes also occur in the composition and asymmetry of the erythrocyte plasma membrane. Although many aspects of the mechanism of export of parasite proteins have become clear, the mechanism by which these membranous compartments are formed and expanded is almost entirely unknown. To determine whether parasite-derived phospholipids play a part in these processes, we applied a metabolic labeling technique that allows phosphatidylcholine to be labeled with a fluorophore. As the host erythrocyte cannot synthesize phospholipids, within infected erythrocytes, only parasite-derived phosphatidylcholine will be labeled with this technique. The results revealed that phosphatidylcholine produced by the parasite is distributed throughout the infected erythrocyte, including the TVN and the erythrocyte plasma membrane, but not Maurer's clefts. Interestingly, labeled phospholipids were also detected in the erythrocyte plasma membrane very soon after invasion of the parasites, indicating that the parasite may add phospholipids to the host erythrocyte during invasion. IMPORTANCE Here, we describe a previously unappreciated way in which the malaria parasite interacts with the host erythrocyte, namely, by the transfer of parasite phospholipids to the erythrocyte plasma membrane. This likely has important consequences for the survival of the parasite in the host cell and the host organism. We show that parasite-derived phospholipids are transferred from the parasite to the host erythrocyte plasma membrane and that other internal membranes that are produced after the parasite has invaded the cell are produced, at least in part, using parasite-derived phospholipids. The one exception to this is the Maurer's cleft, a membranous organelle that is involved in the transport of parasite proteins to the surface of the erythrocyte. This reveals that the Maurer's cleft is produced in a different manner than the other parasite-induced membranes. Overall, these findings provide a platform for the study of a new aspect of the host-parasite interaction.
Collapse
Affiliation(s)
- Tansy Vallintine
- Department of Infection Biology, Faculty of Infectious Disease, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Christiaan van Ooij
- Department of Infection Biology, Faculty of Infectious Disease, London School of Hygiene & Tropical Medicine, London, United Kingdom
| |
Collapse
|
14
|
Anaguano D, Dedkhad W, Brooks CF, Cobb DW, Muralidharan V. Time-resolved proximity biotinylation implicates a porin protein in export of transmembrane malaria parasite effectors. J Cell Sci 2023; 136:jcs260506. [PMID: 37772444 PMCID: PMC10651097 DOI: 10.1242/jcs.260506] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/21/2023] [Indexed: 09/30/2023] Open
Abstract
The malaria-causing parasite, Plasmodium falciparum completely remodels its host red blood cell (RBC) through the export of several hundred parasite proteins, including transmembrane proteins, across multiple membranes to the RBC. However, the process by which these exported membrane proteins are extracted from the parasite plasma membrane for export remains unknown. To address this question, we fused the exported membrane protein, skeleton binding protein 1 (SBP1), with TurboID, a rapid, efficient and promiscuous biotin ligase (SBP1TbID). Using time-resolved proximity biotinylation and label-free quantitative proteomics, we identified two groups of SBP1TbID interactors - early interactors (pre-export) and late interactors (post-export). Notably, two promising membrane-associated proteins were identified as pre-export interactors, one of which possesses a predicted translocon domain, that could facilitate the export of membrane proteins. Further investigation using conditional mutants of these candidate proteins showed that these proteins were essential for asexual growth and localize to the host-parasite interface during early stages of the intraerythrocytic cycle. These data suggest that they might play a role in ushering membrane proteins from the parasite plasma membrane for export to the host RBC.
Collapse
Affiliation(s)
- David Anaguano
- Department of Cellular Biology, University of Georgia, Athens, GA, USA
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602, USA
| | - Watcharatip Dedkhad
- Department of Cellular Biology, University of Georgia, Athens, GA, USA
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602, USA
| | - Carrie F. Brooks
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602, USA
| | - David W. Cobb
- Department of Cellular Biology, University of Georgia, Athens, GA, USA
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602, USA
| | - Vasant Muralidharan
- Department of Cellular Biology, University of Georgia, Athens, GA, USA
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
15
|
Huang L, Jasim I, Alkorjia O, Agca C, Oksman A, Agca Y, Goldberg DE, Benson JD, Almasri M. An impedance based microfluidic sensor for evaluation of individual red blood cell solute permeability. Anal Chim Acta 2023; 1267:341226. [PMID: 37257960 DOI: 10.1016/j.aca.2023.341226] [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: 12/03/2022] [Revised: 04/10/2023] [Accepted: 04/17/2023] [Indexed: 06/02/2023]
Abstract
-In this paper, we investigate a microfluidic based sensing device for cell membrane permeability measurements in real time with applications in rapid assessment of red blood cell (RBC) quality at the individual cell level. The microfluidic chip was designed with unique abilities to line up the RBCs in the centerline of the microchannel using positive dielectrophoresis (p-DEP) forces, rapid mixing of RBCs with various media (e.g. containing permeating or nonpermeating solutes) injected from different inlets to achieve high mixing efficiency. The chip detects the impedance values of the RBCs within 0.19 s from the start of mixing with other media, at ten electrodes along the length of the channel and enables time series measurements of volume change of individual cell caused by cell osmosis in anisosmotic fluids over a 0.8 s postmixing timespan. This technique enables estimating water permeability of individual cell accurately. Here we first present confirmation of a linear voltage-diameter relationship in polystyrene bead standards. Next, we show that under equilibrium conditions, the voltage-volume relationship in rat red blood cells (RBCs) is linear, corresponding to previously published Boyle van 't Hoff plots. Using rat cells as a model for human, we present the first measurement of water permeability in individual red blood cells and confirm that these data align with previously published population level values for human RBC. Finally, we present preliminary evidence for possible application of our device to identify individual RBCs infected with Plasmodium falciparum malaria parasites. Future developments using this device will address the use of whole blood with non-homogenous cell populations, a task currently performed by clinical Coulter counters.
Collapse
Affiliation(s)
- Lining Huang
- Department of Electrical Engineering and Computer Science, University of Missouri-Columbia, MO, USA
| | - Ibrahim Jasim
- Department of Electrical Engineering and Computer Science, University of Missouri-Columbia, MO, USA
| | - Omar Alkorjia
- Department of Electrical Engineering and Computer Science, University of Missouri-Columbia, MO, USA
| | - Cansu Agca
- Department of Veterinary Pathology, University of Missouri-Columbia, MO, USA
| | - Anna Oksman
- Department of Medicine, Division of Infectious Diseases, and Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO, 63130, USA
| | - Yuksel Agca
- Department of Veterinary Pathology, University of Missouri-Columbia, MO, USA
| | - Daniel E Goldberg
- Department of Medicine, Division of Infectious Diseases, and Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO, 63130, USA
| | - James D Benson
- Department of Biology, University of Saskatchewan, Saskatoon, SK, Canada.
| | - Mahmoud Almasri
- Department of Electrical Engineering and Computer Science, University of Missouri-Columbia, MO, USA.
| |
Collapse
|
16
|
Jonsdottir TK, Elsworth B, Cobbold S, Gabriela M, Ploeger E, Parkyn Schneider M, Charnaud SC, Dans MG, McConville M, Bullen HE, Crabb BS, Gilson PR. PTEX helps efficiently traffic haemoglobinases to the food vacuole in Plasmodium falciparum. PLoS Pathog 2023; 19:e1011006. [PMID: 37523385 PMCID: PMC10414648 DOI: 10.1371/journal.ppat.1011006] [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: 11/14/2022] [Revised: 08/10/2023] [Accepted: 07/16/2023] [Indexed: 08/02/2023] Open
Abstract
A key element of Plasmodium biology and pathogenesis is the trafficking of ~10% of the parasite proteome into the host red blood cell (RBC) it infects. To cross the parasite-encasing parasitophorous vacuole membrane, exported proteins utilise a channel-forming protein complex termed the Plasmodium translocon of exported proteins (PTEX). PTEX is obligatory for parasite survival, both in vitro and in vivo, suggesting that at least some exported proteins have essential metabolic functions. However, to date only one essential PTEX-dependent process, the new permeability pathways, has been described. To identify other essential PTEX-dependant proteins/processes, we conditionally knocked down the expression of one of its core components, PTEX150, and examined which pathways were affected. Surprisingly, the food vacuole mediated process of haemoglobin (Hb) digestion was substantially perturbed by PTEX150 knockdown. Using a range of transgenic parasite lines and approaches, we show that two major Hb proteases; falcipain 2a and plasmepsin II, interact with PTEX core components, implicating the translocon in the trafficking of Hb proteases. We propose a model where these proteases are translocated into the PV via PTEX in order to reach the cytostome, located at the parasite periphery, prior to food vacuole entry. This work offers a second mechanistic explanation for why PTEX function is essential for growth of the parasite within its host RBC.
