1
|
Roessner R, Michelarakis N, Gräter F, Aponte-Santamaría C. Mechanical forces control the valency of the malaria adhesin VAR2CSA by exposing cryptic glycan binding sites. PLoS Comput Biol 2023; 19:e1011726. [PMID: 38117828 PMCID: PMC10786402 DOI: 10.1371/journal.pcbi.1011726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 01/12/2024] [Accepted: 12/02/2023] [Indexed: 12/22/2023] Open
Abstract
Plasmodium falciparum (Pf) is responsible for the most lethal form of malaria. VAR2CSA is an adhesin protein expressed by this parasite at the membrane of infected erythrocytes for attachment to the placenta, leading to pregnancy-associated malaria. VAR2CSA is a large 355 kDa multidomain protein composed of nine extracellular domains, a transmembrane helix, and an intracellular domain. VAR2CSA binds to Chondroitin Sulphate A (CSA) of the proteoglycan matrix of the placenta. Shear flow, as the one occurring in blood, has been shown to enhance the (VAR2CSA-mediated) adhesion of Pf-infected erythrocytes on the CSA-matrix. However, the underlying molecular mechanism governing this enhancement has remained elusive. Here, we address this question by using equilibrium, force-probe, and docking-based molecular dynamics simulations. We subjected the VAR2CSA protein-CSA sugar complex to a force mimicking the tensile force exerted on this system due to the shear of the flowing blood. We show that upon this force exertion, VAR2CSA undergoes a large opening conformational transition before the CSA sugar chain dissociates from its main binding site. This preferential order of events is caused by the orientation of the molecule during elongation, as well as the strong electrostatic attraction of the sugar to the main protein binding site. Upon opening, two additional cryptic CSA binding sites get exposed and a functional dodecameric CSA molecule can be stably accommodated at these force-exposed positions. Thus, our results suggest that mechanical forces increase the avidity of VAR2CSA by turning it from a monovalent to a multivalent state. We propose this to be the molecular cause of the observed shear-enhanced adherence. Mechanical control of the valency of VAR2CSA is an intriguing hypothesis that can be tested experimentally and which is of relevance for the understanding of the malaria infection and for the development of anti placental-malaria vaccines targeting VAR2CSA.
Collapse
Affiliation(s)
- Rita Roessner
- Molecular Biomechanics Group, Heidelberg Institute for Theoretical Studies, Heidelberg, Germany
| | - Nicholas Michelarakis
- Molecular Biomechanics Group, Heidelberg Institute for Theoretical Studies, Heidelberg, Germany
| | - Frauke Gräter
- Molecular Biomechanics Group, Heidelberg Institute for Theoretical Studies, Heidelberg, Germany
- Interdisciplinary Center for Scientific Computing, Heidelberg University, Heidelberg, Germany
| | | |
Collapse
|
2
|
Natama HM, Moncunill G, Vidal M, Rouamba T, Aguilar R, Santano R, Rovira-Vallbona E, Jiménez A, Somé MA, Sorgho H, Valéa I, Coulibaly-Traoré M, Coppel RL, Cavanagh D, Chitnis CE, Beeson JG, Angov E, Dutta S, Gamain B, Izquierdo L, Mens PF, Schallig HDFH, Tinto H, Rosanas-Urgell A, Dobaño C. Associations between prenatal malaria exposure, maternal antibodies at birth, and malaria susceptibility during the first year of life in Burkina Faso. Infect Immun 2023; 91:e0026823. [PMID: 37754682 PMCID: PMC10580994 DOI: 10.1128/iai.00268-23] [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: 08/03/2023] [Accepted: 08/12/2023] [Indexed: 09/28/2023] Open
Abstract
In this study, we investigated how different categories of prenatal malaria exposure (PME) influence levels of maternal antibodies in cord blood samples and the subsequent risk of malaria in early childhood in a birth cohort study (N = 661) nested within the COSMIC clinical trial (NCT01941264) in Burkina Faso. Plasmodium falciparum infections during pregnancy and infants' clinical malaria episodes detected during the first year of life were recorded. The levels of maternal IgG and IgG1-4 to 15 P. falciparum antigens were measured in cord blood by quantitative suspension array technology. Results showed a significant variation in the magnitude of maternal antibody levels in cord blood, depending on the PME category, with past placental malaria (PM) more frequently associated with significant increases of IgG and/or subclass levels across three groups of antigens defined as pre-erythrocytic, erythrocytic, and markers of PM, as compared to those from the cord of non-exposed control infants. High levels of antibodies to certain erythrocytic antigens (i.e., IgG to EBA140 and EBA175, IgG1 to EBA175 and MSP142, and IgG3 to EBA140 and MSP5) were independent predictors of protection from clinical malaria during the first year of life. By contrast, high levels of IgG, IgG1, and IgG2 to the VAR2CSA DBL1-2 and IgG4 to DBL3-4 were significantly associated with an increased risk of clinical malaria. These findings indicate that PME categories have different effects on the levels of maternal-derived antibodies to malaria antigens in children at birth, and this might drive heterogeneity to clinical malaria susceptibility in early childhood.
