1
|
Tumbo A, Lorenz FR, Yang ASP, Sefried S, Schindler T, Mpina M, Dangy JP, Milando FA, Rashid MA, Nyaulingo G, Ramadhani K, Jongo S, Felgner PL, Abebe Y, Sim BKL, Church LWP, Richie TL, Billingsley PF, Murshedkar T, Hoffman SL, Abdulla S, Kremsner PG, Mordmüller B, Daubenberger C, Fendel R. PfSPZ Vaccine induces focused humoral immune response in HIV positive and negative Tanzanian adults. EBioMedicine 2024; 108:105364. [PMID: 39353279 PMCID: PMC11464252 DOI: 10.1016/j.ebiom.2024.105364] [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: 01/17/2023] [Revised: 09/11/2024] [Accepted: 09/11/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND PfSPZ Vaccine, a promising pre-erythrocytic stage malaria vaccine candidate based on whole, radiation-attenuated Plasmodium falciparum (Pf) sporozoites (SPZ), has proven safe and effective in mediating sterile protection from malaria in malaria-naïve and exposed healthy adults. Vaccine-induced protection presumably depends on cellular responses to early parasite liver stages, but humoral immunity contributes. METHODS On custom-made Pf protein microarrays, we profiled IgG and IgM responses to PfSPZ Vaccine and subsequent homologous controlled human malaria infection (CHMI) in 21 Tanzanian adults with (n = 12) or without (n = 9) HIV infection. Expression of the main identified immunogens in the pre-erythrocytic parasite stage was verified by immunofluorescence detection using freshly purified PfSPZ and an in vitro model of primary human hepatocytes. FINDINGS Independent of HIV infection status, immunisation induced focused IgG and IgM responses to circumsporozoite surface protein (PfCSP) and merozoite surface protein 5 (PfMSP5). We show that PfMSP5 is detectable on the surface and in the apical complex of PfSPZ. INTERPRETATION Our data demonstrate that HIV infection does not affect the quantity of the total IgG and IgM antibody responses to PfCSP and PfMSP5 after immunization with PfSPZ Vaccine. PfMSP5 represents a highly immunogenic, so far underexplored, target for vaccine-induced antibodies in malaria pre-exposed volunteers. FUNDING This work was supported by the Equatorial Guinea Malaria Vaccine Initiative (EGMVI), the Clinical Trial Platform of the German Center for Infection Research (TTU 03.702), the Swiss Government Excellence Scholarships for Foreign Scholars and Artists (grant 2016.0056) and the Interdisciplinary Center for Clinical Research doctoral program of the Tübingen University Hospital. The funders had no role in design, analysis, or reporting of this study.
Collapse
Affiliation(s)
- Anneth Tumbo
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland; Ifakara Health Institute, Bagamoyo Branch, Bagamoyo, United Republic of Tanzania
| | - Freia-Raphaella Lorenz
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany; Radboud Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands; German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
| | - Annie S P Yang
- Radboud Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Stephanie Sefried
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany; German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany
| | - Tobias Schindler
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland
| | - Maximilian Mpina
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland; Ifakara Health Institute, Bagamoyo Branch, Bagamoyo, United Republic of Tanzania
| | - Jean-Pierre Dangy
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland
| | - Florence A Milando
- Ifakara Health Institute, Bagamoyo Branch, Bagamoyo, United Republic of Tanzania
| | - Mohammed A Rashid
- Ifakara Health Institute, Bagamoyo Branch, Bagamoyo, United Republic of Tanzania
| | - Gloria Nyaulingo
- Ifakara Health Institute, Bagamoyo Branch, Bagamoyo, United Republic of Tanzania
| | - Kamaka Ramadhani
- Ifakara Health Institute, Bagamoyo Branch, Bagamoyo, United Republic of Tanzania
| | - Said Jongo
- Ifakara Health Institute, Bagamoyo Branch, Bagamoyo, United Republic of Tanzania
| | | | - Yonas Abebe
- Sanaria Inc., Rockville, Maryland, United States
| | | | | | | | | | | | | | - Salim Abdulla
- Ifakara Health Institute, Bagamoyo Branch, Bagamoyo, United Republic of Tanzania
| | - Peter G Kremsner
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany; German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany; Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Benjamin Mordmüller
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany; Radboud Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands; Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Claudia Daubenberger
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland.
| | - Rolf Fendel
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany; German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany; Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon.
| |
Collapse
|
2
|
Zhou X, Zhang Q, Chen JH, Dai JF, Kassegne K. Revisiting the antigen markers of vector-borne parasitic diseases identified by immunomics: identification and application to disease control. Expert Rev Proteomics 2024; 21:205-216. [PMID: 38584506 DOI: 10.1080/14789450.2024.2336994] [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: 09/05/2023] [Accepted: 03/03/2024] [Indexed: 04/09/2024]
Abstract
INTRODUCTION Protein microarray is a promising immunomic approach for identifying biomarkers. Based on our previous study that reviewed parasite antigens and recent parasitic omics research, this article expands to include information on vector-borne parasitic diseases (VBPDs), namely, malaria, schistosomiasis, leishmaniasis, babesiosis, trypanosomiasis, lymphatic filariasis, and onchocerciasis. AREAS COVERED We revisit and systematically summarize antigen markers of vector-borne parasites identified by the immunomic approach and discuss the latest advances in identifying antigens for the rational development of diagnostics and vaccines. The applications and challenges of this approach for VBPD control are also discussed. EXPERT OPINION The immunomic approach has enabled the identification and/or validation of antigen markers for vaccine development, diagnosis, disease surveillance, and treatment. However, this approach presents several challenges, including limited sample size, variability in antigen expression, false-positive results, complexity of omics data, validation and reproducibility, and heterogeneity of diseases. In addition, antigen involvement in host immune evasion and antigen sensitivity/specificity are major issues in its application. Despite these limitations, this approach remains promising for controlling VBPD. Advances in technology and data analysis methods should continue to improve candidate antigen identification, as well as the use of a multiantigen approach in diagnostic and vaccine development for VBPD control.
Collapse
Affiliation(s)
- Xia Zhou
- MOE Key Laboratory of Geriatric Diseases and Immunology, School of Biology & Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, China
| | - Qianqian Zhang
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
| | - Jun-Hu Chen
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute of Parasitic Diseases, Chinese Center for Diseases Control and Prevention (Chinese Center for Tropical Diseases Research); National Health Commission of the People's Republic of China (NHC) Key Laboratory of Parasite and Vector Biology; World Health Organization (WHO) Collaborating Center for Tropical Diseases; National Center for International Research on Tropical Diseases, Shanghai, People's Republic of China
- Hainan Tropical Diseases Research Center (Hainan Sub-Center, Chinese Center for Tropical Diseases Research), Haikou, China
| | - Jian-Feng Dai
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
| | - Kokouvi Kassegne
- School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- One Health Center, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
3
|
Lopez-Perez M, Jain A, Davies DH, Vásquez-Jiménez JM, Herrera SM, Oñate J, Felgner PL, Herrera S, Arévalo-Herrera M. Profiling the antibody response of humans protected by immunization with Plasmodium vivax radiation-attenuated sporozoites. Sci Rep 2024; 14:2790. [PMID: 38307966 PMCID: PMC10837454 DOI: 10.1038/s41598-024-53175-0] [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: 10/28/2023] [Accepted: 01/29/2024] [Indexed: 02/04/2024] Open
Abstract
Malaria sterile immunity has been reproducibly induced by immunization with Plasmodium radiation-attenuated sporozoites (RAS). Analyses of sera from RAS-immunized individuals allowed the identification of P. falciparum antigens, such as the circumsporozoite protein (CSP), the basis for the RTS, S and R21Matrix-M vaccines. Similar advances in P. vivax (Pv) vaccination have been elusive. We previously reported 42% (5/12) of sterile protection in malaria-unexposed, Duffy-positive (Fy +) volunteers immunized with PvRAS followed by a controlled human malaria infection (CHMI). Using a custom protein microarray displaying 515 Pv antigens, we found a significantly higher reactivity to PvCSP and one hypothetical protein (PVX_089630) in volunteers protected against P. vivax infection. In mock-vaccinated Fy + volunteers, a strong antibody response to CHMI was also observed. Although the Fy- volunteers immunized with non-irradiated Pv-infected mosquitoes (live sporozoites) did not develop malaria after CHMI, they recognized a high number of antigens, indicating the temporary presence of asexual parasites in peripheral blood. Together, our findings contribute to the understanding of the antibody response to P. vivax infection and allow the identification of novel parasite antigens as vaccine candidates.Trial registration: ClinicalTrials.gov number: NCT01082341.
Collapse
Affiliation(s)
- Mary Lopez-Perez
- Malaria Vaccine and Drug Development Center (MVDC), Cali, Colombia
| | - Aarti Jain
- Department Physiology & Biophysics, Vaccine R&D Center, University of California Irvine, Irvine, CA, USA
| | - D Huw Davies
- Department Physiology & Biophysics, Vaccine R&D Center, University of California Irvine, Irvine, CA, USA
| | | | | | | | - Philip L Felgner
- Department Physiology & Biophysics, Vaccine R&D Center, University of California Irvine, Irvine, CA, USA
| | - Sócrates Herrera
- Malaria Vaccine and Drug Development Center (MVDC), Cali, Colombia
- Caucaseco Scientific Research Center, Cali, Colombia
| | - Myriam Arévalo-Herrera
- Malaria Vaccine and Drug Development Center (MVDC), Cali, Colombia.
- Caucaseco Scientific Research Center, Cali, Colombia.
| |
Collapse
|
4
|
Momoh EO, Ghag SK, White J, Mudeppa DG, Rathod PK. Multiplex Assays for Analysis of Antibody Responses to South Asian Plasmodium falciparum and Plasmodium vivax Malaria Infections. Vaccines (Basel) 2023; 12:1. [PMID: 38276660 PMCID: PMC10818873 DOI: 10.3390/vaccines12010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 12/01/2023] [Accepted: 12/13/2023] [Indexed: 01/27/2024] Open
Abstract
Malaria remains a major global health challenge, causing over 0.6 million yearly deaths. To understand naturally acquired immunity in adult human populations in malaria-prevalent regions, improved serological tools are needed, particularly where multiple malaria parasite species co-exist. Slide-based and bead-based multiplex approaches can help characterize antibodies in malaria patients from endemic regions, but these require pure, well-defined antigens. To efficiently bypass purification steps, codon-optimized malaria antigen genes with N-terminal FLAG-tag and C-terminal Ctag sequences were expressed in a wheat germ cell-free system and adsorbed on functionalized BioPlex beads. In a pilot study, 15 P. falciparum antigens, 8 P. vivax antigens, and a negative control (GFP) were adsorbed individually on functionalized bead types through their Ctag. To validate the multiplexing powers of this platform, 10 P. falciparum-infected patient sera from a US NIH MESA-ICEMR study site in Goa, India, were tested against all 23 parasite antigens. Serial dilution of patient sera revealed variations in potency and breadth of antibodies to various parasite antigens. Individual patients revealed informative variations in immunity to P. falciparum versus P. vivax. This multiplex approach to malaria serology captures varying immunity to different human malaria parasite species and different parasite antigens. This approach can be scaled to track the dynamics of antibody production during one or more human malaria infections.
Collapse
Affiliation(s)
| | | | | | - Devaraja G. Mudeppa
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA; (E.O.M.); (S.K.G.); (J.W.)
| | - Pradipsinh K. Rathod
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA; (E.O.M.); (S.K.G.); (J.W.)
| |
Collapse
|
5
|
Furtado R, Paul M, Zhang J, Sung J, Karell P, Kim RS, Caillat-Zucman S, Liang L, Felgner P, Bauleni A, Gama S, Buchwald A, Taylor T, Seydel K, Laufer M, Delahaye F, Daily JP, Lauvau G. Cytolytic circumsporozoite-specific memory CD4 + T cell clones are expanded during Plasmodium falciparum infection. Nat Commun 2023; 14:7726. [PMID: 38001069 PMCID: PMC10673885 DOI: 10.1038/s41467-023-43376-y] [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: 08/02/2021] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Clinical immunity against Plasmodium falciparum infection develops in residents of malaria endemic regions, manifesting in reduced clinical symptoms during infection and in protection against severe disease but the mechanisms are not fully understood. Here, we compare the cellular and humoral immune response of clinically immune (0-1 episode over 18 months) and susceptible (at least 3 episodes) during a mild episode of Pf malaria infection in a malaria endemic region of Malawi, by analysing peripheral blood samples using high dimensional mass cytometry (CyTOF), spectral flow cytometry and single-cell transcriptomic analyses. In the clinically immune, we find increased proportions of circulating follicular helper T cells and classical monocytes, while the humoral immune response shows characteristic age-related differences in the protected. Presence of memory CD4+ T cell clones with a strong cytolytic ZEB2+ T helper 1 effector signature, sharing identical T cell receptor clonotypes and recognizing the Pf-derived circumsporozoite protein (CSP) antigen are found in the blood of the Pf-infected participants gaining protection. Moreover, in clinically protected participants, ZEB2+ memory CD4+ T cells express lower level of inhibitory and chemotactic receptors. We thus propose that clonally expanded ZEB2+ CSP-specific cytolytic memory CD4+ Th1 cells may contribute to clinical immunity against the sporozoite and liver-stage Pf malaria.
Collapse
Affiliation(s)
- Raquel Furtado
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
- RF: BioNTech US, 40 Erie Street, Cambridge, MA, 02139, USA
| | - Mahinder Paul
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Jinghang Zhang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Joowhan Sung
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Paul Karell
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Ryung S Kim
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Sophie Caillat-Zucman
- Université de Paris, AP-HP, Hôpital Saint-Louis, Laboratoire d'Immunologie et Histocompatiblité, INSERM UMR976, 75010, Paris, France
| | - Li Liang
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Philip Felgner
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Andy Bauleni
- Malaria Alert Centre, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Syze Gama
- Blantyre Malaria Project, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Andrea Buchwald
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Terrie Taylor
- Blantyre Malaria Project, Kamuzu University of Health Sciences, Blantyre, Malawi
- Department of Osteopathic Medical Specialties, Michigan State University, East Lansing, MI, 48824, USA
| | - Karl Seydel
- Blantyre Malaria Project, Kamuzu University of Health Sciences, Blantyre, Malawi
- Department of Osteopathic Medical Specialties, Michigan State University, East Lansing, MI, 48824, USA
| | - Miriam Laufer
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Fabien Delahaye
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
- FD: Precision Oncology, Sanofi, Vitry sur Seine, France
| | - Johanna P Daily
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, 10461, USA.