Collapse
Affiliation(s)
- Thorey K. Jonsdottir
- Malaria Virulence and Drug Discovery Group, Burnet Institute, Melbourne, Australia
- Department of Immunology and Microbiology, University of Melbourne, Melbourne, Australia
| | - Brendan Elsworth
- Malaria Virulence and Drug Discovery Group, Burnet Institute, Melbourne, Australia
| | - Simon Cobbold
- Department of Biochemistry and Molecular Biology, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Melbourne, Australia
| | - Mikha Gabriela
- Malaria Virulence and Drug Discovery Group, Burnet Institute, Melbourne, Australia
- School of Medicine, Deakin University, Geelong, Australia
| | - Ellen Ploeger
- Malaria Virulence and Drug Discovery Group, Burnet Institute, Melbourne, Australia
| | | | - Sarah C. Charnaud
- Malaria Virulence and Drug Discovery Group, Burnet Institute, Melbourne, Australia
| | - Madeline G. Dans
- Malaria Virulence and Drug Discovery Group, Burnet Institute, Melbourne, Australia
| | - Malcolm McConville
- Department of Biochemistry and Molecular Biology, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Melbourne, Australia
| | - Hayley E. Bullen
- Malaria Virulence and Drug Discovery Group, Burnet Institute, Melbourne, Australia
- Department of Immunology and Microbiology, University of Melbourne, Melbourne, Australia
| | - Brendan S. Crabb
- Malaria Virulence and Drug Discovery Group, Burnet Institute, Melbourne, Australia
- Department of Immunology and Microbiology, University of Melbourne, Melbourne, Australia
- Department of Immunology and Pathology, Monash University, Melbourne, Australia
| | - Paul R. Gilson
- Malaria Virulence and Drug Discovery Group, Burnet Institute, Melbourne, Australia
- Department of Immunology and Microbiology, University of Melbourne, Melbourne, Australia
| |
Collapse
|
17
|
Abstract
Malaria remains a significant threat to global health, and despite concerted efforts to curb the disease, malaria-related morbidity and mortality increased in recent years. Malaria is caused by unicellular eukaryotes of the genus Plasmodium, and all clinical manifestations occur during asexual proliferation of the parasite inside host erythrocytes. In the blood stage, Plasmodium proliferates through an unusual cell cycle mode called schizogony. Contrary to most studied eukaryotes, which divide by binary fission, the parasite undergoes several rounds of DNA replication and nuclear division that are not directly followed by cytokinesis, resulting in multinucleated cells. Moreover, despite sharing a common cytoplasm, these nuclei multiply asynchronously. Schizogony challenges our current models of cell cycle regulation and, at the same time, offers targets for therapeutic interventions. Over the recent years, the adaptation of advanced molecular and cell biological techniques have given us deeper insight how DNA replication, nuclear division, and cytokinesis are coordinated. Here, we review our current understanding of the chronological events that characterize the unusual cell division cycle of P. falciparum in the clinically relevant blood stage of infection.
Collapse
|
18
|
The enemy within: lipid asymmetry in intracellular parasite-host interactions. Emerg Top Life Sci 2023; 7:67-79. [PMID: 36820809 DOI: 10.1042/etls20220089] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/26/2023] [Accepted: 02/07/2023] [Indexed: 02/24/2023]
Abstract
Eukaryotic pathogens with an intracellular parasitic lifestyle are shielded from extracellular threats during replication and growth. In addition to many nutrients, parasites scavenge host cell lipids to establish complex membrane structures inside their host cells. To counteract the disturbance of the host cell plasma membrane they have evolved strategies to regulate phospholipid asymmetry. In this review, the function and importance of lipid asymmetry in the interactions of intracellular protozoan parasites with the target and immune cells of the host are highlighted. The malaria parasite Plasmodium infects red blood cells and extensively refurbishes these terminally differentiated cells. Cholesterol depletion and an altered intracellular calcium ion homeostasis can lead to disruption in erythrocyte membrane asymmetry and increased exposure of phosphatidylserine (PS). Binding to the PS receptor on monocytes and macrophages results in phagocytosis and destruction of infected erythrocytes. Leishmania parasites display apoptotic mimicry by actively enhancing PS exposure on their surface to trigger increased infection of macrophages. In extracellular Toxoplasma gondii a P4-type ATPase/CDC50 co-chaperone pair functions as a flippase important for exocytosis of specialised secretory organelles. Identification and functional analysis of parasite lipid-translocating proteins, i.e. flippases, floppases, and scramblases, will be central for the recognition of the molecular mechanisms of parasite/host interactions. Ultimately, a better understanding of parasitic diseases, host immunity, and immune escape by parasites require more research on the dynamics of phospholipid bilayers of parasites and the infected host cell.
Collapse
|
19
|
Ong JJY, Oh J, Yong Ang X, Naidu R, Chu TTT, Hyoung Im J, Manzoor U, Kha Nguyen T, Na SW, Han ET, Davis C, Sun Park W, Chun W, Jun H, Jin Lee S, Na S, Chan JKY, Park Y, Russell B, Chandramohanadas R, Han JH. Optical diffraction tomography and image reconstruction to measure host cell alterations caused by divergent Plasmodium species. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 286:122026. [PMID: 36395614 DOI: 10.1016/j.saa.2022.122026] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/29/2022] [Accepted: 10/18/2022] [Indexed: 06/16/2023]
Abstract
Malaria is a life-threatening infectious disease caused by parasites of the genus Plasmodium. Understanding the biological features of various parasite forms is important for the optical diagnosis and defining pathological states, which are often constrained by the lack of ambient visualization approaches. Here, we employ a label-free tomographic technique to visualize the host red blood cell (RBC) remodeling process and quantify changes in biochemical properties arising from parasitization. Through this, we provide a quantitative body of information pertaining to the influence of host cell environment on growth, survival, and replication of P. falciparum and P. vivax in their respective host cells: mature erythrocytes and young reticulocytes. These exquisite three-dimensional measurements of infected red cells demonstrats the potential of evolving 3D imaging to advance our understanding of Plasmodium biology and host-parasite interactions.
Collapse
Affiliation(s)
- Jessica J Y Ong
- Department of Microbiology and Immunology, University of Otago, Dunedin 9054, New Zealand
| | - Jeonghun Oh
- Department of Physics, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea; KAIST Institute for Health Science and Technology, KAIST, Daejeon 34141, Republic of Korea
| | - Xiang Yong Ang
- Department of Microbiology and Immunology, National University of Singapore, Singapore
| | - Renugah Naidu
- Pillar of Engineering Product Development, Singapore University of Technology and Design, Singapore, Singapore
| | - Trang T T Chu
- Pillar of Engineering Product Development, Singapore University of Technology and Design, Singapore, Singapore
| | - Jae Hyoung Im
- Department of Infectious Disease, Inha University School of Medicine, Incheon 22212, Republic of Korea
| | - Umar Manzoor
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Tuyet Kha Nguyen
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Seok-Won Na
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Christeen Davis
- DBT Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India; Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Won Sun Park
- Department of Physiology, School of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Wanjoo Chun
- Department of Pharmacology, School of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Hojong Jun
- Department of Tropical Medicine, Inha University College of Medicine, Incheon 22212, Republic of Korea
| | - Se Jin Lee
- Department of Obstetrics and Gynecology, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon 24341, South Korea
| | - Sunghun Na
- Department of Obstetrics and Gynecology, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon 24341, South Korea
| | - Jerry K Y Chan
- KK Womens' and Childrens' Hospital, Singapore; Academic Clinical Program in Obstetrics and Gynaecology, Duke-NUS Medical School, 169857, Singapore
| | - YongKeun Park
- Department of Physics, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea; KAIST Institute for Health Science and Technology, KAIST, Daejeon 34141, Republic of Korea; Tomocube Inc, Daejeon 34109, Republic of Korea
| | - Bruce Russell
- Department of Microbiology and Immunology, University of Otago, Dunedin 9054, New Zealand
| | - Rajesh Chandramohanadas
- Department of Microbiology and Immunology, National University of Singapore, Singapore; Pillar of Engineering Product Development, Singapore University of Technology and Design, Singapore, Singapore; DBT Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.
| | - Jin-Hee Han
- Department of Microbiology and Immunology, University of Otago, Dunedin 9054, New Zealand; Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea.
| |
Collapse
|
20
|
CD36-A Host Receptor Necessary for Malaria Parasites to Establish and Maintain Infection. Microorganisms 2022; 10:microorganisms10122356. [PMID: 36557610 PMCID: PMC9785914 DOI: 10.3390/microorganisms10122356] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/21/2022] [Accepted: 11/27/2022] [Indexed: 11/30/2022] Open
Abstract
Plasmodium falciparum-infected erythrocytes (PfIEs) present P. falciparum erythrocyte membrane protein 1 proteins (PfEMP1s) on the cell surface, via which they cytoadhere to various endothelial cell receptors (ECRs) on the walls of human blood vessels. This prevents the parasite from passing through the spleen, which would lead to its elimination. Each P. falciparum isolate has about 60 different PfEMP1s acting as ligands, and at least 24 ECRs have been identified as interaction partners. Interestingly, in every parasite genome sequenced to date, at least 75% of the encoded PfEMP1s have a binding domain for the scavenger receptor CD36 widely distributed on host endothelial cells and many other cell types. Here, we discuss why the interaction between PfIEs and CD36 is optimal to maintain a finely regulated equilibrium that allows the parasite to multiply and spread while causing minimal harm to the host in most infections.
Collapse
|
21
|
Hakimi H, Yamagishi J, Kawazu SI, Asada M. Advances in understanding red blood cell modifications by Babesia. PLoS Pathog 2022; 18:e1010770. [PMID: 36107982 PMCID: PMC9477259 DOI: 10.1371/journal.ppat.1010770] [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] [Indexed: 11/18/2022] Open
Abstract
Babesia are tick-borne protozoan parasites that can infect livestock, pets, wildlife animals, and humans. In the mammalian host, they invade and multiply within red blood cells (RBCs). To support their development as obligate intracellular parasites, Babesia export numerous proteins to modify the RBC during invasion and development. Such exported proteins are likely important for parasite survival and pathogenicity and thus represent candidate drug or vaccine targets. The availability of complete genome sequences and the establishment of transfection systems for several Babesia species have aided the identification and functional characterization of exported proteins. Here, we review exported Babesia proteins; discuss their functions in the context of immune evasion, cytoadhesion, and nutrient uptake; and highlight possible future topics for research and application in this field.