Collapse
Affiliation(s)
- Hamtandi Magloire Natama
- Unité de Recherche Clinique de Nanoro, Institut de Recherche en Sciences de la Santé, Direction Régionale du Centre-Ouest, Nanoro, Burkina Faso
| | - Gemma Moncunill
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic – Universitat de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Marta Vidal
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic – Universitat de Barcelona, Barcelona, Spain
| | - Toussaint Rouamba
- Unité de Recherche Clinique de Nanoro, Institut de Recherche en Sciences de la Santé, Direction Régionale du Centre-Ouest, Nanoro, Burkina Faso
| | - Ruth Aguilar
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic – Universitat de Barcelona, Barcelona, Spain
| | - Rebeca Santano
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic – Universitat de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Eduard Rovira-Vallbona
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic – Universitat de Barcelona, Barcelona, Spain
| | - Alfons Jiménez
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic – Universitat de Barcelona, Barcelona, Spain
- CIBER de Epidemiologia y Salud Pública (CIBERESP), Barcelona, Spain
| | - M. Athanase Somé
- Unité de Recherche Clinique de Nanoro, Institut de Recherche en Sciences de la Santé, Direction Régionale du Centre-Ouest, Nanoro, Burkina Faso
| | - Hermann Sorgho
- Unité de Recherche Clinique de Nanoro, Institut de Recherche en Sciences de la Santé, Direction Régionale du Centre-Ouest, Nanoro, Burkina Faso
| | - Innocent Valéa
- Unité de Recherche Clinique de Nanoro, Institut de Recherche en Sciences de la Santé, Direction Régionale du Centre-Ouest, Nanoro, Burkina Faso
| | - Maminata Coulibaly-Traoré
- Unité de Recherche Clinique de Nanoro, Institut de Recherche en Sciences de la Santé, Direction Régionale du Centre-Ouest, Nanoro, Burkina Faso
| | - Ross L. Coppel
- Infection and Immunity Program, Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - David Cavanagh
- Centre for Immunity, Infection & Evolution, Institute of Immunology & Infection Research, Ashworth Laboratories, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Chetan E. Chitnis
- Malaria Parasite Biology and Vaccines Unit, Department of Parasites and Insect Vectors, Institut Pasteur, Université de Paris, Paris, France
| | | | - Evelina Angov
- U.S. Military Malaria Vaccine Program, Walter Reed Army Institute of Research (WRAIR), Silver Spring, Maryland, USA
| | - Sheetij Dutta
- U.S. Military Malaria Vaccine Program, Walter Reed Army Institute of Research (WRAIR), Silver Spring, Maryland, USA
| | | | - Luis Izquierdo
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic – Universitat de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| | - Petra F. Mens
- Academic Medical Centre at the University of Amsterdam, Amsterdam, the Netherlands
| | | | - Halidou Tinto
- Unité de Recherche Clinique de Nanoro, Institut de Recherche en Sciences de la Santé, Direction Régionale du Centre-Ouest, Nanoro, Burkina Faso
| | - Anna Rosanas-Urgell
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Carlota Dobaño
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic – Universitat de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Barcelona, Spain
| |
Collapse
|
3
|
Disulfide bond and crosslinking analyses reveal inter-domain interactions that contribute to the rigidity of placental malaria VAR2CSA structure and formation of CSA binding channel. Int J Biol Macromol 2023; 226:143-158. [PMID: 36470436 DOI: 10.1016/j.ijbiomac.2022.11.258] [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: 08/26/2022] [Revised: 11/15/2022] [Accepted: 11/24/2022] [Indexed: 12/11/2022]
Abstract
VAR2CSA, a multidomain Plasmodium falciparum protein, mediates the adherence of parasite-infected red blood cells to chondroitin 4-sulfate (C4S) in the placenta, contributing to placental malaria. Therefore, detailed understanding of VAR2CSA structure likely help developing strategies to treat placental malaria. The VAR2CSA ectodomain consists of an N-terminal segment (NTS), six Duffy binding-like (DBL) domains, and three interdomains (IDs) present in sequence NTS-DBL1x-ID1-DBL2x-ID2-DBL3x-DBL4ε-ID3-DBL5ε-DBL6ε. Recent electron microscopy studies showed that VAR2CSA is compactly organized into a globular structure containing C4S-binding channel, and that DBL5ε-DBL6ε arm is attached to the NTS-ID3 core structure. However, the structural elements involved in inter-domain interactions that stabilize the VAR2CSA structure remain largely not understood. Here, limited proteolysis and peptide mapping by mass spectrometry showed that VAR2CSA contains several inter-domain disulfide bonds that stabilize its compact structure. Chemical crosslinking-mass spectrometry showed that all IDs interact with DBL4ε; additionally, IDs interact with other DBL domains, demonstrating that IDs are the key structural scaffolds that shape the functional NTS-ID3 core. Ligand binding analysis suggested that NTS considerably restricts the C4S binding. Overall, our study revealed that inter-domain disulfide bonds and interactions between IDs and DBL domains contribute to the stability of VAR2CSA structural architecture and formation of C4S-binding channel.
Collapse
|
4
|
Talundzic E, Scott S, Owino SO, Campo DS, Lucchi NW, Udhayakumar V, Moore JM, Peterson DS. Polymorphic Molecular Signatures in Variable Regions of the Plasmodium falciparum var2csa DBL3x Domain Are Associated with Virulence in Placental Malaria. Pathogens 2022; 11:520. [PMID: 35631041 PMCID: PMC9147263 DOI: 10.3390/pathogens11050520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/15/2022] [Accepted: 04/24/2022] [Indexed: 11/17/2022] Open
Abstract
The Plasmodium falciparum protein VAR2CSA allows infected erythrocytes to accumulate within the placenta, inducing pathology and poor birth outcomes. Multiple exposures to placental malaria (PM) induce partial immunity against VAR2CSA, making it a promising vaccine candidate. However, the extent to which VAR2CSA genetic diversity contributes to immune evasion and virulence remains poorly understood. The deep sequencing of the var2csa DBL3X domain in placental blood from forty-nine primigravid and multigravid women living in malaria-endemic western Kenya revealed numerous unique sequences within individuals in association with chronic PM but not gravidity. Additional analysis unveiled four distinct sequence types that were variably present in mixed proportions amongst the study population. An analysis of the abundance of each of these sequence types revealed that one was inversely related to infant gestational age, another was inversely related to placental parasitemia, and a third was associated with chronic PM. The categorization of women according to the type to which their dominant sequence belonged resulted in the segregation of types as a function of gravidity: two types predominated in multigravidae whereas the other two predominated in primigravidae. The univariate logistic regression analysis of sequence type dominance further revealed that gravidity, maternal age, placental parasitemia, and hemozoin burden (within maternal leukocytes), reported a lack of antimalarial drug use, and infant gestational age and birth weight influenced the odds of membership in one or more of these sequence predominance groups. Cumulatively, these results show that unique var2csa sequences differentially appear in women with different PM exposure histories and segregate to types independently associated with maternal factors, infection parameters, and birth outcomes. The association of some var2csa sequence types with indicators of pathogenesis should motivate vaccine efforts to further identify and target VAR2CSA epitopes associated with maternal morbidity and poor birth outcomes.