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, 10461, USA.
| | - Grégoire Lauvau
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, 10461, USA.
| |
Collapse
|
6
|
Hassan I, Kanoi BN, Nagaoka H, Sattabongkot J, Udomsangpetch R, Tsuboi T, Takashima E. High-Throughput Antibody Profiling Identifies Targets of Protective Immunity against P. falciparum Malaria in Thailand. Biomolecules 2023; 13:1267. [PMID: 37627332 PMCID: PMC10452476 DOI: 10.3390/biom13081267] [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: 06/12/2023] [Revised: 08/09/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Malaria poses a significant global health challenge, resulting in approximately 600,000 deaths each year. Individuals living in regions with endemic malaria have the potential to develop partial immunity, thanks in part to the presence of anti-plasmodium antibodies. As efforts are made to optimize and implement strategies to reduce malaria transmission and ultimately eliminate the disease, it is crucial to understand how these interventions impact naturally acquired protective immunity. To shed light on this, our study focused on assessing antibody responses to a carefully curated library of P. falciparum recombinant proteins (n = 691) using samples collected from individuals residing in a low-malaria-transmission region of Thailand. We conducted the antibody assays using the AlphaScreen system, a high-throughput homogeneous proximity-based bead assay that detects protein interactions. We observed that out of the 691 variable surface and merozoite stage proteins included in the library, antibodies to 268 antigens significantly correlated with the absence of symptomatic malaria in an univariate analysis. Notably, the most prominent antigens identified were P. falciparum erythrocyte membrane protein 1 (PfEMP1) domains. These results align with our previous research conducted in Uganda, suggesting that similar antigens like PfEMP1s might play a pivotal role in determining infection outcomes in diverse populations. To further our understanding, it remains critical to conduct functional characterization of these identified proteins, exploring their potential as correlates of protection or as targets for vaccine development.
Collapse
Affiliation(s)
- Ifra Hassan
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama 790-8577, Japan; (I.H.); (H.N.)
| | - Bernard N. Kanoi
- Centre for Malaria Elimination, Institute of Tropical Medicine, Mount Kenya University, Thika 01000, Kenya;
| | - Hikaru Nagaoka
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama 790-8577, Japan; (I.H.); (H.N.)
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Rachanee Udomsangpetch
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Salaya, Nakhon Pathom 73170, Thailand;
| | - Takafumi Tsuboi
- Division of Cell-Free Sciences, Proteo-Science Center, Ehime University, Matsuyama 790-8577, Japan;
| | - Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama 790-8577, Japan; (I.H.); (H.N.)
| |
Collapse
|
7
|
Mbambo G, Dwivedi A, Ifeonu OO, Munro JB, Shrestha B, Bromley RE, Hodges T, Adkins RS, Kouriba B, Diarra I, Niangaly A, Kone AK, Coulibaly D, Traore K, Dolo A, Thera MA, Laurens MB, Doumbo OK, Plowe CV, Berry AA, Travassos M, Lyke KE, Silva JC. Immunogenomic profile at baseline predicts host susceptibility to clinical malaria. Front Immunol 2023; 14:1179314. [PMID: 37465667 PMCID: PMC10351378 DOI: 10.3389/fimmu.2023.1179314] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/19/2023] [Indexed: 07/20/2023] Open
Abstract
Introduction Host gene and protein expression impact susceptibility to clinical malaria, but the balance of immune cell populations, cytokines and genes that contributes to protection, remains incompletely understood. Little is known about the determinants of host susceptibility to clinical malaria at a time when acquired immunity is developing. Methods We analyzed peripheral blood mononuclear cells (PBMCs) collected from children who differed in susceptibility to clinical malaria, all from a small town in Mali. PBMCs were collected from children aged 4-6 years at the start, peak and end of the malaria season. We characterized the immune cell composition and cytokine secretion for a subset of 20 children per timepoint (10 children with no symptomatic malaria age-matched to 10 children with >2 symptomatic malarial illnesses), and gene expression patterns for six children (three per cohort) per timepoint. Results We observed differences between the two groups of children in the expression of genes related to cell death and inflammation; in particular, inflammatory genes such as CXCL10 and STAT1 and apoptotic genes such as XAF1 were upregulated in susceptible children before the transmission season began. We also noted higher frequency of HLA-DR+ CD4 T cells in protected children during the peak of the malaria season and comparable levels cytokine secretion after stimulation with malaria schizonts across all three time points. Conclusion This study highlights the importance of baseline immune signatures in determining disease outcome. Our data suggests that differences in apoptotic and inflammatory gene expression patterns can serve as predictive markers of susceptibility to clinical malaria.
Collapse
Affiliation(s)
- Gillian Mbambo
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Ankit Dwivedi
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Olukemi O. Ifeonu
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - James B. Munro
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Biraj Shrestha
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Robin E. Bromley
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Theresa Hodges
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Ricky S. Adkins
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Bourema Kouriba
- Malaria Research and Training Center, International Centers for Excellence in Research (NIH), University of Science Techniques and Technologies of Bamako, Bamako, Mali
| | - Issa Diarra
- Malaria Research and Training Center, International Centers for Excellence in Research (NIH), University of Science Techniques and Technologies of Bamako, Bamako, Mali
| | - Amadou Niangaly
- Malaria Research and Training Center, International Centers for Excellence in Research (NIH), University of Science Techniques and Technologies of Bamako, Bamako, Mali
| | - Abdoulaye K. Kone
- Malaria Research and Training Center, International Centers for Excellence in Research (NIH), University of Science Techniques and Technologies of Bamako, Bamako, Mali
| | - Drissa Coulibaly
- Malaria Research and Training Center, International Centers for Excellence in Research (NIH), University of Science Techniques and Technologies of Bamako, Bamako, Mali
| | - Karim Traore
- Malaria Research and Training Center, International Centers for Excellence in Research (NIH), University of Science Techniques and Technologies of Bamako, Bamako, Mali
| | - Amagana Dolo
- Malaria Research and Training Center, International Centers for Excellence in Research (NIH), University of Science Techniques and Technologies of Bamako, Bamako, Mali
| | - Mahamadou A. Thera
- Malaria Research and Training Center, International Centers for Excellence in Research (NIH), University of Science Techniques and Technologies of Bamako, Bamako, Mali
| | - Matthew B. Laurens
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Ogobara K. Doumbo
- Malaria Research and Training Center, International Centers for Excellence in Research (NIH), University of Science Techniques and Technologies of Bamako, Bamako, Mali
| | - Christopher V. Plowe
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Andrea A. Berry
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Mark Travassos
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Kirsten E. Lyke
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Joana C. Silva
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
- Global Health and Tropical Medicine, Instituto deHigiene e Medicina Tropical, Universidade Nova de Lisboa (GHTM, IHMT, UNL), Lisboa, Portugal
| |
Collapse
|
8
|
Malaria Vaccines. Infect Dis (Lond) 2023. [DOI: 10.1007/978-1-0716-2463-0_536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
9
|
de Jong RM, Alkema M, Oulton T, Dumont E, Teelen K, Nakajima R, de Assis RR, Press KWD, Ngotho P, Tetteh KK, Felgner P, Marti M, Collins KA, Drakeley C, Bousema T, Stone WJ. The acquisition of humoral immune responses targeting Plasmodium falciparum sexual stages in controlled human malaria infections. Front Immunol 2022; 13:930956. [PMID: 35924245 PMCID: PMC9339717 DOI: 10.3389/fimmu.2022.930956] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Individuals infected with P. falciparum develop antibody responses to intra-erythrocytic gametocyte proteins and exported gametocyte proteins present on the surface of infected erythrocytes. However, there is currently limited knowledge on the immunogenicity of gametocyte antigens and the specificity of gametocyte-induced antibody responses. In this study, we assessed antibody responses in participants of two controlled human malaria infection (CHMI) studies by ELISA, multiplexed bead-based antibody assays and protein microarray. By comparing antibody responses in participants with and without gametocyte exposure, we aimed to disentangle the antibody response induced by asexual and sexual stage parasites. We showed that after a single malaria infection, a significant anti-sexual stage humoral response is induced in malaria-naïve individuals, even after exposure to relatively low gametocyte densities (up to ~1,600 gametocytes/mL). In contrast to antibody responses to well-characterised asexual blood stage antigens that were detectable by day 21 after infection, responses to sexual stage antigens (including transmission blocking vaccine candidates Pfs48/45 and Pfs230) were only apparent at 51 days after infection. We found antigens previously associated with early gametocyte or anti-gamete immunity were highly represented among responses linked with gametocyte exposure. Our data provide detailed insights on the induction and kinetics of antibody responses to gametocytes and identify novel antigens that elicit antibody responses exclusively in individuals with gametocyte exposure. Our findings provide target identification for serological assays for surveillance of the malaria infectious reservoir, and support vaccine development by describing the antibody response to leading vaccine antigens after primary infection.
Collapse
Affiliation(s)
- Roos M. de Jong
- Department of Medical Microbiology and Radboud Centre of Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Manon Alkema
- Department of Medical Microbiology and Radboud Centre of Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Tate Oulton
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Elin Dumont
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Karina Teelen
- Department of Medical Microbiology and Radboud Centre of Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Rie Nakajima
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, United States
| | - Rafael Ramiro de Assis
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, United States
| | | | - Priscilla Ngotho
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Kevin K.A. Tetteh
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Phil Felgner
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, United States
| | - Matthias Marti
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Katharine A. Collins
- Department of Medical Microbiology and Radboud Centre of Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Chris Drakeley
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Teun Bousema
- Department of Medical Microbiology and Radboud Centre of Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands,Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Will J.R. Stone
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom,*Correspondence: Will J.R. Stone,
| |
Collapse
|
10
|
Takashima E, Kanoi BN, Nagaoka H, Morita M, Hassan I, Palacpac NMQ, Egwang TG, Horii T, Gitaka J, Tsuboi T. Meta-Analysis of Human Antibodies Against Plasmodium falciparum Variable Surface and Merozoite Stage Antigens. Front Immunol 2022; 13:887219. [PMID: 35757771 PMCID: PMC9218060 DOI: 10.3389/fimmu.2022.887219] [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: 03/01/2022] [Accepted: 05/11/2022] [Indexed: 11/30/2022] Open
Abstract
Concerted efforts to fight malaria have caused significant reductions in global malaria cases and mortality. Sustaining this will be critical to avoid rebound and outbreaks of seasonal malaria. Identifying predictive attributes that define clinical malaria will be key to guide development of second-generation tools to fight malaria. Broadly reactive antibodies against variable surface antigens that are expressed on the surface of infected erythrocytes and merozoites stage antigens are targets of naturally acquired immunity and prime candidates for anti-malaria therapeutics and vaccines. However, predicting the relationship between the antigen-specific antibodies and protection from clinical malaria remains unresolved. Here, we used new datasets and multiple approaches combined with re-analysis of our previous data to assess the multi-dimensional and complex relationship between antibody responses and clinical malaria outcomes. We observed 22 antigens (17 PfEMP1 domains, 3 RIFIN family members, merozoite surface protein 3 (PF3D7_1035400), and merozoites-associated armadillo repeats protein (PF3D7_1035900) that were selected across three different clinical malaria definitions (1,000/2,500/5,000 parasites/µl plus fever). In addition, Principal Components Analysis (PCA) indicated that the first three components (Dim1, Dim2 and Dim3 with eigenvalues of 306, 48, and 29, respectively) accounted for 66.1% of the total variations seen. Specifically, the Dim1, Dim2 and Dim3 explained 52.8%, 8.2% and 5% of variability, respectively. We further observed a significant relationship between the first component scores and age with antibodies to PfEMP1 domains being the key contributing variables. This is consistent with a recent proposal suggesting that there is an ordered acquisition of antibodies targeting PfEMP1 proteins. Thus, although limited, and further work on the significance of the selected antigens will be required, these approaches may provide insights for identification of drivers of naturally acquired protective immunity as well as guide development of additional tools for malaria elimination and eradication.
Collapse
Affiliation(s)
- Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Bernard N Kanoi
- Centre for Research in Infectious Diseases, Directorate of Research and Innovation, Mount Kenya University, Thika, Kenya
| | - Hikaru Nagaoka
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Masayuki Morita
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Ifra Hassan
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Nirianne M Q Palacpac
- Department of Malaria Vaccine Development, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | | | - Toshihiro Horii
- Department of Malaria Vaccine Development, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Jesse Gitaka
- Centre for Research in Infectious Diseases, Directorate of Research and Innovation, Mount Kenya University, Thika, Kenya
| | - Takafumi Tsuboi
- Division of Cell-Free Sciences, Proteo-Science Center, Ehime University, Matsuyama, Japan
| |
Collapse
|
11
|
Nouatin O, Ibáñez J, Fendel R, Ngoa UA, Lorenz FR, Dejon-Agobé JC, Edoa JR, Flügge J, Brückner S, Esen M, Theisen M, Hoffman SL, Moutairou K, Luty AJF, Lell B, Kremsner PG, Adegnika AA, Mordmüller B. Cellular and antibody response in GMZ2-vaccinated Gabonese volunteers in a controlled human malaria infection trial. Malar J 2022; 21:191. [PMID: 35715803 PMCID: PMC9204906 DOI: 10.1186/s12936-022-04169-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 04/23/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Antibody and cellular memory responses following vaccination are important measures of immunogenicity. These immune markers were quantified in the framework of a vaccine trial investigating the malaria vaccine candidate GMZ2. METHODS Fifty Gabonese adults were vaccinated with two formulations (aluminum Alhydrogel and CAF01) of GMZ2 or a control vaccine (Verorab). Vaccine efficacy was assessed using controlled human malaria infection (CHMI) by direct venous inoculation of 3200 live Plasmodium falciparum sporozoites (PfSPZ Challenge). GMZ2-stimulated T and specific B-cell responses were estimated by flow cytometry before and after vaccination. Additionally, the antibody response against 212 P. falciparum antigens was estimated before CHMI by protein microarray. RESULTS Frequencies of pro- and anti-inflammatory CD4+ T cells stimulated with the vaccine antigen GMZ2 as well as B cell profiles did not change after vaccination. IL-10-producing CD4+ T cells and CD20+ IgG+ B cells were increased post-vaccination regardless of the intervention, thus could not be specifically attributed to any malaria vaccine regimen. In contrast, GMZ2-specific antibody response increased after the vaccination, but was not correlated to protection. Antibody responses to several P. falciparum blood and liver stage antigens (MSP1, MSP4, MSP8, PfEMP1, STARP) as well as the breadth of the malaria-specific antibody response were significantly higher in protected study participants. CONCLUSIONS In lifelong malaria exposed adults, the main marker of protection against CHMI is a broad antibody pattern recognizing multiple stages of the plasmodial life cycle. Despite vaccination with GMZ2 using a novel formulation, expansion of the GMZ2-stimulated T cells or the GMZ2-specific B cell response was limited, and the vaccine response could not be identified as a marker of protection against malaria. Trial registration PACTR; PACTR201503001038304; Registered 17 February 2015; https://pactr.samrc.ac.za/TrialDisplay.aspx?TrialID=1038.