Collapse
Affiliation(s)
- Hassan Hakimi
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, Texas, United States of America
- * E-mail: (HH); (MA)
| | - Junya Yamagishi
- Division of Collaboration and Education, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Shin-ichiro Kawazu
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Masahito Asada
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
- * E-mail: (HH); (MA)
| |
Collapse
|
22
|
Daniyan MO, Fisusi FA, Adeoye OB. Neurotransmitters and molecular chaperones interactions in cerebral malaria: Is there a missing link? Front Mol Biosci 2022; 9:965569. [PMID: 36090033 PMCID: PMC9451049 DOI: 10.3389/fmolb.2022.965569] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 07/28/2022] [Indexed: 12/02/2022] Open
Abstract
Plasmodium falciparum is responsible for the most severe and deadliest human malaria infection. The most serious complication of this infection is cerebral malaria. Among the proposed hypotheses that seek to explain the manifestation of the neurological syndrome in cerebral malaria is the vascular occlusion/sequestration/mechanic hypothesis, the cytokine storm or inflammatory theory, or a combination of both. Unfortunately, despite the increasing volume of scientific information on cerebral malaria, our understanding of its pathophysiologic mechanism(s) is still very limited. In a bid to maintain its survival and development, P. falciparum exports a large number of proteins into the cytosol of the infected host red blood cell. Prominent among these are the P. falciparum erythrocytes membrane protein 1 (PfEMP1), P. falciparum histidine-rich protein II (PfHRP2), and P. falciparum heat shock proteins 70-x (PfHsp70-x). Functional activities and interaction of these proteins with one another and with recruited host resident proteins are critical factors in the pathology of malaria in general and cerebral malaria in particular. Furthermore, several neurological impairments, including cognitive, behavioral, and motor dysfunctions, are known to be associated with cerebral malaria. Also, the available evidence has implicated glutamate and glutamatergic pathways, coupled with a resultant alteration in serotonin, dopamine, norepinephrine, and histamine production. While seeking to improve our understanding of the pathophysiology of cerebral malaria, this article seeks to explore the possible links between host/parasite chaperones, and neurotransmitters, in relation to other molecular players in the pathology of cerebral malaria, to explore such links in antimalarial drug discovery.
Collapse
Affiliation(s)
- Michael Oluwatoyin Daniyan
- Department of Pharmacology, Faculty of Pharmacy, Obafemi Awolowo University, Ile-Ife, Osun State, Nigeria
| | - Funmilola Adesodun Fisusi
- Drug Research and Production Unit, Faculty of Pharmacy, Obafemi Awolowo University, Ile-Ife, Osun State, Nigeria
| | - Olufunso Bayo Adeoye
- Department of Biochemistry, Benjamin S. Carson (Snr.) College of Medicine, Babcock University, Ilishan-Remo, Ogun State, Nigeria
| |
Collapse
|
23
|
Shakya B, Kilili GK, Wang L, Nakayasu ES, LaCount DJ. Identification of Exported Plasmodium falciparum Proteins That Bind to the Erythrocyte Cytoskeleton. Microorganisms 2022; 10:1438. [PMID: 35889157 PMCID: PMC9320996 DOI: 10.3390/microorganisms10071438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/11/2022] [Accepted: 07/13/2022] [Indexed: 11/28/2022] Open
Abstract
Plasmodium proteins are exported to the erythrocyte cytoplasm to create an environment that supports parasite replication. Although hundreds of proteins are predicted to be exported through Plasmodium export element (PEXEL)-dependent and -independent mechanisms, the functions of exported proteins are largely uncharacterized. In this study, we used a biochemical screening approach to identify putative exported P. falciparum proteins that bound to inside-out vesicles prepared from erythrocytes. Out of 69 P. falciparum PEXEL-motif proteins tested, 18 bound to inside-out vesicles (IOVs) in two or more independent assays. Using co-affinity purifications followed by mass spectrometry, pairwise co-purification experiments, and the split-luciferase assay, we identified 31 putative protein-protein interactions between erythrocyte cytoskeletal proteins and predicted exported P. falciparum proteins. We further showed that PF3D7_1401600 binds to the spectrin-binding domain of erythrocyte ankyrin via its MESA erythrocyte cytoskeleton binding (MEC) motif and to the N-terminal domains of ankyrin and 4.1R through a fragment that required an intact Plasmodium helical interspersed sub-telomeric (PHIST) domain. Introduction of PF3D7_1401600 into erythrocyte ghosts increased retention in the microsphiltration assay, consistent with previous data that reported a reduction of rigidity in red blood cells infected with PF3D7_1401600-deficient parasites.
Collapse
Affiliation(s)
- Bikash Shakya
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA; (B.S.); (G.K.K.); (L.W.)
| | - Geoffrey Kimiti Kilili
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA; (B.S.); (G.K.K.); (L.W.)
| | - Ling Wang
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA; (B.S.); (G.K.K.); (L.W.)
| | - Ernesto S. Nakayasu
- Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907, USA;
- Pacific Northwest National Laboratory, Biological Sciences Division, Richland, WA 99352, USA
| | - Douglas J. LaCount
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA; (B.S.); (G.K.K.); (L.W.)
| |
Collapse
|
24
|
Shao J. Labeling Strategies for Surface-Exposed Protein Visualization and Determination in Plasmodium falciparum Malaria. Front Cell Infect Microbiol 2022; 12:914297. [PMID: 35755836 PMCID: PMC9226428 DOI: 10.3389/fcimb.2022.914297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/11/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Jinfeng Shao
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| |
Collapse
|
25
|
Goodswen SJ, Kennedy PJ, Ellis JT. Compilation of parasitic immunogenic proteins from 30 years of published research using machine learning and natural language processing. Sci Rep 2022; 12:10349. [PMID: 35725870 PMCID: PMC9208253 DOI: 10.1038/s41598-022-13790-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 05/18/2022] [Indexed: 12/02/2022] Open
Abstract
The World Health Organisation reported in 2020 that six of the top 10 sources of death in low-income countries are parasites. Parasites are microorganisms in a relationship with a larger organism, the host. They acquire all benefits at the host’s expense. A disease develops if the parasitic infection disrupts normal functioning of the host. This disruption can range from mild to severe, including death. Humans and livestock continue to be challenged by established and emerging infectious disease threats. Vaccination is the most efficient tool for preventing current and future threats. Immunogenic proteins sourced from the disease-causing parasite are worthwhile vaccine components (subunits) due to reliable safety and manufacturing capacity. Publications with ‘subunit vaccine’ in their title have accumulated to thousands over the last three decades. However, there are possibly thousands more reporting immunogenicity results without mentioning ‘subunit’ and/or ‘vaccine’. The exact number is unclear given the non-standardised keywords in publications. The study aim is to identify parasite proteins that induce a protective response in an animal model as reported in the scientific literature within the last 30 years using machine learning and natural language processing. Source code to fulfil this aim and the vaccine candidate list obtained is made available.
Collapse
Affiliation(s)
- Stephen J Goodswen
- School of Life Sciences, University of Technology Sydney, 15 Broadway, Ultimo, NSW, 2007, Australia
| | - Paul J Kennedy
- School of Computer Science, Faculty of Engineering and Information Technology and the Australian Artificial Intelligence Institute, University of Technology Sydney, 15 Broadway, Ultimo, NSW, 2007, Australia
| | - John T Ellis
- School of Life Sciences, University of Technology Sydney, 15 Broadway, Ultimo, NSW, 2007, Australia.
| |
Collapse
|
26
|
Jäger J, Patra P, Sanchez CP, Lanzer M, Schwarz US. A particle-based computational model to analyse remodelling of the red blood cell cytoskeleton during malaria infections. PLoS Comput Biol 2022; 18:e1009509. [PMID: 35394995 PMCID: PMC9020725 DOI: 10.1371/journal.pcbi.1009509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 04/20/2022] [Accepted: 03/21/2022] [Indexed: 11/18/2022] Open
Abstract
Red blood cells can withstand the harsh mechanical conditions in the vasculature only because the bending rigidity of their plasma membrane is complemented by the shear elasticity of the underlying spectrin-actin network. During an infection by the malaria parasite Plasmodium falciparum, the parasite mines host actin from the junctional complexes and establishes a system of adhesive knobs, whose main structural component is the knob-associated histidine rich protein (KAHRP) secreted by the parasite. Here we aim at a mechanistic understanding of this dramatic transformation process. We have developed a particle-based computational model for the cytoskeleton of red blood cells and simulated it with Brownian dynamics to predict the mechanical changes resulting from actin mining and KAHRP-clustering. Our simulations include the three-dimensional conformations of the semi-flexible spectrin chains, the capping of the actin protofilaments and several established binding sites for KAHRP. For the healthy red blood cell, we find that incorporation of actin protofilaments leads to two regimes in the shear response. Actin mining decreases the shear modulus, but knob formation increases it. We show that dynamical changes in KAHRP binding affinities can explain the experimentally observed relocalization of KAHRP from ankyrin to actin complexes and demonstrate good qualitative agreement with experiments by measuring pair cross-correlations both in the computer simulations and in super-resolution imaging experiments. Malaria is one of the deadliest infectious diseases and its symptoms are related to the blood stage, when the parasite multiplies within red blood cells. In order to avoid clearance by the spleen, the parasite produces specific factors like the adhesion receptor PfEMP1 and the multifunctional protein KAHRP that lead to the formation of adhesive knobs on the surface of the red blood cells and thus increase residence time in the vasculature. We have developed a computational model for the parasite-induced remodelling of the actin-spectrin network to quantitatively predict the dynamical changes in the mechanical properties of the infected red blood cells and the spatial distribution of the different protein components of the membrane skeleton. Our simulations show that KAHRP can relocate to actin junctions due to dynamical changes in binding affinities, in good qualitative agreement with super-resolution imaging experiments. In the future, our simulation framework can be used to gain further mechanistic insight into the way malaria parasites attack the red blood cell cytoskeleton.