Collapse
Affiliation(s)
- Eldin Talundzic
- Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA; (E.T.); (N.W.L.); (V.U.)
| | - Stephen Scott
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA;
| | - Simon O. Owino
- Boehringer Ingelheim Animal Health, Athens, GA 30601, USA;
| | - David S. Campo
- Molecular Epidemiology and Bioinformatics Laboratory, Division of Viral Hepatitis, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA;
| | - Naomi W. Lucchi
- Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA; (E.T.); (N.W.L.); (V.U.)
| | - Venkatachalam Udhayakumar
- Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA; (E.T.); (N.W.L.); (V.U.)
| | - Julie M. Moore
- Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL 32611, USA
| | - David S. Peterson
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA;
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
5
|
Cryo-EM reveals the architecture of placental malaria VAR2CSA and provides molecular insight into chondroitin sulfate binding. Nat Commun 2021; 12:2956. [PMID: 34011972 PMCID: PMC8134449 DOI: 10.1038/s41467-021-23254-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 04/16/2021] [Indexed: 12/13/2022] Open
Abstract
Placental malaria can have severe consequences for both mother and child and effective vaccines are lacking. Parasite-infected red blood cells sequester in the placenta through interaction between parasite-expressed protein VAR2CSA and the glycosaminoglycan chondroitin sulfate A (CS) abundantly present in the intervillous space. Here, we report cryo-EM structures of the VAR2CSA ectodomain at up to 3.1 Å resolution revealing an overall V-shaped architecture and a complex domain organization. Notably, the surface displays a single significantly electropositive patch, compatible with binding of negatively charged CS. Using molecular docking and molecular dynamics simulations as well as comparative hydroxyl radical protein foot-printing of VAR2CSA in complex with placental CS, we identify the CS-binding groove, intersecting with the positively charged patch of the central VAR2CSA structure. We identify distinctive conserved structural features upholding the macro-molecular domain complex and CS binding capacity of VAR2CSA as well as divergent elements possibly allowing immune escape at or near the CS binding site. These observations will support rational design of second-generation placental malaria vaccines. In placental malaria, interactions between parasite protein VAR2CSA and human glycosaminoglycan chondroitin sulfate A (CS) sequesters infected red blood cells in the placenta. Here, the authors provide cryo-EM structures of VAR2CSA and placental CS, identifying molecular interactions that could guide design of placental malaria vaccines.
Collapse
|
6
|
Ma R, Lian T, Huang R, Renn JP, Petersen JD, Zimmerberg J, Duffy PE, Tolia NH. Structural basis for placental malaria mediated by Plasmodium falciparum VAR2CSA. Nat Microbiol 2021; 6:380-391. [PMID: 33452495 PMCID: PMC7914210 DOI: 10.1038/s41564-020-00858-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 12/17/2020] [Indexed: 01/29/2023]
Abstract
Plasmodium falciparum VAR2CSA binds to chondroitin sulfate A (CSA) on the surface of the syncytiotrophoblast during placental malaria. This interaction facilitates placental sequestration of malaria parasites resulting in severe health outcomes for both the mother and her offspring. Furthermore, CSA is presented by diverse cancer cells and specific targeting of cells by VAR2CSA may become a viable approach for cancer treatment. In the present study, we determined the cryo-electron microscopy structures of the full-length ectodomain of VAR2CSA from P. falciparum strain NF54 in complex with CSA, and VAR2CSA from a second P. falciparum strain FCR3. The architecture of VAR2CSA is composed of a stable core flanked by a flexible arm. CSA traverses the core domain by binding within two channels and CSA binding does not induce major conformational changes in VAR2CSA. The CSA-binding elements are conserved across VAR2CSA variants and are flanked by polymorphic segments, suggesting immune selection outside the CSA-binding sites. This work provides paths for developing interventions against placental malaria and cancer.
Collapse
Affiliation(s)
- Rui Ma
- Host-Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Tengfei Lian
- Laboratory of Membrane Proteins and Structural Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rick Huang
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jonathan P. Renn
- Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jennifer D. Petersen
- Section on Integrative Biophysics, Division of Basic and Translational Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Joshua Zimmerberg
- Section on Integrative Biophysics, Division of Basic and Translational Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Patrick E. Duffy
- Vaccine Development Unit, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA,Pathogenesis and Immunity Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Niraj H. Tolia
- Host-Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA,Correspondence: (N.H.T.)
| |
Collapse
|
7
|
Gamain B, Chêne A, Viebig NK, Tuikue Ndam N, Nielsen MA. Progress and Insights Toward an Effective Placental Malaria Vaccine. Front Immunol 2021; 12:634508. [PMID: 33717176 PMCID: PMC7947914 DOI: 10.3389/fimmu.2021.634508] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 01/06/2021] [Indexed: 12/03/2022] Open
Abstract
In areas where Plasmodium falciparum transmission is endemic, clinical immunity against malaria is progressively acquired during childhood and adults are usually protected against the severe clinical consequences of the disease. Nevertheless, pregnant women, notably during their first pregnancies, are susceptible to placental malaria and the associated serious clinical outcomes. Placental malaria is characterized by the massive accumulation of P. falciparum infected erythrocytes and monocytes in the placental intervillous spaces leading to maternal anaemia, hypertension, stillbirth and low birth weight due to premature delivery, and foetal growth retardation. Remarkably, the prevalence of placental malaria sharply decreases with successive pregnancies. This protection is associated with the development of antibodies directed towards the surface of P. falciparum-infected erythrocytes from placental origin. Placental sequestration is mediated by the interaction between VAR2CSA, a member of the P. falciparum erythrocyte membrane protein 1 family expressed on the infected erythrocytes surface, and the placental receptor chondroitin sulfate A. VAR2CSA stands today as the leading candidate for a placental malaria vaccine. We recently reported the safety and immunogenicity of two VAR2CSA-derived placental malaria vaccines (PRIMVAC and PAMVAC), spanning the chondroitin sulfate A-binding region of VAR2CSA, in both malaria-naïve and P. falciparum-exposed non-pregnant women in two distinct Phase I clinical trials (ClinicalTrials.gov, NCT02658253 and NCT02647489). This review discusses recent advances in placental malaria vaccine development, with a focus on the recent clinical data, and discusses the next clinical steps to undertake in order to better comprehend vaccine-induced immunity and accelerate vaccine development.