Collapse
Affiliation(s)
- Odilon Nouatin
- Centre de Recherches Médicales de Lambaréné, BP : 242, Lambaréné, Gabon. .,Institute of Tropical Medicine, University Tübingen, Wilhelmstr. 27, 72074, Tübingen, Germany. .,German Centre for Infection Research (DZIF), Partner Site, Tübingen, Germany. .,Département de Biochimie Et de Biologie Cellulaire, Faculté des Sciences et Techniques, Université d'Abomey-Calavi, Cotonou, Bénin.
| | - Javier Ibáñez
- Institute of Tropical Medicine, University Tübingen, Wilhelmstr. 27, 72074, Tübingen, Germany.,German Centre for Infection Research (DZIF), Partner Site, Tübingen, Germany
| | - Rolf Fendel
- Centre de Recherches Médicales de Lambaréné, BP : 242, Lambaréné, Gabon. .,Institute of Tropical Medicine, University Tübingen, Wilhelmstr. 27, 72074, Tübingen, Germany. .,German Centre for Infection Research (DZIF), Partner Site, Tübingen, Germany.
| | - Ulysse A Ngoa
- Centre de Recherches Médicales de Lambaréné, BP : 242, Lambaréné, Gabon
| | - Freia-Raphaella Lorenz
- Institute of Tropical Medicine, University Tübingen, Wilhelmstr. 27, 72074, Tübingen, Germany
| | - Jean-Claude Dejon-Agobé
- Centre de Recherches Médicales de Lambaréné, BP : 242, Lambaréné, Gabon.,Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Jean Ronald Edoa
- Centre de Recherches Médicales de Lambaréné, BP : 242, Lambaréné, Gabon.,Institute of Tropical Medicine, University Tübingen, Wilhelmstr. 27, 72074, Tübingen, Germany
| | - Judith Flügge
- Institute of Tropical Medicine, University Tübingen, Wilhelmstr. 27, 72074, Tübingen, Germany.,German Centre for Infection Research (DZIF), Partner Site, Tübingen, Germany
| | - Sina Brückner
- Institute of Tropical Medicine, University Tübingen, Wilhelmstr. 27, 72074, Tübingen, Germany
| | - Meral Esen
- Centre de Recherches Médicales de Lambaréné, BP : 242, Lambaréné, Gabon.,Institute of Tropical Medicine, University Tübingen, Wilhelmstr. 27, 72074, Tübingen, Germany.,German Centre for Infection Research (DZIF), Partner Site, Tübingen, Germany.,Cluster of Excellence: EXC 2124: Controlling Microbes to Fight Infection, Tübingen, Germany
| | - Michael Theisen
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark.,Centre for Medical Parasitology at Department of International Health, Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.,Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Denmark
| | | | - Kabirou Moutairou
- Département de Biochimie Et de Biologie Cellulaire, Faculté des Sciences et Techniques, Université d'Abomey-Calavi, Cotonou, Bénin
| | - Adrian J F Luty
- Centre d'Etude et de Recherche sur le Paludisme Associé à la Grossesse et a l'Enfance, Calavi, Bénin.,MERIT, Université de Paris, Paris, IRD, France
| | - Bertrand Lell
- Centre de Recherches Médicales de Lambaréné, BP : 242, Lambaréné, Gabon.,Department of Medicine I, Division of Infectious Diseases and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | - Peter G Kremsner
- Centre de Recherches Médicales de Lambaréné, BP : 242, Lambaréné, Gabon.,Institute of Tropical Medicine, University Tübingen, Wilhelmstr. 27, 72074, Tübingen, Germany.,German Centre for Infection Research (DZIF), Partner Site, Tübingen, Germany
| | - Ayola A Adegnika
- Centre de Recherches Médicales de Lambaréné, BP : 242, Lambaréné, Gabon.,Institute of Tropical Medicine, University Tübingen, Wilhelmstr. 27, 72074, Tübingen, Germany.,German Centre for Infection Research (DZIF), Partner Site, Tübingen, Germany.,Department of Parasitology, Leiden University Medical Centre (LUMC), 2333 ZA, Leiden, The Netherlands.,Fondation pour la Recherche Scientifique, 72 BP45, Cotonou, Bénin
| | - Benjamin Mordmüller
- Centre de Recherches Médicales de Lambaréné, BP : 242, Lambaréné, Gabon.,Institute of Tropical Medicine, University Tübingen, Wilhelmstr. 27, 72074, Tübingen, Germany.,German Centre for Infection Research (DZIF), Partner Site, Tübingen, Germany.,Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
12
|
Efficacy, T cell activation and antibody responses in accelerated Plasmodium falciparum sporozoite chemoprophylaxis vaccine regimens. NPJ Vaccines 2022; 7:59. [PMID: 35641527 PMCID: PMC9156686 DOI: 10.1038/s41541-022-00473-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 03/18/2022] [Indexed: 11/08/2022] Open
Abstract
Repeated direct venous inoculation of Plasmodium falciparum sporozoites (PfSPZ) together with antimalarial chemoprophylaxis (PfSPZ-CVac) is the most potent way to induce sterile immunity against P. falciparum infection in malaria-naive volunteers. However, established schedules are complex and long. Here, we tested two accelerated three-dose schedules (28- and 10-day regimen) assessing efficacy by controlled human malaria infection (CHMI) against placebo, comparing vaccine-specific T cell and antibody responses by antigen-reactive T cell enrichment (ARTE) and protein microarray, respectively. Both regimens were similarly efficacious (67 and 63% vaccine efficacy) but different in the induction of vaccine-specific T cells and antibodies. The 10-day regimen resulted in higher numbers of antigen-specific CD4+ effector memory pro-inflammatory T cells and a broader antibody response compared with the 28-day regimen. Usually in nature, P. falciparum liver stage lasts about 6.5 days. The short vaccination-interval of the 10-day regimen prolongs the time of continuous exposure to liver-stage parasites, which may explain the stronger response. Besides dose and number of vaccinations, duration of liver-stage exposure is a factor to optimize PfSPZ-CVac immunogenicity.
Collapse
|
13
|
Tayipto Y, Liu Z, Mueller I, Longley RJ. Serology for Plasmodium vivax surveillance: A novel approach to accelerate towards elimination. Parasitol Int 2021; 87:102492. [PMID: 34728377 DOI: 10.1016/j.parint.2021.102492] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 08/30/2021] [Accepted: 10/28/2021] [Indexed: 01/13/2023]
Abstract
Plasmodium vivax is the most widespread causative agent of human malaria in the world. Despite the ongoing implementation of malaria control programs, the rate of case reduction has declined over the last 5 years. Hence, surveillance of malaria transmission should be in place to identify and monitor areas that require intensified malaria control interventions. Serological tools may offer additional insights into transmission intensity over parasite and entomological measures, especially as transmission levels decline. Antibodies can be detected in the host system for months to even years after parasite infections have been cleared from the blood, enabling malaria exposure history to be captured. Because the Plasmodium parasite expresses more than 5000 proteins, it is important to a) understand antibody longevity following infection and b) measure antibodies to more than one antigen in order to accurately inform on the exposure and/or immune status of populations. This review summarises current practices for surveillance of P. vivax malaria, the current state of research into serological exposure markers and their potential role for accelerating malaria elimination, and discusses further studies that need to be undertaken to see such technology implemented in malaria-endemic areas.
Collapse
Affiliation(s)
- Yanie Tayipto
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia; Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Zoe Liu
- The Institute for Mental and Physical Health and Clinical Translation, Barwon Health, Deakin University, Geelong, Victoria, Australia; School of Medicine, Centre for Molecular and Medical Research, Deakin University, Geelong, Australia
| | - Ivo Mueller
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia; Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Rhea J Longley
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia; Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
14
|
Antibody signatures of asymptomatic Plasmodium falciparum malaria infections measured from dried blood spots. Malar J 2021; 20:378. [PMID: 34556121 PMCID: PMC8461960 DOI: 10.1186/s12936-021-03915-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 09/13/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Screening malaria-specific antibody responses on protein microarrays can help identify immune factors that mediate protection against malaria infection, disease, and transmission, as well as markers of past exposure to both malaria parasites and mosquito vectors. Most malaria protein microarray work has used serum as the sample matrix, requiring prompt laboratory processing and a continuous cold chain, thus limiting applications in remote locations. Dried blood spots (DBS) pose minimal biohazard, do not require immediate laboratory processing, and are stable at room temperature for transport, making them potentially superior alternatives to serum. The goals of this study were to assess the viability of DBS as a source for antibody profiling and to use DBS to identify serological signatures of low-density Plasmodium falciparum infections in malaria-endemic regions of Myanmar. METHODS Matched DBS and serum samples from a cross-sectional study in Ingapu Township, Myanmar were probed on protein microarrays populated with P. falciparum antigen fragments. Signal and trends in both sample matrices were compared. A case-control study was then performed using banked DBS samples from malaria-endemic regions of Myanmar, and a regularized logistic regression model was used to identify antibody signatures of ultrasensitive PCR-positive P. falciparum infections. RESULTS Approximately 30% of serum IgG activity was recovered from DBS. Despite this loss of antibody activity, antigen and population trends were well-matched between the two sample matrices. Responses to 18 protein fragments were associated with the odds of asymptomatic P. falciparum infection, albeit with modest diagnostic characteristics (sensitivity 58%, specificity 85%, negative predictive value 88%, and positive predictive value 52%). CONCLUSIONS Malaria-specific antibody responses can be reliably detected, quantified, and analysed from DBS, opening the door to serological studies in populations where serum collection, transport, and storage would otherwise be impossible. While test characteristics of antibody signatures were insufficient for individual diagnosis, serological testing may be useful for identifying exposure to asymptomatic, low-density malaria infections, particularly if sero-surveillance strategies target individuals with low previous exposure as sentinels for population exposure.
Collapse
|
15
|
Antigenic sites in SARS-CoV-2 spike RBD show molecular similarity with pathogenic antigenic determinants and harbors peptides for vaccine development. Immunobiology 2021; 226:152091. [PMID: 34303920 PMCID: PMC8297981 DOI: 10.1016/j.imbio.2021.152091] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 03/13/2021] [Accepted: 03/30/2021] [Indexed: 01/22/2023]
Abstract
The spike protein of coronavirus is key target for drug development and other pharmacological interventions. In current study, we performed an integrative approach to predict antigenic sites in SARS-CoV-2 spike receptor binding domain and found nine potential antigenic sites. The predicted antigenic sites were then assessed for possible molecular similarity with other known antigens in different organisms. Out of nine sites, seven sites showed molecular similarity with 54 antigenic determinants found in twelve pathogenic bacterial species (Mycobacterium tuberculosis, Mycobacterium leprae, Bacillus anthracis, Borrelia burgdorferi, Clostridium perfringens, Clostridium tetani, Helicobacter Pylori, Listeria monocytogenes, Staphylococcus aureus, Streptococcus pyogenes, Vibrio cholera and Yersinia pestis), two malarial parasites (Plasmodium falciparum and Plasmodium knowlesi) and influenza virus A. Most of the bacterial antigens that displayed molecular similarity with antigenic sites in SARS-CoV-2 RBD (receptor binding domain) were toxins and virulent factors. Antigens from Mycobacterium that showed similarity were mainly involved in modulating host cell immune response and ensuring persistence and survival of pathogen in host cells. Presence of a large number of antigenic determinants, similar to those in highly pathogenic microorganisms, not merely accounts for complex etiology of the disease but also provides an explanation for observed pathophysiological complications, such as deregulated immune response, unleashed or dysregulated cytokine secretion (cytokine storm), multiple organ failure etc., that are more evident in aged and immune-compromised patients. Over-representation of antigenic determinants from Plasmodium and Mycobacterium in all antigenic sites suggests that anti-malarial and anti-TB drugs can prove to be clinical beneficial for COVID-19 treatment. Besides this, anti-leprosy, anti-lyme, anti-plague, anti-anthrax drugs/vaccine etc. are also expected to be beneficial in COVID-19 treatment. Moreover, individuals previously immunized/vaccinated or had previous history of malaria, tuberculosis or other disease caused by fifteen microorganisms are expected to display a considerable degree of resistance against SARS-CoV-2 infection. Out of the seven antigenic sites predicted in SARS-CoV-2, a part of two antigenic sites were also predicted as potent T-cell epitopes (KVGGNYNYL444-452 and SVLYNSASF366-374) against MHC class I and three (KRISNCVADYSVLYN356-370, DLCFTNVYADSFVI389-402, and YRVVVLSFELLHA508-520) against MHC class II. All epitopes possessed significantly lower predicted IC50 value which is a prerequisite for a preferred vaccine candidate for COVID-19.
Collapse
|
16
|
Ventimiglia NT, Stucke EM, Coulibaly D, Berry AA, Lyke KE, Laurens MB, Bailey JA, Adams M, Niangaly A, Kone AK, Takala-Harrison S, Kouriba B, Doumbo OK, Felgner PL, Plowe CV, Thera MA, Travassos MA. Malian adults maintain serologic responses to virulent PfEMP1s amid seasonal patterns of fluctuation. Sci Rep 2021; 11:14401. [PMID: 34257318 PMCID: PMC8277812 DOI: 10.1038/s41598-021-92974-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 06/08/2021] [Indexed: 12/03/2022] Open
Abstract
Plasmodium falciparum erythrocyte membrane protein-1s (PfEMP1s), diverse malaria proteins expressed on the infected erythrocyte surface, play an important role in pathogenesis, mediating adhesion to host vascular endothelium. Antibodies to particular non-CD36-binding PfEMP1s are associated with protection against severe disease. We hypothesized that given lifelong P. falciparum exposure, Malian adults would have broad PfEMP1 serorecognition and high seroreactivity levels during follow-up, particularly to non-CD36-binding PfEMP1s such as those that attach to endothelial protein C receptor (EPCR) and intercellular adhesion molecule-1 (ICAM-1). Using a protein microarray, we determined serologic responses to 166 reference PfEMP1 fragments during a dry and subsequent malaria transmission season in Malian adults. Malian adult sera had PfEMP1 serologic responses throughout the year, with decreased reactivity to a small subset of PfEMP1 fragments during the dry season and increases in reactivity to a different subset of PfEMP1 fragments during the subsequent peak malaria transmission season, especially for intracellular PfEMP1 domains. For some individuals, PfEMP1 serologic responses increased after the dry season, suggesting antigenic switching during asymptomatic infection. Adults were more likely to experience variable serorecognition of CD36-binding PfEMP1s than non-CD36-binding PfEMP1s that bind EPCR or ICAM-1, which remained serorecognized throughout the year. Sustained seroreactivity to non-CD36-binding PfEMP1s throughout adulthood amid seasonal fluctuation patterns may reflect underlying protective severe malaria immunity and merits further investigation.
Collapse
Affiliation(s)
| | - Emily M Stucke
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Drissa Coulibaly
- University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Andrea A Berry
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kirsten E Lyke
- University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Jason A Bailey
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Matthew Adams
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Amadou Niangaly
- University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Abdoulaye K Kone
- University of Sciences, Techniques and Technologies, Bamako, Mali
| | | | - Bourema Kouriba
- University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Ogobara K Doumbo
- University of Sciences, Techniques and Technologies, Bamako, Mali
| | | | | | | | | |
Collapse
|
17
|
Systems analysis and controlled malaria infection in Europeans and Africans elucidate naturally acquired immunity. Nat Immunol 2021; 22:654-665. [PMID: 33888898 DOI: 10.1038/s41590-021-00911-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 03/02/2021] [Indexed: 01/31/2023]
Abstract
Controlled human infections provide opportunities to study the interaction between the immune system and malaria parasites, which is essential for vaccine development. Here, we compared immune signatures of malaria-naive Europeans and of Africans with lifelong malaria exposure using mass cytometry, RNA sequencing and data integration, before and 5 and 11 days after venous inoculation with Plasmodium falciparum sporozoites. We observed differences in immune cell populations, antigen-specific responses and gene expression profiles between Europeans and Africans and among Africans with differing degrees of immunity. Before inoculation, an activated/differentiated state of both innate and adaptive cells, including elevated CD161+CD4+ T cells and interferon-γ production, predicted Africans capable of controlling parasitemia. After inoculation, the rapidity of the transcriptional response and clusters of CD4+ T cells, plasmacytoid dendritic cells and innate T cells were among the features distinguishing Africans capable of controlling parasitemia from susceptible individuals. These findings can guide the development of a vaccine effective in malaria-endemic regions.