Collapse
Affiliation(s)
- Julia Jäger
- Institute for Theoretical Physics, Heidelberg University, Heidelberg, Germany
- BioQuant-Center for Quantitative Biology, Heidelberg University, Heidelberg, Germany
| | - Pintu Patra
- Institute for Theoretical Physics, Heidelberg University, Heidelberg, Germany
- BioQuant-Center for Quantitative Biology, Heidelberg University, Heidelberg, Germany
| | - Cecilia P. Sanchez
- Center of Infectious Diseases, Parasitology, University Hospital Heidelberg, Heidelberg, Germany
| | - Michael Lanzer
- Center of Infectious Diseases, Parasitology, University Hospital Heidelberg, Heidelberg, Germany
- * E-mail: (ML); (USS)
| | - Ulrich S. Schwarz
- Institute for Theoretical Physics, Heidelberg University, Heidelberg, Germany
- BioQuant-Center for Quantitative Biology, Heidelberg University, Heidelberg, Germany
- * E-mail: (ML); (USS)
| |
Collapse
|
27
|
Molecular mechanisms of hematological and biochemical alterations in malaria: A review. Mol Biochem Parasitol 2021; 247:111446. [PMID: 34953384 DOI: 10.1016/j.molbiopara.2021.111446] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/20/2021] [Accepted: 12/19/2021] [Indexed: 11/20/2022]
Abstract
Malaria is a dangerous disease that contributes to millions of hospital visits and hundreds of thousands of deaths, especially in children residing in sub-Saharan Africa. Although several interventions such as vector control, case detection, and treatment are already in place, there is no substantive reduction in the disease burden. Several studies in the past have reported the emergence of resistant strains of malaria parasites (MPs) and mosquitoes, and poor adherence and inaccessibility to effective antimalarial drugs as the major factors for this persistent menace of malaria infections. Moreover, victory against MP infections for many years has been hampered by an incomplete understanding of the complex nature of malaria pathogenesis. Very recent studies have identified different complex interactions and hematological alterations induced by malaria parasites. However, no studies have hybridized these alterations for a better understanding of Malaria pathogenesis. Hence, this review thoroughly discusses the molecular mechanisms of all reported hematological and biochemical alterations induced by MPs infections. Specifically, the mechanisms in which MP-infection induces anemia, thrombocytopenia, leukopenia, dyslipidemia, hypoglycemia, oxidative stress, and liver and kidney malfunctions were presented. The study also discussed how MPs evade the host's immune response and suggested strategies to limit evasion of the host's immune response to combat malaria and its complications.
Collapse
|
28
|
Besednova NN, Zaporozhets TS, Andryukov BG, Kryzhanovsky SP, Ermakova SP, Kuznetsova TA, Voronova AN, Shchelkanov MY. Antiparasitic Effects of Sulfated Polysaccharides from Marine Hydrobionts. Mar Drugs 2021; 19:637. [PMID: 34822508 PMCID: PMC8624348 DOI: 10.3390/md19110637] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/07/2021] [Accepted: 11/10/2021] [Indexed: 12/15/2022] Open
Abstract
This review presents materials characterizing sulfated polysaccharides (SPS) of marine hydrobionts (algae and invertebrates) as potential means for the prevention and treatment of protozoa and helminthiasis. The authors have summarized the literature on the pathogenetic targets of protozoa on the host cells and on the antiparasitic potential of polysaccharides from red, brown and green algae as well as certain marine invertebrates. Information about the mechanisms of action of these unique compounds in diseases caused by protozoa has also been summarized. SPS is distinguished by high antiparasitic activity, good solubility and an almost complete absence of toxicity. In the long term, this allows for the consideration of these compounds as effective and attractive candidates on which to base drugs, biologically active food additives and functional food products with antiparasitic activity.
Collapse
Affiliation(s)
- Natalya N. Besednova
- G.P. Somov Research Institute of Epidemiology and Microbiology, Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, 690087 Vladivostok, Russia; (T.S.Z.); (B.G.A.); (T.A.K.); (A.N.V.); (M.Y.S.)
| | - Tatyana S. Zaporozhets
- G.P. Somov Research Institute of Epidemiology and Microbiology, Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, 690087 Vladivostok, Russia; (T.S.Z.); (B.G.A.); (T.A.K.); (A.N.V.); (M.Y.S.)
| | - Boris G. Andryukov
- G.P. Somov Research Institute of Epidemiology and Microbiology, Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, 690087 Vladivostok, Russia; (T.S.Z.); (B.G.A.); (T.A.K.); (A.N.V.); (M.Y.S.)
- School of Biomedicine, Far Eastern Federal University (FEFU), 690091 Vladivostok, Russia
| | - Sergey P. Kryzhanovsky
- Medical Association of the Far Eastern Branch of the Russian Academy of Sciences, 690022 Vladivostok, Russia;
| | - Svetlana P. Ermakova
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 690022 Vladivostok, Russia;
| | - Tatyana A. Kuznetsova
- G.P. Somov Research Institute of Epidemiology and Microbiology, Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, 690087 Vladivostok, Russia; (T.S.Z.); (B.G.A.); (T.A.K.); (A.N.V.); (M.Y.S.)
| | - Anastasia N. Voronova
- G.P. Somov Research Institute of Epidemiology and Microbiology, Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, 690087 Vladivostok, Russia; (T.S.Z.); (B.G.A.); (T.A.K.); (A.N.V.); (M.Y.S.)
| | - Mikhail Y. Shchelkanov
- G.P. Somov Research Institute of Epidemiology and Microbiology, Federal Service for Surveillance on Consumer Rights Protection and Human Wellbeing, 690087 Vladivostok, Russia; (T.S.Z.); (B.G.A.); (T.A.K.); (A.N.V.); (M.Y.S.)
- School of Biomedicine, Far Eastern Federal University (FEFU), 690091 Vladivostok, Russia
- National Scientific Center of Marine Biology, Far Eastern Branch of the Russian Academy of Sciences, 690041 Vladivostok, Russia
- Federal Scientific Center of the East Asia Terrestrial Biodiversity, Far Eastern Branch of the Russian Academy of Sciences, 690022 Vladivostok, Russia
| |
Collapse
|
29
|
Mandala WL, Harawa V, Dzinjalamala F, Tembo D. The role of different components of the immune system against Plasmodium falciparum malaria: Possible contribution towards malaria vaccine development. Mol Biochem Parasitol 2021; 246:111425. [PMID: 34666102 PMCID: PMC8655617 DOI: 10.1016/j.molbiopara.2021.111425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 08/10/2021] [Accepted: 10/08/2021] [Indexed: 12/24/2022]
Abstract
Plasmodium falciparum malaria still remains a major global public health challenge with over 220 million new cases and well over 400,000 deaths annually. Most of the deaths occur in sub-Saharan Africa which bears 90 % of the malaria cases. Such high P. falciparum malaria-related morbidity and mortality rates pose a huge burden on the health and economic wellbeing of the countries affected. Lately, substantial gains have been made in reducing malaria morbidity and mortality through intense malaria control initiatives such as use of effective antimalarials, intensive distribution and use of insecticide-treated nets (ITNs), and implementation of massive indoor residual spraying (IRS) campaigns. However, these gains are being threatened by widespread resistance of the parasite to antimalarials, and the vector to insecticides. Over the years the use of vaccines has proven to be the most reliable, cost-effective and efficient method for controlling the burden and spread of many infectious diseases, especially in resource poor settings with limited public health infrastructure. Nonetheless, this had not been the case with malaria until the most promising malaria vaccine candidate, RTS,S/AS01, was approved for pilot implementation programme in three African countries in 2015. This was regarded as the most important breakthrough in the fight against malaria. However, RTS,S/AS01 has been found to have some limitations, the main ones being low efficacy in certain age groups, poor immunogenicity and need for almost three boosters to attain a reasonable efficacy. Thus, the search for a more robust and effective malaria vaccine still continues and a better understanding of naturally acquired immune responses to the various stages, including the transmissible stages of the parasite, could be crucial in rational vaccine design. This review therefore compiles what is currently known about the basic biology of P. falciparum and the natural malaria immune response against malaria and progress made towards vaccine development.
Collapse
Affiliation(s)
- Wilson L Mandala
- Academy of Medical Sciences, Malawi University of Science and Technology, Thyolo, Malawi; Malawi Liverpool Wellcome Trust, Blantyre, Malawi.
| | | | - Fraction Dzinjalamala
- Academy of Medical Sciences, Malawi University of Science and Technology, Thyolo, Malawi
| | | |
Collapse
|
30
|
Co-chaperone involvement in knob biogenesis implicates host-derived chaperones in malaria virulence. PLoS Pathog 2021; 17:e1009969. [PMID: 34614006 PMCID: PMC8544838 DOI: 10.1371/journal.ppat.1009969] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 10/25/2021] [Accepted: 09/24/2021] [Indexed: 11/19/2022] Open
Abstract
The pathology associated with malaria infection is largely due to the ability of infected human RBCs to adhere to a number of receptors on endothelial cells within tissues and organs. This phenomenon is driven by the export of parasite-encoded proteins to the host cell, the exact function of many of which is still unknown. Here we inactivate the function of one of these exported proteins, PFA66, a member of the J-domain protein family. Although parasites lacking this protein were still able to grow in cell culture, we observed severe defects in normal host cell modification, including aberrant morphology of surface knobs, disrupted presentation of the cytoadherence molecule PfEMP1, and a total lack of cytoadherence, despite the presence of the knob associated protein KAHRP. Complementation assays demonstrate that an intact J-domain is required for recovery to a wild-type phenotype and suggest that PFA66 functions in concert with a HSP70 to carry out host cell modification. Strikingly, this HSP70 is likely to be of host origin. ATPase assays on recombinant protein verify a functional interaction between PFA66 and residual host cell HSP70. Taken together, our data reveal a role for PFA66 in host cell modification, strongly implicate human HSP70s as being essential in this process and uncover a new KAHRP-independent molecular factor required for correct knob biogenesis. To survive in the human body, the malaria parasite invades and lives within human red blood cells. Once within the red blood cell, the parasite renovates the host cell to its own needs. Here we have studied which factors from both parasite and host cell are required for this renovation process, and discover that human chaperone proteins, referred to as HSP70, are required. It appears that a particular parasite-derived protein, PFA66, recruits and modifies the function of the human HSP70. As this interaction between a parasite and human protein is novel and essential for parasite survival, our study identifies a potential Achilles’ Heel which may be targeted for development of new anti-malaria therapies.