Collapse
Affiliation(s)
- Benoît Gamain
- Université de Paris, Inserm, Biologie Intégrée du Globule Rouge, Paris, France.,Institut National de la Transfusion Sanguine, Paris, France
| | - Arnaud Chêne
- Université de Paris, Inserm, Biologie Intégrée du Globule Rouge, Paris, France.,Institut National de la Transfusion Sanguine, Paris, France
| | - Nicola K Viebig
- European Vaccine Initiative, UniversitätsKlinikum Heidelberg, Heidelberg, Germany
| | | | - Morten A Nielsen
- Centre for Medical Parasitology at Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Infectious Diseases, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
8
|
Alonso S, Vidal M, Ruiz-Olalla G, González R, Jairoce C, Manaca MN, Vázquez-Santiago M, Balcells R, Vala A, Rupérez M, Cisteró P, Fuente-Soro L, Angov E, Coppel RL, Gamain B, Cavanagh D, Beeson JG, Nhacolo A, Sevene E, Aponte JJ, Macete E, Aguilar R, Mayor A, Menéndez C, Dobaño C, Moncunill G. HIV infection and placental malaria reduce maternal transfer of multiple antimalarial antibodies in Mozambican women. J Infect 2021; 82:45-57. [PMID: 33636218 DOI: 10.1016/j.jinf.2021.02.024] [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: 09/25/2020] [Revised: 02/15/2021] [Accepted: 02/17/2021] [Indexed: 10/22/2022]
Abstract
OBJECTIVES Maternal Plasmodium falciparum-specific antibodies may contribute to protect infants against severe malaria. Our main objective was to evaluate the impact of maternal HIV infection and placental malaria on the cord blood levels and efficiency of placental transfer of IgG and IgG subclasses. METHODS In a cohort of 341 delivering HIV-negative and HIV-positive mothers from southern Mozambique, we measured total IgG and IgG subclasses in maternal and cord blood pairs by quantitative suspension array technology against eight P. falciparum antigens: Duffy-binding like domains 3-4 of VAR2CSA from the erythrocyte membrane protein 1, erythrocyte-binding antigen 140, exported protein 1 (EXP1), merozoite surface proteins 1, 2 and 5, and reticulocyte-binding-homologue-4.2 (Rh4.2). We performed univariable and multivariable regression models to assess the association of maternal HIV infection, placental malaria, maternal variables and pregnancy outcomes on cord antibody levels and antibody transplacental transfer. RESULTS Maternal antibody levels were the main determinants of cord antibody levels. HIV infection and placental malaria reduced the transfer and cord levels of IgG and IgG1, and this was antigen-dependent. Low birth weight was associated with an increase of IgG2 in cord against EXP1 and Rh4.2. CONCLUSIONS We found lower maternally transferred antibodies in HIV-exposed infants and those born from mothers with placental malaria, which may underlie increased susceptibility to malaria in these children.
Collapse
Affiliation(s)
- Selena Alonso
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain
| | - Marta Vidal
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain
| | - Gemma Ruiz-Olalla
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain
| | - Raquel González
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique
| | - Chenjerai Jairoce
- Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique
| | - M Nelia Manaca
- Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique
| | - Miquel Vázquez-Santiago
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain
| | - Reyes Balcells
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique
| | - Anifa Vala
- Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique
| | - María Rupérez
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique; Present address: London School of Hygiene and Tropical Medicine (LSHTM). Keppel Street, WC1E 7HT, London, UK
| | - Pau Cisteró
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain
| | - Laura Fuente-Soro
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique
| | - Evelina Angov
- U.S. Military Malaria Vaccine Program, Walter Reed Army Institute of Research (WRAIR), Silver Spring, Maryland, USA
| | - Ross L Coppel
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Melbourne, VIC, Australia
| | - Benoit Gamain
- Université Sorbonne Paris Cité, Université Paris Diderot, Inserm, INTS, Unité Biologie Intégrée du Globule Rouge UMR_S1134, Laboratoire d'Excellence GR-Ex, Paris, France
| | - David Cavanagh
- Institute of Immunology & Infection Research and Centre for Immunity, Infection & Evolution, Ashworth Laboratories, School of Biological Sciences, University of Edinburgh, King's Buildings, Charlotte Auerbach Rd, Edinburgh, EH9 3FL, UK
| | | | - Arsenio Nhacolo
- Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique
| | - Esperança Sevene
- Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique; Department of Physiologic Science, Clinical Pharmacology, Faculty of Medicine, Eduardo Mondlane University, Maputo, Mozambique
| | - John J Aponte
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique
| | - Eusébio Macete
- Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique
| | - Ruth Aguilar
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain
| | - Alfredo Mayor
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique
| | - Clara Menéndez
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique
| | - Carlota Dobaño
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique.
| | - Gemma Moncunill
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Carrer Rosselló 153, E-08036, Barcelona, Catalonia, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Rua 12, Cambeve, Vila de Manhiça, CP 1929, Maputo, Mozambique.