Collapse
|
18
|
Wichers JS, Tonkin-Hill G, Thye T, Krumkamp R, Kreuels B, Strauss J, von Thien H, Scholz JAM, Smedegaard Hansson H, Weisel Jensen R, Turner L, Lorenz FR, Schöllhorn A, Bruchhaus I, Tannich E, Fendel R, Otto TD, Lavstsen T, Gilberger TW, Duffy MF, Bachmann A. Common virulence gene expression in adult first-time infected malaria patients and severe cases. eLife 2021; 10:e69040. [PMID: 33908865 PMCID: PMC8102065 DOI: 10.7554/elife.69040] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 04/18/2021] [Indexed: 12/22/2022] Open
Abstract
Sequestration of Plasmodium falciparum(P. falciparum)-infected erythrocytes to host endothelium through the parasite-derived P. falciparum erythrocyte membrane protein 1 (PfEMP1) adhesion proteins is central to the development of malaria pathogenesis. PfEMP1 proteins have diversified and expanded to encompass many sequence variants, conferring each parasite a similar array of human endothelial receptor-binding phenotypes. Here, we analyzed RNA-seq profiles of parasites isolated from 32 P. falciparum-infected adult travellers returning to Germany. Patients were categorized into either malaria naive (n = 15) or pre-exposed (n = 17), and into severe (n = 8) or non-severe (n = 24) cases. For differential expression analysis, PfEMP1-encoding var gene transcripts were de novo assembled from RNA-seq data and, in parallel, var-expressed sequence tags were analyzed and used to predict the encoded domain composition of the transcripts. Both approaches showed in concordance that severe malaria was associated with PfEMP1 containing the endothelial protein C receptor (EPCR)-binding CIDRα1 domain, whereas CD36-binding PfEMP1 was linked to non-severe malaria outcomes. First-time infected adults were more likely to develop severe symptoms and tended to be infected for a longer period. Thus, parasites with more pathogenic PfEMP1 variants are more common in patients with a naive immune status, and/or adverse inflammatory host responses to first infections favor the growth of EPCR-binding parasites.
Collapse
Affiliation(s)
- J Stephan Wichers
- Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical MedicineHamburgGermany
- Centre for Structural Systems BiologyHamburgGermany
- Biology Department, University of HamburgHamburgGermany
| | | | - Thorsten Thye
- Epidemiology and Diagnostics, Bernhard Nocht Institute for Tropical MedicineHamburgGermany
| | - Ralf Krumkamp
- Epidemiology and Diagnostics, Bernhard Nocht Institute for Tropical MedicineHamburgGermany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-RiemsHamburgGermany
| | - Benno Kreuels
- Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine, GermanyHamburgGermany
- Department of Medicine, College of MedicineBlantyreMalawi
- Department of Medicine, University Medical Center Hamburg-EppendorfHamburgGermany
| | - Jan Strauss
- Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical MedicineHamburgGermany
- Centre for Structural Systems BiologyHamburgGermany
- Biology Department, University of HamburgHamburgGermany
| | - Heidrun von Thien
- Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical MedicineHamburgGermany
- Centre for Structural Systems BiologyHamburgGermany
- Biology Department, University of HamburgHamburgGermany
| | - Judith AM Scholz
- Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical MedicineHamburgGermany
| | | | | | | | | | - Anna Schöllhorn
- Institute of Tropical Medicine, University of TübingenTübingenGermany
| | - Iris Bruchhaus
- Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical MedicineHamburgGermany
- Biology Department, University of HamburgHamburgGermany
| | - Egbert Tannich
- Epidemiology and Diagnostics, Bernhard Nocht Institute for Tropical MedicineHamburgGermany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-RiemsHamburgGermany
| | - Rolf Fendel
- Institute of Tropical Medicine, University of TübingenTübingenGermany
- German Center for Infection Research (DZIF), Partner Site TübingenTübingenGermany
| | - Thomas D Otto
- Institute of Infection, Immunity and Inflammation, University of GlasgowGlasgowUnited Kingdom
| | | | - Tim W Gilberger
- Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical MedicineHamburgGermany
- Centre for Structural Systems BiologyHamburgGermany
- Biology Department, University of HamburgHamburgGermany
| | - Michael F Duffy
- Department of Microbiology and Immunology, University of MelbourneMelbourneAustralia
| | - Anna Bachmann
- Molecular Biology and Immunology, Bernhard Nocht Institute for Tropical MedicineHamburgGermany
- Centre for Structural Systems BiologyHamburgGermany
- Biology Department, University of HamburgHamburgGermany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-RiemsHamburgGermany
| |
Collapse
|
19
|
Mwai K, Kibinge N, Tuju J, Kamuyu G, Kimathi R, Mburu J, Chepsat E, Nyamako L, Chege T, Nkumama I, Kinyanjui S, Musenge E, Osier F. protGear: A protein microarray data pre-processing suite. Comput Struct Biotechnol J 2021; 19:2518-2525. [PMID: 34025940 PMCID: PMC8114118 DOI: 10.1016/j.csbj.2021.04.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 11/29/2022] Open
Abstract
Protein microarrays are versatile tools for high throughput study of the human proteome, but systematic and non-systematic sources of bias constrain optimal interpretation and the ultimate utility of the data. Published guidelines to limit technical variability whilst maintaining important biological variation favour DNA-based microarrays that often differ fundamentally in their experimental design. Rigorous tools to guide background correction, the quantification of within-sample variation, normalisation, and batch correction specifically for protein microarrays are limited, require extensive investigation and are not centrally accessible. Here, we develop a generic one-stop-shop pre-processing suite for protein microarrays that is compatible with data from the major protein microarray scanners. Our graphical and tabular interfaces facilitate a detailed inspection of data and are coupled with supporting guidelines that enable users to select the most appropriate algorithms to systematically address bias arising in customized experiments. The localization and distribution of background signal intensities determine the optimal correction strategy. A novel function overcomes the limitations in the interpretation of the coefficient of variation when signal intensities are at the lower end of the detection threshold. We demonstrate essential considerations in the experimental design and their impact on a range of algorithms for normalization and minimization of batch effects. Our user-friendly interactive web-based platform eliminates the need for prowess in programming. The open-source R interface includes illustrative examples, generates an auditable record, enables reproducibility, and can incorporate additional custom scripts through its online repository. This versatility will enhance its broad uptake in the infectious disease and vaccine development community.
Collapse
Affiliation(s)
- Kennedy Mwai
- Epidemiology and Biostatistics Division, School of Public Health, University of the Witwatersrand, Johannesburg, South Africa.,Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Nelson Kibinge
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - James Tuju
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya.,Department of Biotechnology and Biochemistry, Pwani University, Kilifi, Kenya
| | - Gathoni Kamuyu
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Rinter Kimathi
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - James Mburu
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Emily Chepsat
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Lydia Nyamako
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Timothy Chege
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Irene Nkumama
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya.,Centre of Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Samson Kinyanjui
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya.,Department of Biotechnology and Biochemistry, Pwani University, Kilifi, Kenya.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Eustasius Musenge
- Epidemiology and Biostatistics Division, School of Public Health, University of the Witwatersrand, Johannesburg, South Africa
| | - Faith Osier
- Centre for Geographic Medicine Research (Coast), Kenya Medical Research Institute-Wellcome Trust Research Programme, Kilifi, Kenya.,Centre of Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany.,Department of Biotechnology and Biochemistry, Pwani University, Kilifi, Kenya.,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
20
|
Identification of Plasmodium falciparum-specific protein PIESP2 as a novel virulence factor related to cerebral malaria. Int J Biol Macromol 2021; 177:535-547. [PMID: 33631268 DOI: 10.1016/j.ijbiomac.2021.02.145] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 01/21/2021] [Accepted: 02/19/2021] [Indexed: 01/23/2023]
Abstract
Cerebral malaria (CM) is the most severe complication caused by Plasmodium falciparum infection. The pathophysiological changes caused by parasite virulence factors and the human immune response to parasites contribute to CM. To date, very few parasite virulence proteins have been found to participate in CM. Here, we employed comparative genomics analysis and identified parasite-infected erythrocyte specific protein 2 (PIESP2) to be a CM-related protein. We conducted further experimental investigations and found that PIESP2 is an immunogenic protein. PIESP2 expression begins at the early trophozoite stage and progressively increases with parasite development. Although PIESP2 proteins mainly reside within infected red blood cells (IRBCs), some of them are present on the IRBC surface at the pigmented stage. Moreover, blockage of PIESP2 by antiserum apparently inhibited the adhesion of IRBCs to brain microvascular endothelial cells (BMECs). Western blot analysis detected the binding of PIESP2 to BMECs. Transcriptional analysis revealed that the binding of PIESP2 to BMECs can increase the expression of genes involved in the inflammatory response but decrease the expression of genes related to the anchoring junction. Overall, PIESP2 might be associated with CM by mediating the sequestration of IRBCs, inducing the inflammation response, and impairing the integrity of blood-brain barrier.
Collapse
|
21
|
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: 10] [Impact Index Per Article: 2.5] [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
|
22
|
Amlabu E, Nyarko PB, Opoku G, Ibrahim-Dey D, Ilani P, Mensah-Brown H, Akporh GA, Akuh OA, Ayugane EA, Amoh-Boateng D, Kusi KA, Awandare GA. Localization and function of a Plasmodium falciparum protein (PF3D7_1459400) during erythrocyte invasion. Exp Biol Med (Maywood) 2020; 246:10-19. [PMID: 33019810 DOI: 10.1177/1535370220961764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
IMPACT STATEMENT Plasmodium falciparum malaria is a global health problem. Erythrocyte invasion by P. falciparum merozoites appears to be a promising target to curb malaria. We have identified and characterized a novel protein that is involved in erythrocyte invasion. Our data on protein subcellular localization, stage-specific protein expression pattern, and merozoite invasion inhibition by α-peptide antibodies suggest a role for PF3D7_1459400 protein during P. falciparum erythrocyte invasion. Even more, the human immunoepidemiology data present PF3D7_1459400 protein as an immunogenic antigen which could be further exploited for the development of new anti-infective therapy against malaria.
Collapse
Affiliation(s)
- Emmanuel Amlabu
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra LG 54, Ghana.,Department of Biochemistry, Kogi State University, Anyigba P.M.B 1007, Nigeria
| | - Prince B Nyarko
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra LG 54, Ghana
| | - Grace Opoku
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra LG 54, Ghana
| | - Damata Ibrahim-Dey
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra LG 54, Ghana
| | - Philip Ilani
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra LG 54, Ghana
| | - Henrietta Mensah-Brown
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra LG 54, Ghana
| | - Grace A Akporh
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra LG 54, Ghana
| | - Ojo-Ajogu Akuh
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra LG 54, Ghana
| | - Evelyn A Ayugane
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra LG 54, Ghana
| | - David Amoh-Boateng
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra LG 54, Ghana
| | - Kwadwo A Kusi
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra LG 54, Ghana.,Immunology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra LG581, Ghana
| | - Gordon A Awandare
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra LG 54, Ghana
| |
Collapse
|
23
|
Eisenhut M. The Identification of Native Epitopes Eliciting a Protective High-Affinity Immunoglobulin Subclass Response to Blood Stages of Plasmodium falciparum: Protocol for Observational Studies. JMIR Res Protoc 2020; 9:e15690. [PMID: 32706743 PMCID: PMC7395252 DOI: 10.2196/15690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 03/20/2020] [Accepted: 05/27/2020] [Indexed: 11/13/2022] Open
Abstract
Background Antibodies to blood stages protective against complications of Plasmodium falciparum infection were found to be of immunoglobulin G 1 (IgG1) and IgG3 subclasses and of high affinity to the target epitopes. These target epitopes cannot be characterized using recombinant antigens because of a lack of appropriate glycosylation, phosphorylation, methylation, and bisulfide bond formation, which determine the structure of conformational and nonlinear epitopes within the tertiary and quaternary structures of native P. falciparum antigens. Objective This study aims to develop a method for the comprehensive detection of all P. falciparum schizont antigens, eliciting a protective immune response. Methods Purified parasitophorous vacuole membrane–enclosed merozoite structures (PEMSs) containing native schizont antigens are initially generated, separated by two-dimensional (2D) gel electrophoresis and blotted onto nitrocellulose. Antigens eliciting a protective antibody response are visualized by incubation with sera from patients with clinical immunity. This is followed by the elution of low-affinity antibodies with urea and detection of protective antibody responses by incubation with anti-IgG1 and anti-IgG3 antibodies, which were conjugated to horseradish peroxidase. This is followed by visualization with a color reaction. Blot signals are normalized by relating to the intensity of blot staining with a reference antibody and housekeeping antigens. Results are corrected for intensity of exposure by the relation of antibody responses to global P. falciparum antibody titers. Antigens eliciting the protective responses are identified as immunorelevant from the comparison of spot positions, indicating high-affinity IgG1 or IgG3 responses on the western blot, which is unique to or consistently more intensive in clinically immune individuals compared with nonimmune individuals. The results obtained are validated by using affinity chromatography. Results Another group previously applied 2D western blotting to analyze antibody responses to P. falciparum. The sera of patients allowed the detection of 42 antigenic spots on the 2D immunoblot. The spots detected were excised and subjected to mass spectrometry for identification. A total of 19 protein spots were successfully identified and corresponded to 13 distinct proteins. Another group used immunoaffinity chromatography to identify antigens bound by IgGs produced by mice with enhanced immunity to Plasmodium yoelii. Immunorelevant antigens were isolated and identified by immobilizing immunoglobulin from immune mice to a Sephadex column and then passing a blood-stage antigen mixture through the column followed by the elution of specific bound antigens with sodium deoxycholate and the identification of those antigens by western blotting with specific antibodies. Conclusions 2D western blotting using native antigens has the potential to identify antibody responses selective for specific defined isomeric forms of the same protein, including isoforms (protein species) generated by posttranscriptional modifications such as phosphorylation, glycosylation, and methylation. The process involved in 2D western blotting enables highly sensitive detection, high resolution, and preservation of antibody responses during blotting. Validation by immunoaffinity chromatography can compensate for the antigen loss associated with the blotting process. It has the potential for indirect quantification of protective antibody responses by enabling quantification of the amount of eluted antibody bound antigens through mass spectrometry. International Registered Report Identifier (IRRID) PRR1-10.2196/15690
Collapse
Affiliation(s)
- Michael Eisenhut
- Luton&Dunstable University Hospital NHS Foundation Trust, Luton, United Kingdom
| |
Collapse
|
24
|
Camponovo F, Campo JJ, Le TQ, Oberai A, Hung C, Pablo JV, Teng AA, Liang X, Sim BKL, Jongo S, Abdulla S, Tanner M, Hoffman SL, Daubenberger C, Penny MA. Proteome-wide analysis of a malaria vaccine study reveals personalized humoral immune profiles in Tanzanian adults. eLife 2020; 9:53080. [PMID: 32662772 PMCID: PMC7386907 DOI: 10.7554/elife.53080] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 07/10/2020] [Indexed: 12/31/2022] Open
Abstract
Tanzanian adult male volunteers were immunized by direct venous inoculation with radiation-attenuated, aseptic, purified, cryopreserved Plasmodium falciparum (Pf) sporozoites (PfSPZ Vaccine) and protective efficacy assessed by homologous controlled human malaria infection (CHMI). Serum immunoglobulin G (IgG) responses were analyzed longitudinally using a Pf protein microarray covering 91% of the proteome, providing first insights into naturally acquired and PfSPZ Vaccine-induced whole parasite antibody profiles in malaria pre-exposed Africans. Immunoreactivity was identified against 2239 functionally diverse Pf proteins, showing a wide breadth of humoral response. Antibody-based immune ‘fingerprints’ in these individuals indicated a strong person-specific immune response at baseline, with little changes in the overall humoral immunoreactivity pattern measured after immunization. The moderate increase in immunogenicity following immunization and the extensive and variable breadth of humoral immune response observed in the volunteers at baseline suggest that pre-exposure reduces vaccine-induced antigen reactivity in unanticipated ways.