Collapse
|
31
|
Daniyan MO. Heat Shock Proteins as Targets for Novel Antimalarial Drug Discovery. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1340:205-236. [PMID: 34569027 DOI: 10.1007/978-3-030-78397-6_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Plasmodium falciparum, the parasitic agent that is responsible for a severe and dangerous form of human malaria, has a history of long years of cohabitation with human beings with attendant negative consequences. While there have been some gains in the fight against malaria through the application of various control measures and the use of chemotherapeutic agents, and despite the global decline in malaria cases and associated deaths, the continual search for new and effective therapeutic agents is key to achieving sustainable development goals. An important parasite survival strategy, which is also of serious concern to the scientific community, is the rate at which the parasites continually develop resistance to drugs. Among the key players in the parasite's ability to develop resistance, maintain cellular integrity, and survives within an unusual environment of the red blood cells are the molecular chaperones of the heat shock proteins (HSP) family. HSPs constitute a novel avenue for antimalarial drug discovery and by exploring their ubiquitous nature and multifunctional activities, they may be suitable targets for the discovery of multi-targets antimalarial drugs, needed to fight incessant drug resistance. In this chapter, features of selected families of plasmodial HSPs that can be exploited in drug discovery are presented. Also, known applications of HSPs in small molecule screening, their potential usefulness in high throughput drug screening, as well as possible challenges are highlighted.
Collapse
Affiliation(s)
- Michael Oluwatoyin Daniyan
- Department of Pharmacology, Faculty of Pharmacy, Obafemi Awolowo University, Ile-Ife, Osun State, Nigeria.
| |
Collapse
|
32
|
Introductory Chapter: The Importance of Heat Shock Proteins in Survival and Pathogenesis of the Malaria Parasite Plasmodium falciparum. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021. [PMID: 34569019 DOI: 10.1007/978-3-030-78397-6_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/31/2023]
Abstract
Malaria did not die with the end of the age of western colonization but is still a major public health issue in large parts of the world. Despite repeated and concerted efforts to eradicate this disease, it has proved remarkably resilient, and constant vigilance and continuous research are required to discover new chinks in the parasite's armor and alleviate the suffering at both the individual and societal levels. A deeper understanding of the fundamental processes underlying parasite survival, propagation, virulence, and ability to cause disease is the key to the development of desperately needed new therapies and prophylactic drugs. Malaria parasites, by the nature of their lifecycle, are subject to a number of environmental and cellular stresses which they must overcome to survive. To this end, they express a number of heat shock proteins (HSPs), molecules specialized on buffering the effects of external stimuli, but which are also essential for normal cellular biochemistry. In this introductory chapter, I give a brief overview of the diversity of structure, function, and importance of these HSPs, and highlight some of the current and future research questions in this field. Additionally, this chapter acts as a bridge to the other chapters in this book. These chapters, I think you will agree, demonstrate that with regard to HSPs malaria parasites, as in so many things, obey the adage "Same same, but different."
Collapse
|
33
|
Protein Sorting in Plasmodium Falciparum. Life (Basel) 2021; 11:life11090937. [PMID: 34575086 PMCID: PMC8467625 DOI: 10.3390/life11090937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/04/2021] [Accepted: 09/04/2021] [Indexed: 11/23/2022] Open
Abstract
Plasmodium falciparum is a unicellular eukaryote with a very polarized secretory system composed of micronemes rhoptries and dense granules that are required for host cell invasion. P. falciparum, like its relative T. gondii, uses the endolysosomal system to produce the secretory organelles and to ingest host cell proteins. The parasite also has an apicoplast, a secondary endosymbiotic organelle, which depends on vesicular trafficking for appropriate incorporation of nuclear-encoded proteins into the apicoplast. Recently, the central molecules responsible for sorting and trafficking in P. falciparum and T. gondii have been characterized. From these studies, it is now evident that P. falciparum has repurposed the molecules of the endosomal system to the secretory pathway. Additionally, the sorting and vesicular trafficking mechanism seem to be conserved among apicomplexans. This review described the most recent findings on the molecular mechanisms of protein sorting and vesicular trafficking in P. falciparum and revealed that P. falciparum has an amazing secretory machinery that has been cleverly modified to its intracellular lifestyle.
Collapse
|
34
|
Ali F, Wali H, Jan S, Zia A, Aslam M, Ahmad I, Afridi SG, Shams S, Khan A. Analysing the essential proteins set of Plasmodium falciparum PF3D7 for novel drug targets identification against malaria. Malar J 2021; 20:335. [PMID: 34344361 PMCID: PMC8336052 DOI: 10.1186/s12936-021-03865-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/25/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Plasmodium falciparum is an obligate intracellular parasite of humans that causes malaria. Falciparum malaria is a major public health threat to human life responsible for high mortality. Currently, the risk of multi-drug resistance of P. falciparum is rapidly increasing. There is a need to address new anti-malarial therapeutics strategies to combat the drug-resistance threat. METHODS The P. falciparum essential proteins were retrieved from the recently published studies. These proteins were initially scanned against human host and its gut microbiome proteome sets by comparative proteomics analyses. The human host non-homologs essential proteins of P. falciparum were additionally analysed for druggability potential via in silico methods to possibly identify novel therapeutic targets. Finally, the PfAp4AH target was prioritized for pharmacophore modelling based virtual screening and molecular docking analyses to identify potent inhibitors from drug-like compounds databases. RESULTS The analyses identified six P. falciparum essential and human host non-homolog proteins that follow the key druggability features. These druggable targets have not been catalogued so far in the Drugbank repository. These prioritized proteins seem novel and promising drug targets against P. falciparum due to their key protein-protein interactions features in pathogen-specific biological pathways and to hold appropriate drug-like molecule binding pockets. The pharmacophore features based virtual screening of Pharmit resource predicted a lead compound i.e. MolPort-045-917-542 as a promising inhibitor of PfAp4AH among prioritized targets. CONCLUSION The prioritized protein targets may worthy to test in malarial drug discovery programme to overcome the anti-malarial resistance issues. The in-vitro and in-vivo studies might be promising for additional validation of these prioritized lists of drug targets against malaria.
Collapse
Affiliation(s)
- Fawad Ali
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, 23200, Pakistan.,Department of Biochemistry, Hazara University, Mansehra, 21120, Pakistan
| | - Hira Wali
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, 23200, Pakistan
| | - Saadia Jan
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, 23200, Pakistan
| | - Asad Zia
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, 23200, Pakistan
| | - Muneeba Aslam
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, 23200, Pakistan
| | - Imtiaz Ahmad
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, 23200, Pakistan
| | - Sahib Gul Afridi
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, 23200, Pakistan
| | - Sulaiman Shams
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, 23200, Pakistan
| | - Asifullah Khan
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, 23200, Pakistan.
| |
Collapse
|
35
|
Goodswen SJ, Kennedy PJ, Ellis JT. Predicting Protein Therapeutic Candidates for Bovine Babesiosis Using Secondary Structure Properties and Machine Learning. Front Genet 2021; 12:716132. [PMID: 34367264 PMCID: PMC8343536 DOI: 10.3389/fgene.2021.716132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 06/28/2021] [Indexed: 12/02/2022] Open
Abstract
Bovine babesiosis causes significant annual global economic loss in the beef and dairy cattle industry. It is a disease instigated from infection of red blood cells by haemoprotozoan parasites of the genus Babesia in the phylum Apicomplexa. Principal species are Babesia bovis, Babesia bigemina, and Babesia divergens. There is no subunit vaccine. Potential therapeutic targets against babesiosis include members of the exportome. This study investigates the novel use of protein secondary structure characteristics and machine learning algorithms to predict exportome membership probabilities. The premise of the approach is to detect characteristic differences that can help classify one protein type from another. Structural properties such as a protein’s local conformational classification states, backbone torsion angles ϕ (phi) and ψ (psi), solvent-accessible surface area, contact number, and half-sphere exposure are explored here as potential distinguishing protein characteristics. The presented methods that exploit these structural properties via machine learning are shown to have the capacity to detect exportome from non-exportome Babesia bovis proteins with an 86–92% accuracy (based on 10-fold cross validation and independent testing). These methods are encapsulated in freely available Linux pipelines setup for automated, high-throughput processing. Furthermore, proposed therapeutic candidates for laboratory investigation are provided for B. bovis, B. bigemina, and two other haemoprotozoan species, Babesia canis, and Plasmodium falciparum.
Collapse
Affiliation(s)
- Stephen J Goodswen
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW, Australia
| | - Paul J Kennedy
- School of Computer Science, Faculty of Engineering and Information Technology and the Australian Artificial Intelligence Institute, University of Technology Sydney, Ultimo, NSW, Australia
| | - John T Ellis
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW, Australia
| |
Collapse
|
36
|
Goodswen SJ, Kennedy PJ, Ellis JT. Applying Machine Learning to Predict the Exportome of Bovine and Canine Babesia Species That Cause Babesiosis. Pathogens 2021; 10:660. [PMID: 34071992 PMCID: PMC8226867 DOI: 10.3390/pathogens10060660] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 01/08/2023] Open
Abstract
Babesia infection of red blood cells can cause a severe disease called babesiosis in susceptible hosts. Bovine babesiosis causes global economic loss to the beef and dairy cattle industries, and canine babesiosis is considered a clinically significant disease. Potential therapeutic targets against bovine and canine babesiosis include members of the exportome, i.e., those proteins exported from the parasite into the host red blood cell. We developed three machine learning-derived methods (two novel and one adapted) to predict for every known Babesia bovis, Babesia bigemina, and Babesia canis protein the probability of being an exportome member. Two well-studied apicomplexan-related species, Plasmodium falciparum and Toxoplasma gondii, with extensive experimental evidence on their exportome or excreted/secreted proteins were used as important benchmarks for the three methods. Based on 10-fold cross validation and multiple train-validation-test splits of training data, we expect that over 90% of the predicted probabilities accurately provide a secretory or non-secretory indicator. Only laboratory testing can verify that predicted high exportome membership probabilities are creditable exportome indicators. However, the presented methods at least provide those proteins most worthy of laboratory validation and will ultimately save time and money.