| |
Collapse
|
9
|
Tomlinson A, Semblat JP, Gamain B, Chêne A. VAR2CSA-Mediated Host Defense Evasion of Plasmodium falciparum Infected Erythrocytes in Placental Malaria. Front Immunol 2021; 11:624126. [PMID: 33633743 PMCID: PMC7900151 DOI: 10.3389/fimmu.2020.624126] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/23/2020] [Indexed: 12/04/2022] Open
Abstract
Over 30 million women living in P. falciparum endemic areas are at risk of developing malaria during pregnancy every year. Placental malaria is characterized by massive accumulation of infected erythrocytes in the intervillous space of the placenta, accompanied by infiltration of immune cells, particularly monocytes. The consequent local inflammation and the obstruction of the maternofetal exchanges can lead to severe clinical outcomes for both mother and child. Even if protection against the disease can gradually be acquired following successive pregnancies, the malaria parasite has developed a large panel of evasion mechanisms to escape from host defense mechanisms and manipulate the immune system to its advantage. Infected erythrocytes isolated from placentas of women suffering from placental malaria present a unique phenotype and express the pregnancy-specific variant VAR2CSA of the Plasmodium falciparum Erythrocyte Membrane Protein (PfEMP1) family at their surface. The polymorphic VAR2CSA protein is able to mediate the interaction of infected erythrocytes with a variety of host cells including placental syncytiotrophoblasts and leukocytes but also with components of the immune system such as non-specific IgM. This review summarizes the described VAR2CSA-mediated host defense evasion mechanisms employed by the parasite during placental malaria to ensure its survival and persistence.
Collapse
Affiliation(s)
- Alice Tomlinson
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, INSERM, Paris, France.,Institut National de la Transfusion Sanguine, Paris, France.,Laboratory of Excellence GR-Ex, Paris, France
| | - Jean-Philippe Semblat
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, INSERM, Paris, France.,Institut National de la Transfusion Sanguine, Paris, France.,Laboratory of Excellence GR-Ex, Paris, France
| | - Benoît Gamain
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, INSERM, Paris, France.,Institut National de la Transfusion Sanguine, Paris, France.,Laboratory of Excellence GR-Ex, Paris, France
| | - Arnaud Chêne
- Université de Paris, Biologie Intégrée du Globule Rouge, UMR_S1134, BIGR, INSERM, Paris, France.,Institut National de la Transfusion Sanguine, Paris, France.,Laboratory of Excellence GR-Ex, Paris, France
| |
Collapse
|
10
|
Bewley MC, Gautam L, Jagadeeshaprasad MG, Gowda DC, Flanagan JM. Molecular architecture and domain arrangement of the placental malaria protein VAR2CSA suggests a model for carbohydrate binding. J Biol Chem 2020; 295:18589-18603. [PMID: 33122198 PMCID: PMC7939466 DOI: 10.1074/jbc.ra120.014676] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 10/13/2020] [Indexed: 11/29/2022] Open
Abstract
VAR2CSA is the placental-malaria-specific member of the antigenically variant Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) family. It is expressed on the surface of Plasmodium falciparum-infected host red blood cells and binds to specific chondroitin-4-sulfate chains of the placental proteoglycan receptor. The functional ∼310 kDa ectodomain of VAR2CSA is a multidomain protein that requires a minimum 12-mer chondroitin-4-sulfate molecule for specific, high affinity receptor binding. However, it is not known how the individual domains are organized and interact to create the receptor-binding surface, limiting efforts to exploit its potential as an effective vaccine or drug target. Using small angle X-ray scattering and single particle reconstruction from negative-stained electron micrographs of the ectodomain and multidomain constructs, we have determined the structural architecture of VAR2CSA. The relative locations of the domains creates two distinct pores that can each accommodate the 12-mer of chondroitin-4-sulfate, suggesting a model for receptor binding. This model has important implications for understanding cytoadherence of infected red blood cells and potentially provides a starting point for developing novel strategies to prevent and/or treat placental malaria.
Collapse
Affiliation(s)
- Maria C Bewley
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Lovely Gautam
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Mashanipalya G Jagadeeshaprasad
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - D Channe Gowda
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA.
| | - John M Flanagan
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA.
| |
Collapse
|
11
|
Kanoi BN, Nagaoka H, Morita M, Tsuboi T, Takashima E. Leveraging the wheat germ cell-free protein synthesis system to accelerate malaria vaccine development. Parasitol Int 2020; 80:102224. [PMID: 33137499 DOI: 10.1016/j.parint.2020.102224] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/04/2020] [Accepted: 09/16/2020] [Indexed: 01/29/2023]
Abstract
Vaccines against infectious diseases have had great successes in the history of public health. Major breakthroughs have occurred in the development of vaccine-based interventions against viral and bacterial pathogens through the application of classical vaccine design strategies. In contrast the development of a malaria vaccine has been slow. Plasmodium falciparum malaria affects millions of people with nearly half of the world population at risk of infection. Decades of dedicated research has taught us that developing an effective vaccine will be time consuming, challenging, and expensive. Nevertheless, recent advancements such as the optimization of robust protein synthesis platforms, high-throughput immunoscreening approaches, reverse vaccinology, structural design of immunogens, lymphocyte repertoire sequencing, and the utilization of artificial intelligence, have renewed the prospects of an accelerated discovery of the key antigens in malaria. A deeper understanding of the major factors underlying the immunological and molecular mechanisms of malaria might provide a comprehensive approach to identifying novel and highly efficacious vaccines. In this review we discuss progress in novel antigen discoveries that leverage on the wheat germ cell-free protein synthesis system (WGCFS) to accelerate malaria vaccine development.
Collapse
Affiliation(s)
- Bernard N Kanoi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan
| | - Hikaru Nagaoka
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan
| | - Masayuki Morita
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan
| | - Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan.
| |
Collapse
|
12
|
Gnidehou S, Yanow SK. VAR2CSA Antibodies in Non-Pregnant Populations. Trends Parasitol 2020; 37:65-76. [PMID: 33067131 DOI: 10.1016/j.pt.2020.09.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/16/2020] [Accepted: 09/18/2020] [Indexed: 11/18/2022]
Abstract
The Plasmodium falciparum protein VAR2CSA is a critical mediator of placental malaria, and VAR2CSA antibodies (IgGs) are important to protect pregnant women. Although infrequently detected outside pregnancy, VAR2CSA IgGs were reported in men and children from Colombia and Brazil and in select African populations. These findings raise questions about the specificity of VAR2CSA IgGs and the mechanisms by which they are acquired outside pregnancy. Here we review the data on VAR2CSA IgGs in men and children from different malaria-endemic regions. We discuss experimental factors that may affect interpretation of the serological data and consider the biological relevance of VAR2CSA IgGs in non-pregnant populations. We propose potential mechanisms for the acquisition of VARCSA IgGs outside of pregnancy. We identify knowledge gaps and research priorities.