Collapse
Affiliation(s)
- Flavia Camponovo
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | | | | | - Amit Oberai
- Antigen Discovery Inc, Irvine, United States
| | | | | | - Andy A Teng
- Antigen Discovery Inc, Irvine, United States
| | | | | | - Said Jongo
- Ifakara Health Institute, Dar es Salaam, United Republic of Tanzania
| | - Salim Abdulla
- Ifakara Health Institute, Dar es Salaam, United Republic of Tanzania
| | - Marcel Tanner
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | | | - Claudia Daubenberger
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - Melissa A Penny
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| |
Collapse
|
25
|
Hickey B, Teneza-Mora N, Lumsden J, Reyes S, Sedegah M, Garver L, Hollingdale MR, Banania JG, Ganeshan H, Dowler M, Reyes A, Tamminga C, Singer A, Simmons A, Belmonte M, Belmonte A, Huang J, Inoue S, Velasco R, Abot S, Vasquez CS, Guzman I, Wong M, Twomey P, Wojnarski M, Moon J, Alcorta Y, Maiolatesi S, Spring M, Davidson S, Chaudhury S, Villasante E, Richie TL, Epstein JE. IMRAS-A clinical trial of mosquito-bite immunization with live, radiation-attenuated P. falciparum sporozoites: Impact of immunization parameters on protective efficacy and generation of a repository of immunologic reagents. PLoS One 2020; 15:e0233840. [PMID: 32555601 PMCID: PMC7299375 DOI: 10.1371/journal.pone.0233840] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 05/12/2020] [Indexed: 12/31/2022] Open
Abstract
Background Immunization with radiation-attenuated sporozoites (RAS) by mosquito bite provides >90% sterile protection against Plasmodium falciparum (Pf) malaria in humans. RAS invade hepatocytes but do not replicate. CD8+ T cells recognizing parasite-derived peptides on the surface of infected hepatocytes are likely the primary protective mechanism. We conducted a randomized clinical trial of RAS immunization to assess safety, to achieve 50% vaccine efficacy (VE) against controlled human malaria infection (CHMI), and to generate reagents from protected and non-protected subjects for future identification of protective immune mechanisms and antigens. Methods Two cohorts (Cohort 1 and Cohort 2) of healthy, malaria-naïve, non-pregnant adults age 18–50 received five monthly immunizations with infected (true-immunized, n = 21) or non-infected (mock-immunized, n = 5) mosquito bites and underwent homologous CHMI at 3 weeks. Immunization parameters were selected for 50% protection based on prior clinical data. Leukapheresis was done to collect plasma and peripheral blood mononuclear cells. Results Adverse event rates were similar in true- and mock-immunized subjects. Two true- and two mock-immunized subjects developed large local reactions likely caused by mosquito salivary gland antigens. In Cohort 1, 11 subjects received 810–1235 infected bites; 6/11 (55%) were protected against CHMI vs. 0/3 mock-immunized and 0/6 infectivity controls (VE 55%). In Cohort 2, 10 subjects received 839–1131 infected bites with a higher first dose and a reduced fifth dose; 9/10 (90%) were protected vs. 0/2 mock-immunized and 0/6 controls (VE 90%). Three/3 (100%) protected subjects administered three booster immunizations were protected against repeat CHMI vs. 0/6 controls (VE 100%). Cohort 2 uniquely showed a significant rise in IFN-γ responses after the third and fifth immunizations and higher antibody responses to CSP. Conclusions PfRAS were generally safe and well tolerated. Cohort 2 had a higher first dose, reduced final dose, higher antibody responses to CSP and significant rise of IFN-γ responses after the third and fifth immunizations. Whether any of these factors contributed to increased protection in Cohort 2 requires further investigation. A cryobank of sera and cells from protected and non-protected individuals was generated for future immunological studies and antigen discovery. Trial registration ClinicalTrials.gov NCT01994525.
Collapse
Affiliation(s)
- Bradley Hickey
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Nimfa Teneza-Mora
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Joanne Lumsden
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation, Bethesda, MD, United States of America
| | - Sharina Reyes
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation, Bethesda, MD, United States of America
| | - Martha Sedegah
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Lindsey Garver
- Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
| | - Michael R. Hollingdale
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation, Bethesda, MD, United States of America
- * E-mail:
| | - Jo Glenna Banania
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation, Bethesda, MD, United States of America
| | - Harini Ganeshan
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation, Bethesda, MD, United States of America
| | - Megan Dowler
- Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
| | - Anatalio Reyes
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation, Bethesda, MD, United States of America
| | - Cindy Tamminga
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Alexandra Singer
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Alicia Simmons
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation, Bethesda, MD, United States of America
| | - Maria Belmonte
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation, Bethesda, MD, United States of America
| | - Arnel Belmonte
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation, Bethesda, MD, United States of America
| | - Jun Huang
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation, Bethesda, MD, United States of America
| | - Sandra Inoue
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation, Bethesda, MD, United States of America
| | - Rachel Velasco
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation, Bethesda, MD, United States of America
| | - Steve Abot
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation, Bethesda, MD, United States of America
| | - Carlos S. Vasquez
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation, Bethesda, MD, United States of America
| | - Ivelese Guzman
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation, Bethesda, MD, United States of America
| | - Mimi Wong
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation, Bethesda, MD, United States of America
| | - Patrick Twomey
- Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
| | - Mariusz Wojnarski
- Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
| | - James Moon
- Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
| | - Yolanda Alcorta
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation, Bethesda, MD, United States of America
| | - Santina Maiolatesi
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, United States of America
- Henry M. Jackson Foundation, Bethesda, MD, United States of America
| | - Michele Spring
- Henry M. Jackson Foundation, Bethesda, MD, United States of America
- Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
| | - Silas Davidson
- Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
| | - Sidhartha Chaudhury
- Walter Reed Army Institute of Research, Silver Spring, MD, United States of America
- Biotechnology HPC Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Frederick, MD, United States of America
| | - Eileen Villasante
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Thomas L. Richie
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, United States of America
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Judith E. Epstein
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, United States of America
| |
Collapse
|
26
|
Imported Malaria in Countries where Malaria Is Not Endemic: a Comparison of Semi-immune and Nonimmune Travelers. Clin Microbiol Rev 2020; 33:33/2/e00104-19. [PMID: 32161068 DOI: 10.1128/cmr.00104-19] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The continuous increase in long-distance travel and recent large migratory movements have changed the epidemiological characteristics of imported malaria in countries where malaria is not endemic (here termed non-malaria-endemic countries). While malaria was primarily imported to nonendemic countries by returning travelers, the proportion of immigrants from malaria-endemic regions and travelers visiting friends and relatives (VFRs) in malaria-endemic countries has continued to increase. VFRs and immigrants from malaria-endemic countries now make up the majority of malaria patients in many nonendemic countries. Importantly, this group is characterized by various degrees of semi-immunity to malaria, resulting from repeated exposure to infection and a gradual decline of protection as a result of prolonged residence in non-malaria-endemic regions. Most studies indicate an effect of naturally acquired immunity in VFRs, leading to differences in the parasitological features, clinical manifestation, and odds for severe malaria and clinical complications between immune VFRs and nonimmune returning travelers. There are no valid data indicating evidence for differing algorithms for chemoprophylaxis or antimalarial treatment in semi-immune versus nonimmune malaria patients. So far, no robust biomarkers exist that properly reflect anti-parasite or clinical immunity. Until they are found, researchers should rigorously stratify their study results using surrogate markers, such as duration of time spent outside a malaria-endemic country.
Collapse
|
27
|
Proietti C, Krause L, Trieu A, Dodoo D, Gyan B, Koram KA, Rogers WO, Richie TL, Crompton PD, Felgner PL, Doolan DL. Immune Signature Against Plasmodium falciparum Antigens Predicts Clinical Immunity in Distinct Malaria Endemic Communities. Mol Cell Proteomics 2020; 19:101-113. [PMID: 31658979 PMCID: PMC6944240 DOI: 10.1074/mcp.ra118.001256] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 10/23/2019] [Indexed: 01/14/2023] Open
Abstract
A large body of evidence supports the role of antibodies directed against the Plasmodium spp. parasite in the development of naturally acquired immunity to malaria, however an antigen signature capable of predicting protective immunity against Plasmodium remains to be identified. Key challenges for the identification of a predictive immune signature include the high dimensionality of data produced by high-throughput technologies and the limitation of standard statistical tests in accounting for synergetic interactions between immune responses to multiple targets. In this study, using samples collected from young children in Ghana at multiple time points during a longitudinal study, we adapted a predictive modeling framework which combines feature selection and machine learning techniques to identify an antigen signature of clinical immunity to malaria. Our results show that an individual's immune status can be accurately predicted by measuring antibody responses to a small defined set of 15 target antigens. We further demonstrate that the identified immune signature is highly versatile and capable of providing precise and accurate estimates of clinical protection from malaria in an independent geographic community. Our findings pave the way for the development of a robust point-of-care test to identify individuals at high risk of disease and which could be applied to monitor the impact of vaccinations and other interventions. This approach could be also translated to biomarker discovery for other infectious diseases.
Collapse
Affiliation(s)
- Carla Proietti
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia; QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Lutz Krause
- The University of Queensland Diamantina Institute, Brisbane, QLD, Australia
| | - Angela Trieu
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Daniel Dodoo
- Noguchi Memorial Institute for Medical Research, University of Ghana, Ghana
| | - Ben Gyan
- Noguchi Memorial Institute for Medical Research, University of Ghana, Ghana
| | - Kwadwo A Koram
- Noguchi Memorial Institute for Medical Research, University of Ghana, Ghana
| | | | | | - Peter D Crompton
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland
| | - Philip L Felgner
- Department of Medicine, Division of Infectious Diseases, University of California Irvine, Irvine, California
| | - Denise L Doolan
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia; QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.
| |
Collapse
|
28
|
Tran TM, Crompton PD. Decoding the complexities of human malaria through systems immunology. Immunol Rev 2019; 293:144-162. [PMID: 31680289 DOI: 10.1111/imr.12817] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 09/27/2019] [Accepted: 09/30/2019] [Indexed: 12/18/2022]
Abstract
The complexity of the Plasmodium parasite and its life cycle poses a challenge to our understanding of the host immune response against malaria. Studying human immune responses during natural and experimental Plasmodium infections can enhance our understanding of malaria-protective immunity and inform the design of disease-modifying adjunctive therapies and next-generation malaria vaccines. Systems immunology can complement conventional approaches to facilitate our understanding of the complex immune response to the highly dynamic malaria parasite. In this review, recent studies that used systems-based approaches to evaluate human immune responses during natural and experimental Plasmodium falciparum and Plasmodium vivax infections as well as during immunization with candidate malaria vaccines are summarized and related to each other. The potential for next-generation technologies to address the current limitations of systems-based studies of human malaria are discussed.
Collapse
Affiliation(s)
- Tuan M Tran
- Division of Infectious Diseases, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.,Ryan White Center for Pediatric Infectious Disease and Global Health, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Peter D Crompton
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
29
|
Whittaker K, Burgess R, Jones V, Yang Y, Zhou W, Luo S, Wilson J, Huang R. Quantitative proteomic analyses in blood: A window to human health and disease. J Leukoc Biol 2019; 106:759-775. [PMID: 31329329 DOI: 10.1002/jlb.mr1118-440r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/11/2019] [Accepted: 06/24/2019] [Indexed: 12/13/2022] Open
Affiliation(s)
| | | | | | | | | | - Shuhong Luo
- RayBiotech Life Norcross Georgia USA
- RayBiotech Life Guangzhou Guangdong China
- South China Biochip Research Center Guangzhou Guangdong China
| | | | - Ruo‐Pan Huang
- RayBiotech Life Norcross Georgia USA
- RayBiotech Life Guangzhou Guangdong China
- South China Biochip Research Center Guangzhou Guangdong China
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityGuangzhou Medical University Guangzhou China
- Guangdong Provincial Hospital of Chinese Medicine Guangzhou China
| |
Collapse
|
30
|
Venkatesh A, Jain A, Davies H, Periera L, Maki JN, Gomes E, Felgner PL, Srivastava S, Patankar S, Rathod PK. Hospital-derived antibody profiles of malaria patients in Southwest India. Malar J 2019; 18:138. [PMID: 30995911 PMCID: PMC6472095 DOI: 10.1186/s12936-019-2771-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 04/08/2019] [Indexed: 11/10/2022] Open
Abstract
Background Naturally acquired immunity to malaria across the globe varies in intensity and protective powers. Many of the studies on immunity are from hyperendemic regions of Africa. In Asia, particularly in India, there are unique opportunities for exploring and understanding malaria immunity relative to host age, co-occurrence of Plasmodium falciparum and Plasmodium vivax infections, varying travel history, and varying disease severity. Variation in immunity in hospital settings is particularly understudied. Methods A US NIH ICEMR (South Asia) team examined the level of immunity in an Indian malaria patient population visiting or admitted to Goa Medical College and Hospital in Goa, India. Sera from 200 patients of different ages, in different seasons, infected with P. falciparum or P. vivax or both species, and with different clinical severity were applied to an established protein array system with over 1000 P. falciparum and P. vivax antigens. Differential binding of patient IgG to different antigens was measured. Results Even though Goa itself has much more P. vivax than P. falciparum, IgG reactivity towards P. falciparum antigens was very strong and comparable to that seen in regions of the world with high P. falciparum endemicity. Of 248 seropositive P. falciparum antigens, the strongest were VAR, MSP10, HSP70, PTP5, AP2, AMA1, and SYN6. In P. vivax patients, ETRAMPs, MSPs, and ApiAP2, sexual stage antigen s16, RON3 were the strongest IgG binders. Both P. falciparum and P. vivax patients also revealed strong binding to new antigens with unknown functions. Seropositives showed antigens unique to the young (HSP40, ACS6, GCVH) or to non-severe malaria (MSP3.8 and PHIST). Conclusion Seroreactivity at a major hospital in Southwest India reveals antibody responses to P. falciparum and P. vivax in a low malaria transmission region with much migration. In addition to markers of transmission, the data points to specific leads for possible protective immunity against severe disease. Several, but not all, key antigens overlap with work from different settings around the globe and from other parts of India. Together, these studies confidently help define antigens with the greatest potential chance of universal application for surveillance and possibly for disease protection, in many different parts of India and the world. Electronic supplementary material The online version of this article (10.1186/s12936-019-2771-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Apoorva Venkatesh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Aarti Jain
- Vaccine R&D Center, University of California, Irvine, CA, 92697, USA
| | - Huw Davies
- Vaccine R&D Center, University of California, Irvine, CA, 92697, USA
| | - Ligia Periera
- Department of Chemistry and Department of Global Health, University of Washington, Seattle, WA, 98195, USA
| | - Jennifer N Maki
- Department of Chemistry and Department of Global Health, University of Washington, Seattle, WA, 98195, USA
| | - Edwin Gomes
- Department of Medicine, Goa Medical College and Hospital, Bambolim, Goa, 403202, India
| | - Philip L Felgner
- Vaccine R&D Center, University of California, Irvine, CA, 92697, USA
| | - Sanjeeva Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Swati Patankar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Pradipsinh K Rathod
- Department of Chemistry and Department of Global Health, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
31
|
Jaenisch T, Heiss K, Fischer N, Geiger C, Bischoff FR, Moldenhauer G, Rychlewski L, Sié A, Coulibaly B, Seeberger PH, Wyrwicz LS, Breitling F, Loeffler FF. High-density Peptide Arrays Help to Identify Linear Immunogenic B-cell Epitopes in Individuals Naturally Exposed to Malaria Infection. Mol Cell Proteomics 2019; 18:642-656. [PMID: 30630936 PMCID: PMC6442360 DOI: 10.1074/mcp.ra118.000992] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 11/28/2018] [Indexed: 01/31/2023] Open
Abstract
High-density peptide arrays are an excellent means to profile anti-plasmodial antibody responses. Different protein intrinsic epitopes can be distinguished, and additional insights are gained, when compared with assays involving the full-length protein. Distinct reactivities to specific epitopes within one protein may explain differences in published results, regarding immunity or susceptibility to malaria. We pursued three approaches to find specific epitopes within important plasmodial proteins, (1) twelve leading vaccine candidates were mapped as overlapping 15-mer peptides, (2) a bioinformatical approach served to predict immunogenic malaria epitopes which were subsequently validated in the assay, and (3) randomly selected peptides from the malaria proteome were screened as a control. Several peptide array replicas were prepared, employing particle-based laser printing, and were used to screen 27 serum samples from a malaria-endemic area in Burkina Faso, West Africa. The immunological status of the individuals was classified as "protected" or "unprotected" based on clinical symptoms, parasite density, and age. The vaccine candidate screening approach resulted in significant hits in all twelve proteins and allowed us (1) to verify many known immunogenic structures, (2) to map B-cell epitopes across the entire sequence of each antigen and (3) to uncover novel immunogenic epitopes. Predicting immunogenic regions in the proteome of the human malaria parasite Plasmodium falciparum, via the bioinformatics approach and subsequent array screening, confirmed known immunogenic sequences, such as in the leading malaria vaccine candidate CSP and discovered immunogenic epitopes derived from hypothetical or unknown proteins.