Collapse
Affiliation(s)
- Stephen J. Goodswen
- School of Life Sciences, University of Technology Sydney, 15 Broadway, Ultimo, NSW 2007, Australia;
| | - Paul J. Kennedy
- School of Computer Science, Faculty of Engineering and Information Technology, Australian Artificial Intelligence Institute, University of Technology Sydney, 15 Broadway, Ultimo, NSW 2007, Australia;
| | - John T. Ellis
- School of Life Sciences, University of Technology Sydney, 15 Broadway, Ultimo, NSW 2007, Australia;
| |
Collapse
|
37
|
Soni K, Kempf G, Manalastas-Cantos K, Hendricks A, Flemming D, Guizetti J, Simon B, Frischknecht F, Svergun DI, Wild K, Sinning I. Structural analysis of the SRP Alu domain from Plasmodium falciparum reveals a non-canonical open conformation. Commun Biol 2021; 4:600. [PMID: 34017052 PMCID: PMC8137916 DOI: 10.1038/s42003-021-02132-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 04/22/2021] [Indexed: 12/25/2022] Open
Abstract
The eukaryotic signal recognition particle (SRP) contains an Alu domain, which docks into the factor binding site of translating ribosomes and confers translation retardation. The canonical Alu domain consists of the SRP9/14 protein heterodimer and a tRNA-like folded Alu RNA that adopts a strictly 'closed' conformation involving a loop-loop pseudoknot. Here, we study the structure of the Alu domain from Plasmodium falciparum (PfAlu), a divergent apicomplexan protozoan that causes human malaria. Using NMR, SAXS and cryo-EM analyses, we show that, in contrast to its prokaryotic and eukaryotic counterparts, the PfAlu domain adopts an 'open' Y-shaped conformation. We show that cytoplasmic P. falciparum ribosomes are non-discriminative and recognize both the open PfAlu and closed human Alu domains with nanomolar affinity. In contrast, human ribosomes do not provide high affinity binding sites for either of the Alu domains. Our analyses extend the structural database of Alu domains to the protozoan species and reveal species-specific differences in the recognition of SRP Alu domains by ribosomes.
Collapse
Affiliation(s)
- Komal Soni
- Heidelberg University Biochemistry Center (BZH), Heidelberg, Germany
| | - Georg Kempf
- Heidelberg University Biochemistry Center (BZH), Heidelberg, Germany
| | | | - Astrid Hendricks
- Heidelberg University Biochemistry Center (BZH), Heidelberg, Germany
| | - Dirk Flemming
- Heidelberg University Biochemistry Center (BZH), Heidelberg, Germany
| | - Julien Guizetti
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Bernd Simon
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Friedrich Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Klemens Wild
- Heidelberg University Biochemistry Center (BZH), Heidelberg, Germany
| | - Irmgard Sinning
- Heidelberg University Biochemistry Center (BZH), Heidelberg, Germany.
| |
Collapse
|
38
|
Castro-Salguedo C, Mendez-Cuadro D, Moneriz C. Erythrocyte membrane proteins involved in the immune response to Plasmodium falciparum and Plasmodium vivax infection. Parasitol Res 2021; 120:1789-1797. [PMID: 33797613 DOI: 10.1007/s00436-021-07135-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 03/21/2021] [Indexed: 11/29/2022]
Abstract
Invasion of Plasmodium into the red blood cell involves the interactions of a substantial number of proteins, with red cell membrane proteins as the most involved throughout the process from entry to exit. The objective of this work was to identify proteins of the human erythrocyte membrane capable of generating an antigenic response to P. falciparum and P. vivax infection, with the goal of searching for new molecular targets of interest with an immunological origin to prevent Plasmodium infection. To identify these proteins, an immunoproteomic technique was carried out in four stages: protein separation (electrophoresis), detection of antigenic proteins (western blotting), identification of proteins of interest (mass spectrometry), and interpretation of the data (bioinformatic analysis). Four proteins were identified from extracts of membrane proteins from erythrocytes infected with P. falciparum: Spectrin, Ankyrin-1, Band 3 and band 4.2, and a single protein was identified from erythrocytes infected with P. vivax: Band 3. These results demonstrate that modifications in the red blood cell membrane during infection with P. falciparum and P. vivax can generate an immune response, altering proteins of great structural and functional importance.
Collapse
Affiliation(s)
- Cristian Castro-Salguedo
- Biochemistry and Diseases Research Group, Facultad de Medicina, Universidad de Cartagena, Cartagena, 130015, Colombia.,Grupo de Investigaciones Biomédicas-GIB, Universidad de San Buenaventura, Cartagena, 130010, Colombia
| | - Darío Mendez-Cuadro
- Analytical Chemistry and Biomedicine Group, Facultad de Ciencias Exactas y Naturales, Universidad de Cartagena, Cartagena, 130015, Colombia
| | - Carlos Moneriz
- Biochemistry and Diseases Research Group, Facultad de Medicina, Universidad de Cartagena, Cartagena, 130015, Colombia.
| |
Collapse
|
39
|
Abstract
Obligate intracellular malaria parasites reside within a vacuolar compartment generated during invasion which is the principal interface between pathogen and host. To subvert their host cell and support their metabolism, these parasites coordinate a range of transport activities at this membrane interface that are critically important to parasite survival and virulence, including nutrient import, waste efflux, effector protein export, and uptake of host cell cytosol. Here, we review our current understanding of the transport mechanisms acting at the malaria parasite vacuole during the blood and liver-stages of development with a particular focus on recent advances in our understanding of effector protein translocation into the host cell by the Plasmodium Translocon of EXported proteins (PTEX) and small molecule transport by the PTEX membrane-spanning pore EXP2. Comparison to Toxoplasma gondii and other related apicomplexans is provided to highlight how similar and divergent mechanisms are employed to fulfill analogous transport activities.
Collapse
Affiliation(s)
- Josh R. Beck
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, United States of America
| | - Chi-Min Ho
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
| |
Collapse
|
40
|
Zhou M, Varol A, Efferth T. Multi-omics approaches to improve malaria therapy. Pharmacol Res 2021; 167:105570. [PMID: 33766628 DOI: 10.1016/j.phrs.2021.105570] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/02/2021] [Accepted: 03/16/2021] [Indexed: 01/07/2023]
Abstract
Malaria contributes to the most widespread infectious diseases worldwide. Even though current drugs are commercially available, the ever-increasing drug resistance problem by malaria parasites poses new challenges in malaria therapy. Hence, searching for efficient therapeutic strategies is of high priority in malaria control. In recent years, multi-omics technologies have been extensively applied to provide a more holistic view of functional principles and dynamics of biological mechanisms. We briefly review multi-omics technologies and focus on recent malaria progress conducted with the help of various omics methods. Then, we present up-to-date advances for multi-omics approaches in malaria. Next, we describe resistance phenomena to established antimalarial drugs and underlying mechanisms. Finally, we provide insight into novel multi-omics approaches, new drugs and vaccine developments and analyze current gaps in multi-omics research. Although multi-omics approaches have been successfully used in malaria studies, they are still limited. Many gaps need to be filled to bridge the gap between basic research and treatment of malaria patients. Multi-omics approaches will foster a better understanding of the molecular mechanisms of Plasmodium that are essential for the development of novel drugs and vaccines to fight this disastrous disease.
Collapse
Affiliation(s)
- Min Zhou
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
| | - Ayşegül Varol
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany.
| |
Collapse
|
41
|
Fraser M, Jing W, Bröer S, Kurth F, Sander LE, Matuschewski K, Maier AG. Breakdown in membrane asymmetry regulation leads to monocyte recognition of P. falciparum-infected red blood cells. PLoS Pathog 2021; 17:e1009259. [PMID: 33600495 PMCID: PMC7891792 DOI: 10.1371/journal.ppat.1009259] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 12/28/2020] [Indexed: 02/06/2023] Open
Abstract
The human malaria parasite Plasmodium falciparum relies on lipids to survive; this makes its lipid metabolism an attractive drug target. The lipid phosphatidylserine (PS) is usually confined to the inner leaflet of the red blood cell membrane (RBC) bilayer; however, some studies suggest that infection with the intracellular parasite results in the presence of this lipid in the RBC membrane outer leaflet, where it could act as a recognition signal to phagocytes. Here, we used fluorescent lipid analogues and probes to investigate the enzymatic reactions responsible for maintaining asymmetry between membrane leaflets, and found that in parasitised RBCs the maintenance of membrane asymmetry was partly disrupted, and PS was increased in the outer leaflet. We examined the underlying causes for the differences between uninfected and infected RBCs using fluorescent dyes and probes, and found that calcium levels increased in the infected RBC cytoplasm, whereas membrane cholesterol was depleted from the erythrocyte plasma membrane. We explored the resulting effect of PS exposure on enhanced phagocytosis by monocytes, and show that infected RBCs must expend energy to limit phagocyte recognition, and provide experimental evidence that PS exposure contributes to phagocytic recognition of P. falciparum-infected RBCs. Together, these findings underscore the pivotal role for PS exposure on the surface of Plasmodium falciparum-infected erythrocytes for in vivo interactions with the host immune system, and provide a rationale for targeted antimalarial drug design.