Collapse
Affiliation(s)
- Sedami Gnidehou
- Campus Saint-Jean, University of Alberta, Edmonton, AB, Canada; Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada.
| | - Stephanie K Yanow
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada; School of Public Health, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
13
|
Doritchamou JYA, Morrison R, Renn JP, Ribeiro J, Duan J, Fried M, Duffy PE. Placental malaria vaccine candidate antigen VAR2CSA displays atypical domain architecture in some Plasmodium falciparum strains. Commun Biol 2019; 2:457. [PMID: 31840102 PMCID: PMC6897902 DOI: 10.1038/s42003-019-0704-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 11/13/2019] [Indexed: 12/15/2022] Open
Abstract
Two vaccines based on Plasmodium falciparum protein VAR2CSA are currently in clinical evaluation to prevent placental malaria (PM), but a deeper understanding of var2csa variability could impact vaccine design. Here we identified atypical extended or truncated VAR2CSA extracellular structures and confirmed one extended structure in a Malian maternal isolate, using a novel protein fragment assembly method for RNA-seq and DNA-seq data. Extended structures included one or two additional DBL domains downstream of the conventional NTS-DBL1X-6ɛ domain structure, with closest similarity to DBLɛ in var2csa and non-var2csa genes. Overall, 4/82 isolates displayed atypical VAR2CSA structures. The maternal isolate expressing an extended VAR2CSA bound to CSA, but its recombinant VAR2CSA bound less well to CSA than VAR2CSANF54 and showed lower reactivity to naturally acquired parity-dependent antibody. Our protein fragment sequence assembly approach has revealed atypical VAR2CSA domain architectures that impact antigen reactivity and function, and should inform the design of VAR2CSA-based vaccines.
Collapse
Affiliation(s)
- Justin Y. A. Doritchamou
- Laboratory of Malaria Immunology & Vaccinology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD USA
| | - Robert Morrison
- Laboratory of Malaria Immunology & Vaccinology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD USA
| | - Jonathan P. Renn
- Laboratory of Malaria Immunology & Vaccinology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD USA
| | - Jose Ribeiro
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD USA
| | - Junhui Duan
- Laboratory of Malaria Immunology & Vaccinology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD USA
| | - Michal Fried
- Laboratory of Malaria Immunology & Vaccinology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD USA
| | - Patrick E. Duffy
- Laboratory of Malaria Immunology & Vaccinology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD USA
| |
Collapse
|
14
|
Moxon CA, Gibbins MP, McGuinness D, Milner DA, Marti M. New Insights into Malaria Pathogenesis. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2019; 15:315-343. [PMID: 31648610 DOI: 10.1146/annurev-pathmechdis-012419-032640] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Malaria remains a major public health threat in tropical and subtropical regions across the world. Even though less than 1% of malaria infections are fatal, this leads to about 430,000 deaths per year, predominantly in young children in sub-Saharan Africa. Therefore, it is imperative to understand why a subset of infected individuals develop severe syndromes and some of them die and what differentiates these cases from the majority that recovers. Here, we discuss progress made during the past decade in our understanding of malaria pathogenesis, focusing on the major human parasite Plasmodium falciparum.
Collapse
Affiliation(s)
- Christopher A Moxon
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, United Kingdom; ,
| | - Matthew P Gibbins
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, United Kingdom; ,
| | - Dagmara McGuinness
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, United Kingdom; ,
| | - Danny A Milner
- American Society for Clinical Pathology, Chicago, Illinois 60603, USA.,Department of Immunology and Infectious Disease, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Matthias Marti
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, United Kingdom; , .,Department of Immunology and Infectious Disease, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| |
Collapse
|
15
|
Phosphorylation of the VAR2CSA extracellular region is associated with enhanced adhesive properties to the placental receptor CSA. PLoS Biol 2019; 17:e3000308. [PMID: 31181082 PMCID: PMC6586358 DOI: 10.1371/journal.pbio.3000308] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 06/20/2019] [Accepted: 05/16/2019] [Indexed: 12/26/2022] Open
Abstract
Plasmodium falciparum is the main cause of disease and death from malaria. P. falciparum virulence resides in the ability of infected erythrocytes (IEs) to sequester in various tissues through the interaction between members of the polymorphic P. falciparum erythrocyte membrane protein 1 (PfEMP1) adhesin family to various host receptors. Here, we investigated the effect of phosphorylation of variant surface antigen 2-CSA (VAR2CSA), a member of the PfEMP1 family associated to placental sequestration, on its capacity to adhere to chondroitin sulfate A (CSA) present on the placental syncytium. We showed that phosphatase treatment of IEs impairs cytoadhesion to CSA. MS analysis of recombinant VAR2CSA phosphosites prior to and after phosphatase treatment, as well as of native VAR2CSA expressed on IEs, identified critical phosphoresidues associated with CSA binding. Site-directed mutagenesis on recombinant VAR2CSA of 3 phosphoresidues localised within the CSA-binding region confirmed in vitro their functional importance. Furthermore, using clustered regularly interspaced short palindromic repeats/CRISPR-associated protein-9 nuclease (CRISPR/Cas9), we generated a parasite line in which the phosphoresidue T934 is changed to alanine and showed that this mutation strongly impairs IEs cytoadhesion to CSA. Taken together, these results demonstrate that phosphorylation of the extracellular region of VAR2CSA plays a major role in IEs cytoadhesion to CSA and provide new molecular insights for strategies aiming to reduce the morbidity and mortality of PM.