Collapse
Affiliation(s)
- Thomas Jaenisch
- From the ‡Center for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Im Neuenheimer Feld 324, D 69120 Heidelberg, Germany;; §German Center for Infectious Disease Research, Heidelberg (DZIF);; ¶HEiKA - Heidelberg Karlsruhe Research Partnership, Heidelberg University, Karlsruhe Institute of Technology (KIT), Germany;.
| | - Kirsten Heiss
- From the ‡Center for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Im Neuenheimer Feld 324, D 69120 Heidelberg, Germany;; §German Center for Infectious Disease Research, Heidelberg (DZIF)
| | - Nico Fischer
- From the ‡Center for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Im Neuenheimer Feld 324, D 69120 Heidelberg, Germany;; §German Center for Infectious Disease Research, Heidelberg (DZIF);; ¶HEiKA - Heidelberg Karlsruhe Research Partnership, Heidelberg University, Karlsruhe Institute of Technology (KIT), Germany
| | - Carolin Geiger
- From the ‡Center for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Im Neuenheimer Feld 324, D 69120 Heidelberg, Germany;; §German Center for Infectious Disease Research, Heidelberg (DZIF)
| | - F Ralf Bischoff
- ‖German Cancer Research Center, Im Neuenheimer Feld 280, D 69120 Heidelberg, Germany
| | - Gerhard Moldenhauer
- ‖German Cancer Research Center, Im Neuenheimer Feld 280, D 69120 Heidelberg, Germany
| | - Leszek Rychlewski
- BioInfoBank Institute, Św. Marcin 80/82 lok. 355, 61-809 Poznań, Poland
| | - Ali Sié
- Centre de Recherche en Santé de Nouna, BP 02 Nouna, Rue Namory Keita, Burkina Faso
| | - Boubacar Coulibaly
- Centre de Recherche en Santé de Nouna, BP 02 Nouna, Rue Namory Keita, Burkina Faso
| | - Peter H Seeberger
- §§Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, D 14476 Potsdam, Germany
| | - Lucjan S Wyrwicz
- Department of Oncology and Radiotherapy, M Sklodowska Curie Memorial Cancer Center, Wawelska 15, 02-034 Warsaw, Poland
| | - Frank Breitling
- ‖‖Institute of Microstructure Technology, Karlsruhe Institute of Technology, Germany Hermann-von-Helmholtz-Platz 1, D 76344 Eggenstein-Leopoldshafen, Germany
| | - Felix F Loeffler
- ¶HEiKA - Heidelberg Karlsruhe Research Partnership, Heidelberg University, Karlsruhe Institute of Technology (KIT), Germany;; §§Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, D 14476 Potsdam, Germany;.
| |
Collapse
|
32
|
Kobayashi T, Jain A, Liang L, Obiero JM, Hamapumbu H, Stevenson JC, Thuma PE, Lupiya J, Chaponda M, Mulenga M, Mamini E, Mharakurwa S, Gwanzura L, Munyati S, Mutambu S, Felgner P, Davies DH, Moss WJ. Distinct Antibody Signatures Associated with Different Malaria Transmission Intensities in Zambia and Zimbabwe. mSphere 2019; 4:e00061-19. [PMID: 30918058 PMCID: PMC6437277 DOI: 10.1128/mspheredirect.00061-19] [Citation(s) in RCA: 15] [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: 02/06/2019] [Accepted: 02/08/2019] [Indexed: 12/30/2022] Open
Abstract
Antibodies to Plasmodium falciparum are specific biomarkers that can be used to monitor parasite exposure over broader time frames than microscopy, rapid diagnostic tests, or molecular assays. Consequently, seroprevalence surveys can assist with monitoring the impact of malaria control interventions, particularly in the final stages of elimination, when parasite incidence is low. The protein array format to measure antibodies to diverse P. falciparum antigens requires only small sample volumes and is high throughput, permitting the monitoring of malaria transmission on large spatial and temporal scales. We expanded the use of a protein microarray to assess malaria transmission in settings beyond those with a low malaria incidence. Antibody responses in children and adults were profiled, using a P. falciparum protein microarray, through community-based surveys in three areas in Zambia and Zimbabwe at different stages of malaria control and elimination. These three epidemiological settings had distinct serological profiles reflective of their malaria transmission histories. While there was little correlation between transmission intensity and antibody signals (magnitude or breadth) in adults, there was a clear correlation in children younger than 5 years of age. Antibodies in adults appeared to be durable even in the absence of significant recent transmission, whereas antibodies in children provided a more accurate picture of recent levels of transmission intensity. Seroprevalence studies in children could provide a valuable marker of progress toward malaria elimination.IMPORTANCE As malaria approaches elimination in many areas of the world, monitoring the effect of control measures becomes more important but challenging. Low-level infections may go undetected by conventional tests that depend on parasitemia, particularly in immune individuals, who typically show no symptoms of malaria. In contrast, antibodies persist after parasitemia and may provide a more accurate picture of recent exposure. Only a few parasite antigens-mainly vaccine candidates-have been evaluated in seroepidemiological studies. We examined antibody responses to 500 different malaria proteins in blood samples collected through community-based surveillance from areas with low, medium, and high malaria transmission intensities. The breadth of the antibody responses in adults was broad in all three settings and was a poor correlate of recent exposure. In contrast, children represented a better sentinel population for monitoring recent malaria transmission. These data will help inform the use of multiplex serology for malaria surveillance.
Collapse
Affiliation(s)
- Tamaki Kobayashi
- Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Aarti Jain
- Vaccine Research & Development Center, Department of Physiology & Biophysics, School of Medicine, University of California, Irvine, Irvine, California, USA
| | - Li Liang
- Vaccine Research & Development Center, Department of Physiology & Biophysics, School of Medicine, University of California, Irvine, Irvine, California, USA
| | - Joshua M Obiero
- Vaccine Research & Development Center, Department of Physiology & Biophysics, School of Medicine, University of California, Irvine, Irvine, California, USA
| | | | - Jennifer C Stevenson
- Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Macha Research Trust, Choma, Zambia
| | - Philip E Thuma
- Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Macha Research Trust, Choma, Zambia
| | - James Lupiya
- Tropical Diseases Research Centre, Ndola, Zambia
| | | | | | - Edmore Mamini
- Biomedical Research and Training Institute, Harare, Zimbabwe
| | | | | | - Shungu Munyati
- Biomedical Research and Training Institute, Harare, Zimbabwe
| | - Susan Mutambu
- National Institute of Health Research, Harare, Zimbabwe
| | - Philip Felgner
- Vaccine Research & Development Center, Department of Physiology & Biophysics, School of Medicine, University of California, Irvine, Irvine, California, USA
| | - D Huw Davies
- Vaccine Research & Development Center, Department of Physiology & Biophysics, School of Medicine, University of California, Irvine, Irvine, California, USA
| | - William J Moss
- Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
33
|
Cooper MM, Loiseau C, McCarthy JS, Doolan DL. Human challenge models: tools to accelerate the development of malaria vaccines. Expert Rev Vaccines 2019; 18:241-251. [PMID: 30732492 DOI: 10.1080/14760584.2019.1580577] [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] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Malaria challenge models, where healthy human volunteers are intentionally infected with Plasmodium species parasites under controlled conditions, can be undertaken in several well-defined ways. These challenge models enable evaluation of the kinetics of parasite growth and clearance, host-pathogen interactions and the host immune response. They can facilitate discovery of candidate diagnostic biomarkers and novel vaccine targets. As translational tools they can facilitate testing of candidate vaccines and drugs and evaluation of diagnostic tests. AREAS COVERED Until recently, malaria human challenge models have been limited to only a few Plasmodium falciparum strains and used exclusively in malaria-naïve volunteers in non-endemic regions. Several recent advances include the use of alternate P. falciparum strains and other species of Plasmodia, as well as strains attenuated by chemical, radiation or genetic modification, and the conduct of studies in pre-exposed individuals. Herein, we discuss how this diversification is enabling more thorough vaccine efficacy testing and informing rational vaccine development. EXPERT OPINION The ability to comprehensively evaluate vaccine efficacy in controlled settings will continue to accelerate the translation of candidate malaria vaccines to the clinic, and inform the development and optimisation of potential vaccines that would be effective against multiple strains in geographically and demographically diverse settings.
Collapse
Affiliation(s)
- Martha M Cooper
- a Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine , James Cook University , Cairns , Australia
| | - Claire Loiseau
- a Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine , James Cook University , Cairns , Australia
| | - James S McCarthy
- b Infectious Diseases Programme , QIMR Berghofer Medical Research Institute , Brisbane , Australia
| | - Denise L Doolan
- a Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine , James Cook University , Cairns , Australia
| |
Collapse
|
34
|
Obiero JM, Campo JJ, Scholzen A, Randall A, Bijker EM, Roestenberg M, Hermsen CC, Teng A, Jain A, Davies DH, Sauerwein RW, Felgner PL. Antibody Biomarkers Associated with Sterile Protection Induced by Controlled Human Malaria Infection under Chloroquine Prophylaxis. mSphere 2019; 4:e00027-19. [PMID: 30787114 PMCID: PMC6382972 DOI: 10.1128/mspheredirect.00027-19] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 01/21/2019] [Indexed: 12/25/2022] Open
Abstract
Immunization with sporozoites under chloroquine chemoprophylaxis (CPS) induces distinctly preerythrocytic and long-lasting sterile protection against homologous controlled human malaria infection (CHMI). To identify possible humoral immune correlates of protection, plasma samples were collected from 38 CPS-immunized Dutch volunteers for analysis using a whole Plasmodium falciparum proteome microarray with 7,455 full-length or segmented protein features displaying about 91% of the total P. falciparum proteome. We identified 548 reactive antigens representing 483 unique proteins. Using the breadth of antibody responses for each subject in a mixture-model algorithm, we observed a trimodal pattern, with distinct groups of 16 low responders, 19 medium responders, and 3 high responders. Fifteen out of 16 low responders, 12 of the 19 medium responders, and 3 out of 3 high responders were fully protected from a challenge infection. In the medium-responder group, we identified six novel antigens associated with protection (area under the curve [AUC] value of ≥0.75; P < 0.05) and six other antigens that were specifically increased in nonprotected volunteers (AUC value of ≤0.25; P < 0.05). When used in combination, the multiantigen classifier predicts CPS-induced protective efficacy with 83% sensitivity and 88% specificity. The antibody response patterns characterized in this study represent surrogate markers that may provide rational guidance for clinical vaccine development.IMPORTANCE Infection by Plasmodium parasites has been a major cause of mortality and morbidity in humans for thousands of years. Despite the considerable reduction of deaths, according to the WHO, over 5 billion people are still at risk, with about 216 million worldwide cases occurring in 2016. More compelling, 15 countries in sub-Saharan Africa bore 80% of the worldwide malaria burden. Complete eradication has been challenging, and the development of an affordable and effective vaccine will go a long way in achieving elimination. However, identifying vaccine candidate targets has been difficult. In the present study, we use a highly effective immunization protocol that confers long-lasting sterile immunity in combination with a whole P. falciparum proteome microarray to identify antibody responses associated with protection. This study characterizes a novel antibody profile associated with sterile protective immunity and trimodal humoral responses that sheds light on the possible mechanism of CPS-induced immunity against P. falciparum parasites.
Collapse
Affiliation(s)
- Joshua M Obiero
- Vaccine Research and Development Center, Department of Physiology and Biophysics, University of California, Irvine, Irvine, California, USA
| | | | - Anja Scholzen
- Radboud University Medical Center, Department of Medical Microbiology, Nijmegen, The Netherlands
| | - Arlo Randall
- Antigen Discovery, Inc., Irvine, California, USA
| | - Else M Bijker
- Radboud University Medical Center, Department of Medical Microbiology, Nijmegen, The Netherlands
| | - Meta Roestenberg
- Radboud University Medical Center, Department of Medical Microbiology, Nijmegen, The Netherlands
| | - Cornelus C Hermsen
- Radboud University Medical Center, Department of Medical Microbiology, Nijmegen, The Netherlands
| | - Andy Teng
- Antigen Discovery, Inc., Irvine, California, USA
| | - Aarti Jain
- Vaccine Research and Development Center, Department of Physiology and Biophysics, University of California, Irvine, Irvine, California, USA
| | - D Huw Davies
- Vaccine Research and Development Center, Department of Physiology and Biophysics, University of California, Irvine, Irvine, California, USA
| | - Robert W Sauerwein
- Radboud University Medical Center, Department of Medical Microbiology, Nijmegen, The Netherlands
| | - Philip L Felgner
- Vaccine Research and Development Center, Department of Physiology and Biophysics, University of California, Irvine, Irvine, California, USA
| |
Collapse
|
35
|
Taghavian O, Jain A, Joyner CJ, Ketchum S, Nakajima R, Jasinskas A, Liang L, Fong R, King C, Greenhouse B, Murphy M, Bailey J, Galinski MR, Barnwell JW, Plowe CV, Davies DH, Felgner PL. Antibody Profiling by Proteome Microarray with Multiplex Isotype Detection Reveals Overlap between Human and Aotus nancymaae Controlled Malaria Infections. Proteomics 2019; 18. [PMID: 29266845 DOI: 10.1002/pmic.201700277] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 11/21/2017] [Indexed: 12/13/2022]
Abstract
The development of vaccines against malaria and serodiagnostic tests for detecting recent exposure requires tools for antigen discovery and suitable animal models. The protein microarray is a high-throughput, sample sparing technique, with applications in infectious disease research, clinical diagnostics, epidemiology, and vaccine development. We recently demonstrated Qdot-based indirect immunofluorescence together with portable optical imager ArrayCAM using single isotype detection could replicate data using the conventional laser confocal scanner system. We developed a multiplexing protocol for simultaneous detection of IgG, IgA, and IgM and compared samples from a controlled human malaria infection model with those from controlled malaria infections of Aotus nancymaae, a widely used non-human primate model of human malaria. IgG profiles showed the highest concordance in number of reactive antigens; thus, of the 139 antigens recognized by human IgG antibody, 111 were also recognized by Aotus monkeys. Interestingly, IgA profiles were largely non-overlapping. Finally, on the path toward wider deployment of the portable platform, we show excellent correlations between array data obtained in five independent laboratories around the United States using the multiplexing protocol (R2 : 0.60-0.92). This study supports the use of this platform for wider deployment, particularly in endemic areas where such a tool will have the greatest impact on global human health.