Collapse
Affiliation(s)
- Merryn Fraser
- Research School of Biology, The Australian National University, Canberra, Australia
- Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, Germany
| | - Weidong Jing
- Research School of Biology, The Australian National University, Canberra, Australia
| | - Stefan Bröer
- Research School of Biology, The Australian National University, Canberra, Australia
| | - Florian Kurth
- Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Germany
- Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Department of Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Leif-Erik Sander
- Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Germany
| | - Kai Matuschewski
- Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, Germany
| | - Alexander G. Maier
- Research School of Biology, The Australian National University, Canberra, Australia
| |
Collapse
|
42
|
Ghosh D, Stumhofer JS. The spleen: "epicenter" in malaria infection and immunity. J Leukoc Biol 2021; 110:753-769. [PMID: 33464668 PMCID: PMC8518401 DOI: 10.1002/jlb.4ri1020-713r] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/10/2020] [Accepted: 12/11/2020] [Indexed: 12/14/2022] Open
Abstract
The spleen is a complex secondary lymphoid organ that plays a crucial role in controlling blood‐stage infection with Plasmodium parasites. It is tasked with sensing and removing parasitized RBCs, erythropoiesis, the activation and differentiation of adaptive immune cells, and the development of protective immunity, all in the face of an intense inflammatory environment. This paper describes how these processes are regulated following infection and recognizes the gaps in our current knowledge, highlighting recent insights from human infections and mouse models.
Collapse
Affiliation(s)
- Debopam Ghosh
- Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Jason S Stumhofer
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
43
|
Dousti M, Manzano-Román R, Rashidi S, Barzegar G, Ahmadpour NB, Mohammadi A, Hatam G. A proteomic glimpse into the effect of antimalarial drugs on Plasmodium falciparum proteome towards highlighting possible therapeutic targets. Pathog Dis 2021; 79:ftaa071. [PMID: 33202000 DOI: 10.1093/femspd/ftaa071] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 11/13/2020] [Indexed: 02/07/2023] Open
Abstract
There is no effective vaccine against malaria; therefore, chemotherapy is to date the only choice to fight against this infectious disease. However, there is growing evidences of drug-resistance mechanisms in malaria treatments. Therefore, the identification of new drug targets is an urgent need for the clinical management of the disease. Proteomic approaches offer the chance of determining the effects of antimalarial drugs on the proteome of Plasmodium parasites. Accordingly, we reviewed the effects of antimalarial drugs on the Plasmodium falciparum proteome pointing out the relevance of several proteins as possible drug targets in malaria treatment. In addition, some of the P. falciparum stage-specific altered proteins and parasite-host interactions might play important roles in pathogenicity, survival, invasion and metabolic pathways and thus serve as potential sources of drug targets. In this review, we have identified several proteins, including thioredoxin reductase, helicases, peptidyl-prolyl cis-trans isomerase, endoplasmic reticulum-resident calcium-binding protein, choline/ethanolamine phosphotransferase, purine nucleoside phosphorylase, apical membrane antigen 1, glutamate dehydrogenase, hypoxanthine guanine phosphoribosyl transferase, heat shock protein 70x, knob-associated histidine-rich protein and erythrocyte membrane protein 1, as promising antimalarial drugs targets. Overall, proteomic approaches are able to partially facilitate finding possible drug targets. However, the integration of other 'omics' and specific pharmaceutical techniques with proteomics may increase the therapeutic properties of the critical proteins identified in the P. falciparum proteome.
Collapse
Affiliation(s)
- Majid Dousti
- Department of Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Raúl Manzano-Román
- Proteomics Unit, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007, Salamanca, Spain
| | - Sajad Rashidi
- Department of Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Gholamreza Barzegar
- Department of Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Alireza Mohammadi
- Department of Disease Control, Komijan Treatment and Health Network, Arak University of Medical Science, Iran
| | - Gholamreza Hatam
- Basic Sciences in Infectious Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
44
|
Edkins AL, Boshoff A. General Structural and Functional Features of Molecular Chaperones. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1340:11-73. [PMID: 34569020 DOI: 10.1007/978-3-030-78397-6_2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Molecular chaperones are a group of structurally diverse and highly conserved ubiquitous proteins. They play crucial roles in facilitating the correct folding of proteins in vivo by preventing protein aggregation or facilitating the appropriate folding and assembly of proteins. Heat shock proteins form the major class of molecular chaperones that are responsible for protein folding events in the cell. This is achieved by ATP-dependent (folding machines) or ATP-independent mechanisms (holders). Heat shock proteins are induced by a variety of stresses, besides heat shock. The large and varied heat shock protein class is categorised into several subfamilies based on their sizes in kDa namely, small Hsps (HSPB), J domain proteins (Hsp40/DNAJ), Hsp60 (HSPD/E; Chaperonins), Hsp70 (HSPA), Hsp90 (HSPC), and Hsp100. Heat shock proteins are localised to different compartments in the cell to carry out tasks specific to their environment. Most heat shock proteins form large oligomeric structures, and their functions are usually regulated by a variety of cochaperones and cofactors. Heat shock proteins do not function in isolation but are rather part of the chaperone network in the cell. The general structural and functional features of the major heat shock protein families are discussed, including their roles in human disease. Their function is particularly important in disease due to increased stress in the cell. Vector-borne parasites affecting human health encounter stress during transmission between invertebrate vectors and mammalian hosts. Members of the main classes of heat shock proteins are all represented in Plasmodium falciparum, the causative agent of cerebral malaria, and they play specific functions in differentiation, cytoprotection, signal transduction, and virulence.
Collapse
Affiliation(s)
- Adrienne Lesley Edkins
- Biomedical Biotechnology Research Unit (BioBRU), Department of Biochemistry and Microbiology, Rhodes University, Makhanda/Grahamstown, South Africa.
- Rhodes University, Makhanda/Grahamstown, South Africa.
| | - Aileen Boshoff
- Rhodes University, Makhanda/Grahamstown, South Africa.
- Biotechnology Innovation Centre, Rhodes University, Makhanda/Grahamstown, South Africa.
| |
Collapse
|
45
|
Keswani T, Delcroix-Genete D, Herbert F, Leleu I, Lambert C, Draheim M, Salome-Desnoulez S, Saliou JM, Cazenave PA, Silvie O, Roland J, Pied S. Plasmodium yoelii Uses a TLR3-Dependent Pathway to Achieve Mammalian Host Parasitism. THE JOURNAL OF IMMUNOLOGY 2020; 205:3071-3082. [PMID: 33148715 DOI: 10.4049/jimmunol.1901317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 09/28/2020] [Indexed: 02/06/2023]
Abstract
Malaria is associated with complicated immunopathogenesis. In this study, we provide evidence for an unexpected role of TLR3 in promoting the establishment of Plasmodium yoelii infection through delayed clearance of parasitemia in wild type C57BL/6jRj (B6) compared with TLR3 knockout mice. In this study, we confirmed an increased expression of Tlr3, Trif, Tbk1, and Irf7/Irf3 in the liver 42 h postinfection and the initiation of an early burst of proinflammatory response such as Ifng, NF-kB, and Tnfa in B6 mice that may promote parasite fitness. Interestingly, in the absence of TLR3, we showed the involvement of high IFN-γ and lower type I IFN response in the early clearance of parasitemia. In parallel, we observed an increase in splenic NK and NKT cells expressing TLR3 in infected B6 mice, suggesting a role for TLR sensing in the innate immune response. Finally, we find evidence that the increase in the frequency of CD19+TLR3+ B cells along with reduced levels of total IgG in B6 mice possibly suggests the initiation of TLR3-dependent pathway early during P. yoelii infection. Our results thus reveal a new mechanism in which a parasite-activated TLR3 pathway promotes blood stage infection along with quantitative and qualitative differences in Ab responses.
Collapse
Affiliation(s)
- Tarun Keswani
- Team 10: Tropical Biomes & Immunopathophysiology, Université de Lille, Centre Hospitalier Régional Universitaire de Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 9017 - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France
| | - Delphine Delcroix-Genete
- Team 10: Tropical Biomes & Immunopathophysiology, Université de Lille, Centre Hospitalier Régional Universitaire de Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 9017 - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France
| | - Fabien Herbert
- Team 10: Tropical Biomes & Immunopathophysiology, Université de Lille, Centre Hospitalier Régional Universitaire de Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 9017 - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France
| | - Ines Leleu
- Team 10: Tropical Biomes & Immunopathophysiology, Université de Lille, Centre Hospitalier Régional Universitaire de Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 9017 - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France
| | - Claire Lambert
- Team 10: Tropical Biomes & Immunopathophysiology, Université de Lille, Centre Hospitalier Régional Universitaire de Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 9017 - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France
| | - Marion Draheim
- Team 10: Tropical Biomes & Immunopathophysiology, Université de Lille, Centre Hospitalier Régional Universitaire de Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 9017 - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France
| | | | - Jean Michel Saliou
- Team 10: Tropical Biomes & Immunopathophysiology, Université de Lille, Centre Hospitalier Régional Universitaire de Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 9017 - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France
| | - Pierre-André Cazenave
- Team 10: Tropical Biomes & Immunopathophysiology, Université de Lille, Centre Hospitalier Régional Universitaire de Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 9017 - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France
| | - Olivier Silvie
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, 75013 Paris, France
| | - Jacques Roland
- Team 10: Tropical Biomes & Immunopathophysiology, Université de Lille, Centre Hospitalier Régional Universitaire de Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 9017 - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France
| | - Sylviane Pied
- Team 10: Tropical Biomes & Immunopathophysiology, Université de Lille, Centre Hospitalier Régional Universitaire de Lille, CNRS, INSERM, Institut Pasteur de Lille, U1019 - UMR 9017 - Centre d'Infection et d'Immunité de Lille, F-59000 Lille, France;
| |
Collapse
|
46
|
Yang B, Wang X, Jiang N, Sang X, Feng Y, Chen R, Wang X, Chen Q. Interaction Analysis of a Plasmodium falciparum PHISTa-like Protein and PfEMP1 Proteins. Front Microbiol 2020; 11:611190. [PMID: 33281807 PMCID: PMC7691434 DOI: 10.3389/fmicb.2020.611190] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 10/26/2020] [Indexed: 11/13/2022] Open
Abstract
Plasmodium falciparum extensively remodels host cells by translocating numerous proteins into the cytoplasm of red blood cells (RBCs) after invasion. Among these exported proteins, members of the Plasmodium helical interspersed subtelomeric (PHIST) family are crucial for host cell remodeling and host-parasite interactions, and thereby contribute to malaria pathogenesis. Herein, we explored the function of PF3D7_1372300, a member of the PHIST/PHISTa-like subfamily. PF3D7_1372300 was highly transcribed and expressed during the blood stage of P. falciparum, and distributed throughout RBCs, but most abundant at the erythrocyte membrane. Specific interaction of PF3D7_1372300 with the cytoplasmic tail of P. falciparum erythrocyte membrane protein 1 (PfEMP1) was revealed by immunofluorescence assay, in vitro intermolecular interaction assays. The interaction sites of PF3D7_1372300 with PfEMP1 ATS domain were found involved more than 30 amino acids (aa) at several positions. The findings deepen our understanding of host-parasite interactions and malaria pathogenesis.