Collapse
|
16
|
Salinas ND, Tang WK, Tolia NH. Blood-Stage Malaria Parasite Antigens: Structure, Function, and Vaccine Potential. J Mol Biol 2019; 431:4259-4280. [PMID: 31103771 DOI: 10.1016/j.jmb.2019.05.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 04/22/2019] [Accepted: 05/08/2019] [Indexed: 10/26/2022]
Abstract
Plasmodium parasites are the causative agent of malaria, a disease that kills approximately 450,000 individuals annually, with the majority of deaths occurring in children under the age of 5 years and the development of a malaria vaccine is a global health priority. Plasmodium parasites undergo a complex life cycle requiring numerous diverse protein families. The blood stage of parasite development results in the clinical manifestation of disease. A vaccine that disrupts the blood stage is highly desired and will aid in the control of malaria. The blood stage comprises multiple steps: invasion of, asexual growth within, and egress from red blood cells. This review focuses on blood-stage antigens with emphasis on antigen structure, antigen function, neutralizing antibodies, and vaccine potential.
Collapse
Affiliation(s)
- Nichole D Salinas
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD,, 20892, USA
| | - Wai Kwan Tang
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD,, 20892, USA
| | - Niraj H Tolia
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD,, 20892, USA.
| |
Collapse
|
17
|
Seitz J, Morales-Prieto DM, Favaro RR, Schneider H, Markert UR. Molecular Principles of Intrauterine Growth Restriction in Plasmodium Falciparum Infection. Front Endocrinol (Lausanne) 2019; 10:98. [PMID: 30930847 PMCID: PMC6405475 DOI: 10.3389/fendo.2019.00098] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 02/01/2019] [Indexed: 12/21/2022] Open
Abstract
Malaria in pregnancy still constitutes a particular medical challenge in tropical and subtropical regions. Of the five Plasmodium species that are pathogenic to humans, infection with Plasmodium falciparum leads to fulminant progression of the disease with massive impact on pregnancy. Severe anemia of the mother, miscarriage, stillbirth, preterm delivery and intrauterine growth restriction (IUGR) with reduced birth weight are frequent complications that lead to more than 10,000 maternal and 200,000 perinatal deaths annually in sub-Saharan Africa alone. P. falciparum can adhere to the placenta via the expression of the surface antigen VAR2CSA, which leads to sequestration of infected erythrocytes in the intervillous space. This process induces a placental inflammation with involvement of immune cells and humoral factors. Especially, monocytes get activated and change the release of soluble mediators, including a variety of cytokines. This proinflammatory environment contributes to disorders of angiogenesis, blood flow, autophagy, and nutrient transport in the placenta and erythropoiesis. Collectively, they impair placental functions and, consequently, fetal growth. The discovery that women in endemic regions develop a certain immunity against VAR2CSA-expressing parasites with increasing number of pregnancies has redefined the understanding of malaria in pregnancy and offers strategies for the development of vaccines. The following review gives an overview of molecular processes in P. falciparum infection in pregnancy which may be involved in the development of IUGR.
Collapse
Affiliation(s)
- Johanna Seitz
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany
| | | | - Rodolfo R. Favaro
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany
| | - Henning Schneider
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
- Department of Obstetrics and Gynecology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Udo Rudolf Markert
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany
| |
Collapse
|
18
|
Frimpong A, Kusi KA, Ofori MF, Ndifon W. Novel Strategies for Malaria Vaccine Design. Front Immunol 2018; 9:2769. [PMID: 30555463 PMCID: PMC6281765 DOI: 10.3389/fimmu.2018.02769] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 11/12/2018] [Indexed: 12/19/2022] Open
Abstract
The quest for a licensed effective vaccine against malaria remains a global priority. Even though classical vaccine design strategies have been successful for some viral and bacterial pathogens, little success has been achieved for Plasmodium falciparum, which causes the deadliest form of malaria due to its diversity and ability to evade host immune responses. Nevertheless, recent advances in vaccinology through high throughput discovery of immune correlates of protection, lymphocyte repertoire sequencing and structural design of immunogens, provide a comprehensive approach to identifying and designing a highly efficacious vaccine for malaria. In this review, we discuss novel vaccine approaches that can be employed in malaria vaccine design.
Collapse
Affiliation(s)
- Augustina Frimpong
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana.,Immunology Department, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana.,African Institute for Mathematical Sciences, Cape Coast, Ghana
| | - Kwadwo Asamoah Kusi
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana.,Immunology Department, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Michael Fokuo Ofori
- Department of Biochemistry, Cell and Molecular Biology, West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana.,Immunology Department, College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Wilfred Ndifon
- African Institute for Mathematical Sciences, Cape Coast, Ghana.,African Institute for Mathematical Sciences, University of Stellenbosch, Cape Town, South Africa
| |
Collapse
|
19
|
Ubillos I, Jiménez A, Vidal M, Bowyer PW, Gaur D, Dutta S, Gamain B, Coppel R, Chauhan V, Lanar D, Chitnis C, Angov E, Beeson J, Cavanagh D, Campo JJ, Aguilar R, Dobaño C. Optimization of incubation conditions of Plasmodium falciparum antibody multiplex assays to measure IgG, IgG 1-4, IgM and IgE using standard and customized reference pools for sero-epidemiological and vaccine studies. Malar J 2018; 17:219. [PMID: 29859096 PMCID: PMC5984756 DOI: 10.1186/s12936-018-2369-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/28/2018] [Indexed: 12/15/2022] Open
Abstract