Collapse
Affiliation(s)
- Omid Taghavian
- Department of Medicine, Division of Infectious Diseases, University of California Irvine, Irvine, CA, USA
| | - Aarti Jain
- Department of Medicine, Division of Infectious Diseases, University of California Irvine, Irvine, CA, USA
| | - Chester J Joyner
- International Center for Malaria Research, Education and Development, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | | | - Rie Nakajima
- Department of Medicine, Division of Infectious Diseases, University of California Irvine, Irvine, CA, USA
| | - Algis Jasinskas
- Department of Medicine, Division of Infectious Diseases, University of California Irvine, Irvine, CA, USA
| | - Li Liang
- Department of Medicine, Division of Infectious Diseases, University of California Irvine, Irvine, CA, USA
| | - Rich Fong
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, OH, USA
| | - Christopher King
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, OH, USA
| | - Bryan Greenhouse
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Maxwell Murphy
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Jason Bailey
- Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Mary R Galinski
- International Center for Malaria Research, Education and Development, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.,Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, GA, USA
| | - John W Barnwell
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Christopher V Plowe
- Howard Hughes Medical Institute/Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD, USA
| | - D Huw Davies
- Department of Medicine, Division of Infectious Diseases, University of California Irvine, Irvine, CA, USA
| | - Philip L Felgner
- Department of Medicine, Division of Infectious Diseases, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
36
|
Abstract
The varied landscape of the adaptive immune response is determined by the peptides presented by immune cells, derived from viral or microbial pathogens or cancerous cells. The study of immune biomarkers or antigens is not new, and classical methods such as agglutination, enzyme-linked immunosorbent assay, or Western blotting have been used for many years to study the immune response to vaccination or disease. However, in many of these traditional techniques, protein or peptide identification has often been the bottleneck. Recent progress in genomics and mass spectrometry have led to many of the rapid advances in proteomics approaches. Immunoproteomics describes a rapidly growing collection of approaches that have the common goal of identifying and measuring antigenic peptides or proteins. This includes gel-based, array-based, mass spectrometry-based, DNA-based, or in silico approaches. Immunoproteomics is yielding an understanding of disease and disease progression, vaccine candidates, and biomarkers. This review gives an overview of immunoproteomics and closely related technologies that are used to define the full set of protein antigens targeted by the immune system during disease.
Collapse
Affiliation(s)
- Kelly M Fulton
- Human Health Therapeutics Research Centre, National Research Council of Canada, Ottawa, ON, Canada
| | - Isabel Baltat
- Human Health Therapeutics Research Centre, National Research Council of Canada, Ottawa, ON, Canada
| | - Susan M Twine
- Human Health Therapeutics Research Centre, National Research Council of Canada, Ottawa, ON, Canada.
| |
Collapse
|
37
|
Schreeg ME, Marr HS, Tarigo JL, Sherrill MK, Outi HK, Scholl EH, Bird DM, Vigil A, Hung C, Nakajima R, Liang L, Trieu A, Doolan DL, Thomas JE, Levy MG, Reichard MV, Felgner PL, Cohn LA, Birkenheuer AJ. Identification of Cytauxzoon felis antigens via protein microarray and assessment of expression library immunization against cytauxzoonosis. Clin Proteomics 2018; 15:44. [PMID: 30618510 PMCID: PMC6310948 DOI: 10.1186/s12014-018-9218-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 12/14/2018] [Indexed: 01/17/2023] Open
Abstract
Background Cytauxzoonosis is a disease of felids in North America caused by the tick-transmitted apicomplexan parasite Cytauxzoon felis. Cytauxzoonosis is particularly virulent for domestic cats, but no vaccine currently exists. The parasite cannot be cultivated in vitro, presenting a significant limitation for vaccine development. Methods Recent sequencing of the C. felis genome has identified over 4300 putative protein-encoding genes. From this pool we constructed a protein microarray containing 673 putative C. felis proteins. This microarray was probed with sera from C. felis-infected and naïve cats to identify differentially reactive antigens which were incorporated into two expression library vaccines, one polyvalent and one monovalent. We assessed the efficacy of these vaccines to prevent of infection and/or disease in a tick-challenge model. Results Probing of the protein microarray resulted in identification of 30 differentially reactive C. felis antigens that were incorporated into the two expression library vaccines. However, expression library immunization failed to prevent infection or disease in cats challenged with C. felis. Conclusions Protein microarray facilitated high-throughput identification of novel antigens, substantially increasing the pool of characterized C. felis antigens. These antigens should be considered for development of C. felis vaccines, diagnostics, and therapeutics. Electronic supplementary material The online version of this article (10.1186/s12014-018-9218-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Megan E Schreeg
- 1College of Veterinary Medicine, North Carolina State University, Research Building Room 464, 1060 William Moore Drive, Raleigh, NC 27607 USA
| | - Henry S Marr
- 1College of Veterinary Medicine, North Carolina State University, Research Building Room 464, 1060 William Moore Drive, Raleigh, NC 27607 USA
| | - Jaime L Tarigo
- 1College of Veterinary Medicine, North Carolina State University, Research Building Room 464, 1060 William Moore Drive, Raleigh, NC 27607 USA.,2College of Veterinary Medicine, University of Georgia, 501 D.W. Brooks Drive, Athens, GA 30602 USA
| | - Meredith K Sherrill
- 3College of Veterinary Medicine, University of Missouri, 1600 East Rollins, Columbia, MO 65211 USA
| | - Hilton K Outi
- 3College of Veterinary Medicine, University of Missouri, 1600 East Rollins, Columbia, MO 65211 USA
| | - Elizabeth H Scholl
- 4College of Agriculture and Life Sciences, North Carolina State University, 2501 Founders Dr, Raleigh, NC 27607 USA
| | - David M Bird
- 4College of Agriculture and Life Sciences, North Carolina State University, 2501 Founders Dr, Raleigh, NC 27607 USA
| | - Adam Vigil
- 5School of Medicine, University of California Irvine, 1001 Health Sciences Rd, Irvine, CA 92617 USA
| | - Chris Hung
- 5School of Medicine, University of California Irvine, 1001 Health Sciences Rd, Irvine, CA 92617 USA
| | - Rie Nakajima
- 5School of Medicine, University of California Irvine, 1001 Health Sciences Rd, Irvine, CA 92617 USA
| | - Li Liang
- 5School of Medicine, University of California Irvine, 1001 Health Sciences Rd, Irvine, CA 92617 USA
| | - Angela Trieu
- 6QIMR Berghofer Medical Research Institute, 300 Herston Rd, Brisbane City, QLD 4006 Australia
| | - Denise L Doolan
- 6QIMR Berghofer Medical Research Institute, 300 Herston Rd, Brisbane City, QLD 4006 Australia.,7Australian Institute of Tropical Health and Medicine, James Cook University, 1 James Cook Dr, Douglas, QLD 4814 Australia
| | - Jennifer E Thomas
- 8Center for Veterinary Health Sciences, Oklahoma State University, 208 S McFarland St, Stillwater, OK 74078 USA
| | - Michael G Levy
- 1College of Veterinary Medicine, North Carolina State University, Research Building Room 464, 1060 William Moore Drive, Raleigh, NC 27607 USA
| | - Mason V Reichard
- 8Center for Veterinary Health Sciences, Oklahoma State University, 208 S McFarland St, Stillwater, OK 74078 USA
| | - Philip L Felgner
- 5School of Medicine, University of California Irvine, 1001 Health Sciences Rd, Irvine, CA 92617 USA
| | - Leah A Cohn
- 3College of Veterinary Medicine, University of Missouri, 1600 East Rollins, Columbia, MO 65211 USA
| | - Adam J Birkenheuer
- 1College of Veterinary Medicine, North Carolina State University, Research Building Room 464, 1060 William Moore Drive, Raleigh, NC 27607 USA
| |
Collapse
|
38
|
Kassegne K, Abe EM, Cui YB, Chen SB, Xu B, Deng WP, Shen HM, Wang Y, Chen JH, Zhou XN. Contribution of Plasmodium immunomics: potential impact for serological testing and surveillance of malaria. Expert Rev Proteomics 2018; 16:117-129. [PMID: 30513025 DOI: 10.1080/14789450.2019.1554441] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Introduction: Plasmodium vivax (Pv) and P. knowlesi account together for a considerable share of the global burden of malaria, along with P. falciparum (Pf). However, inaccurate diagnosis and undetectable asymptomatic/submicroscopic malaria infections remain very challenging. Blood-stage antigens involved in either invasion of red blood cells or sequestration/cytoadherence of parasitized erythrocytes have been immunomics-characterized, and are vital for the detection of malaria incidence. Areas covered: We review the recent advances in Plasmodium immunomics to discuss serological markers with potential for specific and sensitive diagnosis of malaria. Insights on alternative use of immunomics to assess malaria prevalence are also highlighted. Finally, we provide practical applications of serological markers as diagnostics, with an emphasis on dot immunogold filtration assay which holds promise for malaria diagnosis and epidemiological surveys. Expert commentary: The approach largely contributes to Pf and Pv research in identifying promising non-orthologous antigens able to detect malaria incidence and to differentiate between past and recent infections. However, further studies to profiling naturally acquired immune responses are expected in order to help discover/validate serological markers of no cross-seroreactivity and guide control interventions. More so, the application of immunomics to knowlesi infections would help validate the recently identified antigens and contribute to the discovery of additional biomarkers of exposure, immunity, or both.
Collapse
Affiliation(s)
- Kokouvi Kassegne
- a National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health , National Centre for International Research on Tropical Diseases, WHO Collaborating Center for Tropical Diseases, Shanghai, People's Republic of China
| | - Eniola Michael Abe
- a National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health , National Centre for International Research on Tropical Diseases, WHO Collaborating Center for Tropical Diseases, Shanghai, People's Republic of China
| | - Yan-Bing Cui
- a National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health , National Centre for International Research on Tropical Diseases, WHO Collaborating Center for Tropical Diseases, Shanghai, People's Republic of China
| | - Shen-Bo Chen
- a National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health , National Centre for International Research on Tropical Diseases, WHO Collaborating Center for Tropical Diseases, Shanghai, People's Republic of China
| | - Bin Xu
- a National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health , National Centre for International Research on Tropical Diseases, WHO Collaborating Center for Tropical Diseases, Shanghai, People's Republic of China
| | - Wang-Ping Deng
- a National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health , National Centre for International Research on Tropical Diseases, WHO Collaborating Center for Tropical Diseases, Shanghai, People's Republic of China
| | - Hai-Mo Shen
- a National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health , National Centre for International Research on Tropical Diseases, WHO Collaborating Center for Tropical Diseases, Shanghai, People's Republic of China
| | - Yue Wang
- b Institute of Parasitic Diseases , Zhejiang Academy of Medical Sciences , Hangzhou , People's Republic of China
| | - Jun-Hu Chen
- a National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health , National Centre for International Research on Tropical Diseases, WHO Collaborating Center for Tropical Diseases, Shanghai, People's Republic of China
| | - Xiao-Nong Zhou
- a National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health , National Centre for International Research on Tropical Diseases, WHO Collaborating Center for Tropical Diseases, Shanghai, People's Republic of China
| |
Collapse
|
39
|
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: 28] [Impact Index Per Article: 4.7] [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
|
40
|
Ito D, Takashima E, Yamasaki T, Hatano S, Hasegawa T, Miura K, Morita M, Thongkukiatkul A, Diakite M, Long CA, Sattabongkot J, Udomsangpetch R, Iriko H, Ishino T, Tsuboi T. Antibodies against a Plasmodium falciparum RON12 inhibit merozoite invasion into erythrocytes. Parasitol Int 2018; 68:87-91. [PMID: 30342119 DOI: 10.1016/j.parint.2018.10.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 10/10/2018] [Accepted: 10/16/2018] [Indexed: 10/28/2022]
Abstract
Proteins coating Plasmodium merozoite surface and secreted from its apical organelles are considered as promising vaccine candidates for blood-stage malaria. The rhoptry neck protein 12 of Plasmodium falciparum (PfRON12) was recently reported as a protein specifically expressed in schizonts and localized to the rhoptry neck of merozoites. Here, we assessed its potential as a vaccine candidate. We expressed a recombinant PfRON12 protein by a wheat germ cell-free system to obtain anti-PfRON12 antibody. Immunoblot analysis of schizont lysates detected a single band at approximately 40 kDa under reducing conditions, consistent with the predicted molecular weight. Additionally, anti-PfRON12 antibody recognized a single band around 80 kDa under non-reducing conditions, suggesting native PfRON12 forms a disulfide-bond-mediated multimer. Immunofluorescence assay and immunoelectron microscopy revealed that PfRON12 localized to the rhoptry neck of merozoites in schizonts and to the surface of free merozoites. The biological activity of anti-PfRON12 antibody was tested by in vitro growth inhibition assay (GIA), and the rabbit antibodies significantly inhibited merozoite invasion of erythrocytes. We then investigated whether PfRON12 is immunogenic in P. falciparum-infected individuals. The sera from P. falciparum infected individuals in Thailand and Mali reacted with the recombinant PfRON12. Furthermore, human anti-PfRON12 antibodies affinity-purified from Malian serum samples inhibited merozoite invasion of erythrocytes in vitro. Moreover, pfron12 is highly conserved with only 4 non-synonymous mutations in the coding sequence from approximately 200 isolates deposited in PlasmoDB. These results suggest that PfRON12 might be a potential blood-stage vaccine candidate antigen against P. falciparum.