Collapse
Affiliation(s)
- Baoling Yang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, Shenyang Agricultural University, Shenyang, China.,College of Food Science and Technology, Shenyang Agricultural University, Shenyang, China.,The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, China
| | - Xiaofeng Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences (CAS), University of Chinese Academy of Sciences, Beijing, China
| | - Ning Jiang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, Shenyang Agricultural University, Shenyang, China.,The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, China
| | - Xiaoyu Sang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, Shenyang Agricultural University, Shenyang, China.,The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, China
| | - Ying Feng
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, Shenyang Agricultural University, Shenyang, China.,The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, China
| | - Ran Chen
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, Shenyang Agricultural University, Shenyang, China.,The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, China
| | - Xinyi Wang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, Shenyang Agricultural University, Shenyang, China.,College of Basic Sciences, Shenyang Agricultural University, Shenyang, China
| | - Qijun Chen
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, Shenyang Agricultural University, Shenyang, China.,The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, China
| |
Collapse
|
47
|
Live-Cell FRET Reveals that Malaria Nutrient Channel Proteins CLAG3 and RhopH2 Remain Associated throughout Their Tortuous Trafficking. mBio 2020; 11:mBio.01354-20. [PMID: 32900800 PMCID: PMC7482060 DOI: 10.1128/mbio.01354-20] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Malaria parasites increase their host erythrocyte's permeability to various nutrients, fueling intracellular pathogen development and replication. The plasmodial surface anion channel (PSAC) mediates this uptake and is linked to the parasite-encoded RhopH complex, consisting of CLAG3, RhopH2, and RhopH3. While interactions between these subunits are well established, it is not clear whether they remain associated from their synthesis in developing merozoites through erythrocyte invasion and trafficking to the host membrane. Here, we explored protein-protein interactions between RhopH subunits using live-cell imaging and Förster resonance energy transfer (FRET) experiments. Using the green fluorescent protein (GFP) derivatives mCerulean and mVenus, we generated single- and double-tagged parasite lines for fluorescence measurements. While CLAG3-mCerulean served as an efficient FRET donor for RhopH2-mVenus within rhoptry organelles, mCerulean targeted to this organelle via a short signal sequence produced negligible FRET. Upon merozoite egress and reinvasion, these tagged RhopH subunits were deposited into the new host cell's parasitophorous vacuole; these proteins were then exported and trafficked to the erythrocyte membrane, where CLAG3 and RhopH2 remained fully associated. Fluorescence intensity measurements identified stoichiometric increases in exported RhopH protein when erythrocytes are infected with two parasites; whole-cell patch-clamp revealed a concomitant increase in PSAC functional copy number and a dose effect for RhopH contribution to ion and nutrient permeability. These studies establish live-cell FRET imaging in human malaria parasites, reveal that RhopH subunits traffic to their host membrane destination without dissociation, and suggest quantitative contribution to PSAC formation.IMPORTANCE Malaria parasites grow within circulating red blood cells and uptake nutrients through a pore on their host membrane. Here, we used gene editing to tag CLAG3 and RhopH2, two proteins linked to the nutrient pore, with fluorescent markers and tracked these proteins in living infected cells. After their synthesis in mature parasites, imaging showed that both proteins are packaged into membrane-bound rhoptries. When parasites ruptured their host cells and invaded new red blood cells, these proteins were detected within a vacuole around the parasite before they migrated and inserted in the surface membrane of the host cell. Using simultaneous labeling of CLAG3 and RhopH2, we determined that these proteins interact tightly during migration and after surface membrane insertion. Red blood cells infected with two parasites had twice the protein at their surface and a parallel increase in the number of nutrient pores. Our work suggests that these proteins directly facilitate parasite nutrient uptake from human plasma.
Collapse
|
48
|
Kiyuka PK, Meri S, Khattab A. Complement in malaria: immune evasion strategies and role in protective immunity. FEBS Lett 2020; 594:2502-2517. [PMID: 32181490 PMCID: PMC8653895 DOI: 10.1002/1873-3468.13772] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 03/03/2020] [Accepted: 03/03/2020] [Indexed: 12/12/2022]
Abstract
The malaria parasite has for long been thought to escape host complement attack as a survival strategy. However, it was only recently that complement evasion mechanisms of the parasite were described. Simultaneously, the role of complement in antibody-mediated naturally acquired and vaccine-induced protection against malaria has also been reported. Such findings should be considered in future vaccine design, given the current need to develop more efficacious vaccines against malaria. Parasite antigens derived from molecules mediating functions crucial for parasite survival, such as complement evasion, or parasite antigens against which antibody responses lead to an efficient complement attack could present new candidates for vaccines. In this review, we discuss recent findings on complement evasion by the malaria parasites and the emerging role of complement in antibody-mediated protection against malaria. We emphasize that immune responses to vaccines based on complement inhibitors should not only induce complement-activating antibodies but also neutralize the escape mechanisms of the parasite.
Collapse
Affiliation(s)
- Patience Kerubo Kiyuka
- Department of Bacteriology and Immunology, Translational Immunology Research Program, Haartman Institute, University of Helsinki, Finland
- KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-Coast, Kilifi, Kenya
| | - Seppo Meri
- Department of Bacteriology and Immunology, Translational Immunology Research Program, Haartman Institute, University of Helsinki, Finland
- Helsinki University Central Hospital, Finland
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Ayman Khattab
- Department of Bacteriology and Immunology, Translational Immunology Research Program, Haartman Institute, University of Helsinki, Finland
- Department of Nucleic Acid Research, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications, Alexandria, Egypt
| |
Collapse
|
49
|
Osii RS, Otto TD, Garside P, Ndungu FM, Brewer JM. The Impact of Malaria Parasites on Dendritic Cell-T Cell Interaction. Front Immunol 2020; 11:1597. [PMID: 32793231 PMCID: PMC7393936 DOI: 10.3389/fimmu.2020.01597] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 06/16/2020] [Indexed: 12/13/2022] Open
Abstract
Malaria is caused by apicomplexan parasites of the genus Plasmodium. While infection continues to pose a risk for the majority of the global population, the burden of disease mainly resides in Sub-Saharan Africa. Although immunity develops against disease, this requires years of persistent exposure and is not associated with protection against infection. Repeat infections occur due to the parasite's ability to disrupt or evade the host immune responses. However, despite many years of study, the mechanisms of this disruption remain unclear. Previous studies have demonstrated a parasite-induced failure in dendritic cell (DCs) function affecting the generation of helper T cell responses. These T cells fail to help B cell responses, reducing the production of antibodies that are necessary to control malaria infection. This review focuses on our current understanding of the effect of Plasmodium parasite on DC function, DC-T cell interaction, and T cell activation. A better understanding of how parasites disrupt DC-T cell interactions will lead to new targets and approaches to reinstate adaptive immune responses and enhance parasite immunity.
Collapse
Affiliation(s)
- Rowland S Osii
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, United Kingdom.,KEMRI-CGMRC/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Thomas D Otto
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Paul Garside
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Francis M Ndungu
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, United Kingdom.,KEMRI-CGMRC/Wellcome Trust Research Programme, Kilifi, Kenya.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - James M Brewer
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
50
|
Dziekan JM, Wirjanata G, Dai L, Go KD, Yu H, Lim YT, Chen L, Wang LC, Puspita B, Prabhu N, Sobota RM, Nordlund P, Bozdech Z. Cellular thermal shift assay for the identification of drug-target interactions in the Plasmodium falciparum proteome. Nat Protoc 2020; 15:1881-1921. [PMID: 32341577 DOI: 10.1038/s41596-020-0310-z] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 01/13/2020] [Indexed: 02/06/2023]
Abstract
Despite decades of research, little is known about the cellular targets and the mode of action of the vast majority of antimalarial drugs. We recently demonstrated that the cellular thermal shift assay (CETSA) protocol in its two variants: the melt curve and the isothermal dose-response, represents a comprehensive strategy for the identification of antimalarial drug targets. CETSA enables proteome-wide target screening for unmodified antimalarial compounds with undetermined mechanisms of action, providing quantitative evidence about direct drug-protein interactions. The experimental workflow involves treatment of P. falciparum-infected erythrocytes with a compound of interest, heat exposure to denature proteins, soluble protein isolation, enzymatic digestion, peptide labeling with tandem mass tags, offline fractionation, and liquid chromatography-tandem mass spectrometry analysis. Methodological optimizations necessary for the analysis of this intracellular parasite are discussed, including enrichment of parasitized cells and hemoglobin depletion strategies to overcome high hemoglobin abundance in the host red blood cells. We outline an effective data processing workflow using the mineCETSA R package, which enables prioritization of drug-target candidates for follow-up studies. The entire protocol can be completed within 2 weeks.
Collapse
Affiliation(s)
- Jerzy Michal Dziekan
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Grennady Wirjanata
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Lingyun Dai
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- The Second Clinical Medical College of Jinan University, First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People's Hospital, Shenzhen, China
| | - Ka Diam Go
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Han Yu
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Yan Ting Lim
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Functional Proteomics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Liyan Chen
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Functional Proteomics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Loo Chien Wang
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Functional Proteomics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Brenda Puspita
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Nayana Prabhu
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Radoslaw M Sobota
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- Functional Proteomics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
| | - Pär Nordlund
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.
| | - Zbynek Bozdech
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|