Background The quantitative suspension array technology (qSAT) is a useful platform for malaria immune marker discovery. However, a major challenge for large sero-epidemiological and malaria vaccine studies is the comparability across laboratories, which requires the access to standardized control reagents for assay optimization, to monitor performance and improve reproducibility. Here, the Plasmodium falciparum antibody reactivities of the newly available WHO reference reagent for anti-malaria human plasma (10/198) and of additional customized positive controls were examined with seven in-house qSAT multiplex assays measuring IgG, IgG1–4 subclasses, IgM and IgE against a panel of 40 antigens. The different positive controls were tested at different incubation times and temperatures (4 °C overnight, 37 °C 2 h, room temperature 1 h) to select the optimal conditions. Results Overall, the WHO reference reagent had low IgG2, IgG4, IgM and IgE, and also low anti-CSP antibody levels, thus this reagent was enriched with plasmas from RTS,S-vaccinated volunteers to be used as standard for CSP-based vaccine studies. For the IgM assay, another customized plasma pool prepared with samples from malaria primo-infected adults with adequate IgM levels proved to be more adequate as a positive control. The range and magnitude of IgG and IgG1–4 responses were highest when the WHO reference reagent was incubated with antigen-coupled beads at 4 °C overnight. IgG levels measured in the negative control did not vary between incubations at 37 °C 2 h and 4 °C overnight, indicating no difference in unspecific binding. Conclusions With this study, the immunogenicity profile of the WHO reference reagent, including seven immunoglobulin isotypes and subclasses, and more P. falciparum antigens, also those included in the leading RTS,S malaria vaccine, was better characterized. Overall, incubation of samples at 4 °C overnight rendered the best performance for antibody measurements against the antigens tested. Although the WHO reference reagent performed well to measure IgG to the majority of the common P. falciparum blood stage antigens tested, customized pools may need to be used as positive controls depending on the antigens (e.g. pre-erythrocytic proteins of low natural immunogenicity) and isotypes/subclasses (e.g. IgM) under study. Electronic supplementary material The online version of this article (10.1186/s12936-018-2369-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Itziar Ubillos
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Carrer Rosselló 153 (CEK Building), 08036, Barcelona, Catalonia, Spain
| | - Alfons Jiménez
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Carrer Rosselló 153 (CEK Building), 08036, Barcelona, Catalonia, Spain.,CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Marta Vidal
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Carrer Rosselló 153 (CEK Building), 08036, Barcelona, Catalonia, Spain
| | - Paul W Bowyer
- Bacteriology Division, MHRA-NIBSC, South Mimms, Potter Bars, EN6 3QG, UK
| | - Deepak Gaur
- Laboratory of Malaria and Vaccine Research, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India.,Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Sheetij Dutta
- U.S. Military Malaria Vaccine Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Benoit Gamain
- Université Sorbonne Paris Cité, Université Paris Diderot, Inserm, INTS, Unité Biologie Intégrée du Globule Rouge UMR_S1134, Laboratoire d'Excellence GR-Ex, Paris, France
| | - Ross Coppel
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - Virander Chauhan
- Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - David Lanar
- U.S. Military Malaria Vaccine Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Chetan Chitnis
- Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Evelina Angov
- U.S. Military Malaria Vaccine Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - James Beeson
- Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, VIC, Australia
| | - David Cavanagh
- Institute of Immunology & Infection Research and Centre for Immunity, Infection & Evolution, Ashworth Laboratories, School of Biological Sciences, University of Edinburgh, King's Buildings, Charlotte Auerbach Rd, Edinburgh, EH9 3FL, UK
| | - Joseph J Campo
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Carrer Rosselló 153 (CEK Building), 08036, Barcelona, Catalonia, Spain
| | - Ruth Aguilar
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Carrer Rosselló 153 (CEK Building), 08036, Barcelona, Catalonia, Spain
| | - Carlota Dobaño
- ISGlobal, Hospital Clínic-Universitat de Barcelona, Carrer Rosselló 153 (CEK Building), 08036, Barcelona, Catalonia, Spain.
| |
Collapse
|
20
|
Plasmodium falciparum PfEMP1 Modulates Monocyte/Macrophage Transcription Factor Activation and Cytokine and Chemokine Responses. Infect Immun 2017; 86:IAI.00447-17. [PMID: 29038124 PMCID: PMC5736827 DOI: 10.1128/iai.00447-17] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 10/03/2017] [Indexed: 12/11/2022] Open
Abstract
Immunity to Plasmodium falciparum malaria is slow to develop, and it is often asserted that malaria suppresses host immunity, although this is poorly understood and the molecular basis for such activity remains unknown. P. falciparum erythrocyte membrane protein 1 (PfEMP1) is a virulence factor that plays a key role in parasite-host interactions. We investigated the immunosuppressive effect of PfEMP1 on monocytes/macrophages, which are central to the antiparasitic innate response. RAW macrophages and human primary monocytes were stimulated with wild-type 3D7 or CS2 parasites or transgenic PfEMP1-null parasites. To study the immunomodulatory effect of PfEMP1, transcription factor activation and cytokine and chemokine responses were measured. The level of activation of NF-κB was significantly lower in macrophages stimulated with parasites that express PfEMP1 at the red blood cell surface membrane than in macrophages stimulated with PfEMP1-null parasites. Modulation of additional transcription factors, including CREB, also occurred, resulting in reduced immune gene expression and decreased tumor necrosis factor (TNF) and interleukin-10 (IL-10) release. Similarly, human monocytes released less IL-1β, IL-6, IL-10, monocyte chemoattractant protein 1 (MCP-1), macrophage inflammatory protein 1α (MIP-1α), MIP-1β, and TNF specifically in response to VAR2CSA PfEMP1-containing parasites than in response to PfEMP1-null parasites, suggesting that this immune regulation by PfEMP1 is important in naturally occurring infections. These results indicate that PfEMP1 is an immunomodulatory molecule that affects the activation of a range of transcription factors, dampening cytokine and chemokine responses. Therefore, these findings describe a potential molecular basis for immune suppression by P. falciparum.
Collapse
|
21
|
Abstract
Parasites of the genus Plasmodium have a complex life cycle. They alternate between their final mosquito host and their intermediate hosts. The parasite can be either extra- or intracellular, depending on the stage of development. By modifying their shape, motility, and metabolic requirements, the parasite adapts to the different environments in their different hosts. The parasite has evolved to escape the multiple immune mechanisms in the host that try to block parasite development at the different stages of their development. In this article, we describe the mechanisms reported thus far that allow the Plasmodium parasite to evade innate and adaptive immune responses.
Collapse
Affiliation(s)
- Laurent Rénia
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Yun Shan Goh
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore
| |
Collapse
|