Collapse
Affiliation(s)
- Daisuke Ito
- Division of Medical Zoology, Department of Microbiology and Immunology, Faculty of Medicine, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan.
| | - Tsutomu Yamasaki
- Department of Molecular and Clinical Diagnosis, School of Pharmacy, Shujitsu University, Okayama 703-8516, Japan
| | - Shinya Hatano
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan
| | - Tomoyuki Hasegawa
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, United States
| | - Masayuki Morita
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan
| | - Amporn Thongkukiatkul
- Department of Biology, Faculty of Science, Burapha University, Chonburi 20131, Thailand
| | - Mahamadou Diakite
- Malaria Research and Training Centre, Faculty of Medicine, Pharmacy and Odonto-stomatology, University of Sciences, Techniques, and Technologies of Bamako, Point G, BP 1805, Mali
| | - Carole A Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, United States
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Rachanee Udomsangpetch
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Salaya, Nakhosn Pathom, 73170, Thailand
| | - Hideyuki Iriko
- Division of Global Infectious Diseases, Department of Public Health, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, Hyogo 654-0142, Japan
| | - Tomoko Ishino
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon Ehime, 791-0295, Japan
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan.
| |
Collapse
|
41
|
|
42
|
Arévalo-Pinzón G, González-González M, Suárez CF, Curtidor H, Carabias-Sánchez J, Muro A, LaBaer J, Patarroyo MA, Fuentes M. Self-assembling functional programmable protein array for studying protein-protein interactions in malaria parasites. Malar J 2018; 17:270. [PMID: 30016987 PMCID: PMC6050706 DOI: 10.1186/s12936-018-2414-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 07/10/2018] [Indexed: 12/30/2022] Open
Abstract
Background Plasmodium vivax is the most widespread malarial species, causing significant morbidity worldwide. Knowledge is limited regarding the molecular mechanism of invasion due to the lack of a continuous in vitro culture system for these species. Since protein–protein and host–cell interactions play an essential role in the microorganism’s invasion and replication, elucidating protein function during invasion is critical when developing more effective control methods. Nucleic acid programmable protein array (NAPPA) has thus become a suitable technology for studying protein–protein and host–protein interactions since producing proteins through the in vitro transcription/translation (IVTT) method overcomes most of the drawbacks encountered to date, such as heterologous protein production, stability and purification. Results Twenty P. vivax proteins on merozoite surface or in secretory organelles were selected and successfully cloned using gateway technology. Most constructs were displayed in the array expressed in situ, using the IVTT method. The Pv12 protein was used as bait for evaluating array functionality and co-expressed with P. vivax cDNA display in the array. It was found that Pv12 interacted with Pv41 (as previously described), as well as PvMSP142kDa, PvRBP1a, PvMSP8 and PvRAP1. Conclusions NAPPA is a high-performance technique enabling co-expression of bait and query in situ, thereby enabling interactions to be analysed rapidly and reproducibly. It offers a fresh alternative for studying protein–protein and ligand–receptor interactions regarding a parasite which is difficult to cultivate (i.e. P. vivax). Electronic supplementary material The online version of this article (10.1186/s12936-018-2414-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gabriela Arévalo-Pinzón
- Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50 # 26-20, Bogotá, Colombia.,PhD Programme in Biomedical and Biological Sciences, Universidad del Rosario, Carrera 24 # 63C-69, Bogotá, Colombia
| | - María González-González
- Proteomics Unit, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007, Salamanca, Spain.,Department of Medicine and General Cytometry Service-Nucleus, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007, Salamanca, Spain
| | - Carlos Fernando Suárez
- Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50 # 26-20, Bogotá, Colombia.,Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A.), Calle 222 # 55-37, Bogotá, Colombia
| | - Hernando Curtidor
- Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50 # 26-20, Bogotá, Colombia.,School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 # 63C-69, Bogotá, Colombia
| | | | - Antonio Muro
- Unidad de Investigación Enfermedades Infecciosas y Tropicales (e-INTRO), Instituto de Investigación Biomédica de Salamanca-Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (IBSAL-CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Universitario Miguel de Unamuno s/n, 37007, Salamanca, Spain
| | - Joshua LaBaer
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Manuel Alfonso Patarroyo
- Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50 # 26-20, Bogotá, Colombia.,School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 # 63C-69, Bogotá, Colombia
| | - Manuel Fuentes
- Proteomics Unit, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007, Salamanca, Spain. .,Department of Medicine and General Cytometry Service-Nucleus, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007, Salamanca, Spain.
| |
Collapse
|
43
|
Abstract
Probabilistic modeling is fundamental to the statistical analysis of complex data. In addition to forming a coherent description of the data-generating process, probabilistic models enable parameter inference about given datasets. This procedure is well developed in the Bayesian perspective, in which one infers probability distributions describing to what extent various possible parameters agree with the data. In this paper, we motivate and review probabilistic modeling for adaptive immune receptor repertoire data then describe progress and prospects for future work, from germline haplotyping to adaptive immune system deployment across tissues. The relevant quantities in immune sequence analysis include not only continuous parameters such as gene use frequency but also discrete objects such as B-cell clusters and lineages. Throughout this review, we unravel the many opportunities for probabilistic modeling in adaptive immune receptor analysis, including settings for which the Bayesian approach holds substantial promise (especially if one is optimistic about new computational methods). From our perspective, the greatest prospects for progress in probabilistic modeling for repertoires concern ancestral sequence estimation for B-cell receptor lineages, including uncertainty from germline genotype, rearrangement, and lineage development.
Collapse
Affiliation(s)
- Branden Olson
- Computational Biology Program Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N., Mail stop: M1-B514 Seattle, WA 98109-1024 phone: +1 206 667 7318
| | - Frederick A. Matsen
- Computational Biology Program Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N., Mail stop: M1-B514 Seattle, WA 98109-1024 phone: +1 206 667 7318
| |
Collapse
|
44
|
Sakamoto H, Takeo S, Takashima E, Miura K, Kanoi BN, Kaneko T, Han ET, Tachibana M, Matsuoka K, Sattabongkot J, Udomsangpetch R, Ishino T, Tsuboi T. Identification of target proteins of clinical immunity to Plasmodium falciparum in a region of low malaria transmission. Parasitol Int 2017; 67:203-208. [PMID: 29217416 DOI: 10.1016/j.parint.2017.12.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/29/2017] [Accepted: 12/04/2017] [Indexed: 01/03/2023]
Abstract
The target molecules of antibodies against falciparum malaria remain largely unknown. Recently we have identified multiple proteins as targets of immunity against Plasmodium falciparum using African serum samples. To investigate whether potential targets of clinical immunity differ with transmission intensity, we assessed immune responses in residents of low malaria transmission region in Thailand. Malaria asymptomatic volunteers (Asy: n=19) and symptomatic patients (Sym: n=21) were enrolled into the study. Serum immunoreactivity to 186 wheat germ cell-free system (WGCFS)-synthesized recombinant P. falciparum asexual-blood stage proteins were determined by AlphaScreen, and subsequently compared between the study groups. Forty proteins were determined as immunoreactive with antibody responses to 35 proteins being higher in Asy group than in Sym group. Among the 35 proteins, antibodies to MSP3, MSPDBL1, RH2b, and MSP7 were significantly higher in Asy than Sym (unadjusted p<0.005) suggesting these antigens may have a protective role in clinical malaria. MSP3 reactivity remained significantly different between Asy and Sym groups even after multiple comparison adjustments (adjusted p=0.033). Interestingly, while our two preceding studies using African sera were conducted differently (e.g., cross-sectional vs. longitudinal design, observed clinical manifestation vs. functional activity), those studies similarly identified MSP3 and MSPDBL1 as potential targets of protective immunity. This study further provides a strong rationale for the application of WGCFS-based immunoprofiling to malaria vaccine candidate and biomarker discovery even in low or reduced malaria transmission settings.
Collapse
Affiliation(s)
- Hirokazu Sakamoto
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan
| | - Satoru Takeo
- 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
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Bernard N Kanoi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan
| | - Takamasa Kaneko
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon, Gangwon-do 200-701, Republic of Korea
| | - Mayumi Tachibana
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Ehime 791-0295, Japan
| | - Kazuhiro Matsuoka
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Ehime 791-0295, Japan
| | - Jetsumon Sattabongkot
- Department of Entomology, Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand
| | - Rachanee Udomsangpetch
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Salaya, Nakhosn Pathom 73170, Thailand
| | - Tomoko Ishino
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Ehime 791-0295, Japan
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan.
| |
Collapse
|
45
|
Mechanisms of naturally acquired immunity to P. falciparum and approaches to identify merozoite antigen targets. Parasitology 2017; 145:839-847. [PMID: 29144217 DOI: 10.1017/s0031182017001949] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Malaria is one the most serious infectious diseases with over 200 million clinical cases annually. Most cases of the severe disease are caused by Plasmodium falciparum. The blood stage of Plasmodium parasite is entirely responsible for malaria-associated pathology. The population most susceptible to severe malaria are children under the age of 5, with low levels of immunity. It is only after many years of repeated exposure that individuals living in endemic areas develop clinical immunity. This form of protection prevents clinical episodes by substantially reducing parasite burden. Naturally acquired immunity predominantly targets blood-stage parasites with antibody responses being the main mediators of protection. The targets of clinical immunity are the extracellular merozoite and the infected erythrocyte surface, with the extremely diverse PfEMP1 proteins the main target here. This observation provides a strong rationale that an effective anti-malaria vaccine targeting blood-stage parasites is achievable. Thus the identification of antigenic targets of naturally acquired immunity remains an important step towards the formulation of novel vaccine combinations before testing their efficacy in clinical trials. This review summarizes the main findings to date defining antigenic targets present on the extracellular merozoite associated with naturally acquired immunity to P. falciparum malaria.
Collapse
|
46
|
Haralambieva IH, Gibson MJ, Kennedy RB, Ovsyannikova IG, Warner ND, Grill DE, Poland GA. Characterization of rubella-specific humoral immunity following two doses of MMR vaccine using proteome microarray technology. PLoS One 2017; 12:e0188149. [PMID: 29145521 PMCID: PMC5690594 DOI: 10.1371/journal.pone.0188149] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 11/01/2017] [Indexed: 11/18/2022] Open
Abstract
Introduction//Background The lack of standardization of the currently used commercial anti-rubella IgG antibody assays leads to frequent misinterpretation of results for samples with low/equivocal antibody concentration. The use of alternative approaches in rubella serology could add new information leading to a fuller understanding of rubella protective immunity and neutralizing antibody response after vaccination. Methods We applied microarray technology to measure antibodies to all rubella virus proteins in 75 high and 75 low rubella virus-specific antibody responders after two MMR vaccine doses. These data were used in multivariate penalized logistic regression modeling of rubella-specific neutralizing antibody response after vaccination. Results We measured antibodies to all rubella virus structural proteins (i.e., the glycoproteins E1 and E2 and the capsid C protein) and to the non-structural protein P150. Antibody levels to each of these proteins were: correlated with the neutralizing antibody titer (p<0.006); demonstrated differences between the high and the low antibody responder groups (p<0.008); and were components of the model associated with/predictive of vaccine-induced rubella virus-specific neutralizing antibody titers (misclassification error = 0.2). Conclusion Our study supports the use of this new technology, as well as the use of antibody profiles/patterns (rather than single antibody measures) as biomarkers of neutralizing antibody response and correlates of protective immunity in rubella virus serology.
Collapse
Affiliation(s)
- Iana H. Haralambieva
- Mayo Vaccine Research Group, Mayo Clinic and Foundation, Rochester, MN, United States of America
| | - Michael J. Gibson
- Mayo Vaccine Research Group, Mayo Clinic and Foundation, Rochester, MN, United States of America
| | - Richard B. Kennedy
- Mayo Vaccine Research Group, Mayo Clinic and Foundation, Rochester, MN, United States of America
| | - Inna G. Ovsyannikova
- Mayo Vaccine Research Group, Mayo Clinic and Foundation, Rochester, MN, United States of America
| | - Nathaniel D. Warner
- Division of Biomedical Statistics and Informatics- Department of Health Science Research, Mayo Clinic and Foundation, Rochester, MN, United States of America
| | - Diane E. Grill
- Division of Biomedical Statistics and Informatics- Department of Health Science Research, Mayo Clinic and Foundation, Rochester, MN, United States of America
| | - Gregory A. Poland
- Mayo Vaccine Research Group, Mayo Clinic and Foundation, Rochester, MN, United States of America
- * E-mail:
| |
Collapse
|
47
|
Schussek S, Trieu A, Apte SH, Sidney J, Sette A, Doolan DL. Novel Plasmodium antigens identified via genome-based antibody screen induce protection associated with polyfunctional T cell responses. Sci Rep 2017; 7:15053. [PMID: 29118376 PMCID: PMC5678182 DOI: 10.1038/s41598-017-15354-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 10/25/2017] [Indexed: 12/13/2022] Open
Abstract
The development of vaccines against complex intracellular pathogens, such as Plasmodium spp., where protection is likely mediated by cellular immune responses, has proven elusive. The availability of whole genome, proteome and transcriptome data has the potential to advance rational vaccine development but yet there are no licensed vaccines against malaria based on antigens identified from genomic data. Here, we show that the Plasmodium yoelii orthologs of four Plasmodium falciparum proteins identified by an antibody-based genome-wide screening strategy induce a high degree of sterile infection-blocking protection against sporozoite challenge in a stringent rodent malaria model. Protection increased in multi-antigen formulations. Importantly, protection was highly correlated with the induction of multifunctional triple-positive T cells expressing high amounts of IFN-γ, IL-2 and TNF. These data demonstrate that antigens identified by serological screening are targets of multifunctional cellular immune responses that correlate with protection. Our results provide experimental validation for the concept of rational vaccine design from genomic sequence data.
Collapse
Affiliation(s)
- Sophie Schussek
- QIMR Berghofer Medical Research Institute, Infectious Diseases Programme, Herston, QLD 4006, Australia.,University of Queensland, School of Medicine, Herston, QLD 4006, Australia
| | - Angela Trieu
- QIMR Berghofer Medical Research Institute, Infectious Diseases Programme, Herston, QLD 4006, Australia
| | - Simon H Apte
- QIMR Berghofer Medical Research Institute, Infectious Diseases Programme, Herston, QLD 4006, Australia
| | - John Sidney
- La Jolla Institute of Allergy and Immunology, San Diego, CA, 92121, USA
| | - Alessandro Sette
- La Jolla Institute of Allergy and Immunology, San Diego, CA, 92121, USA
| | - Denise L Doolan
- QIMR Berghofer Medical Research Institute, Infectious Diseases Programme, Herston, QLD 4006, Australia. .,Centre for Biosecurity and Tropical Infectious Diseases, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD 4879, Australia.
| |
Collapse
|
48
|
Perraut R, Varela ML, Joos C, Diouf B, Sokhna C, Mbengue B, Tall A, Loucoubar C, Touré A, Mercereau-Puijalon O. Association of antibodies to Plasmodium falciparum merozoite surface protein-4 with protection against clinical malaria. Vaccine 2017; 35:6720-6726. [PMID: 29042203 DOI: 10.1016/j.vaccine.2017.10.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 09/10/2017] [Accepted: 10/05/2017] [Indexed: 11/17/2022]
Abstract
Identification of parasite antigens targeted by immune effector mechanisms that confer protection against malaria is important for the design of a multi-component malaria vaccine. Here, the association of antibodies reacting with the Plasmodium falciparum merozoite surface protein-4 (MSP4) with protection against clinical malaria was investigated in a Senegalese community living in an area of moderate, seasonal malaria transmission. Blood samples were collected at the end of an 8-month long dry season without any recorded parasite transmission from 206 residents enrolled in a prospective follow-up study. Active daily clinical monitoring was implemented during the subsequent five months. Entomologic monitoring documented parasite transmission during the first three months of follow-up. Serum IgG levels were determined by ELISA against three MSP4 baculovirus-encoded recombinant protein constructs, namely the full-length MSP4p40, MSP4p30 devoid of a highly polymorphic sequence stretch and the conserved C-terminal EGF-containing MSP4p20, as well as against a merozoite crude extract. Community seroprevalence against all three constructs was quite high, the lowest being against MSP4p30. Seroprevalence and antibody levels against the three MSP4 constructs were age-dependent. IgG1 dominated the anti-MSP4p20 responses, while both IgG1 and IgG3 were observed against MSP4p40. Anti-MSP4 antibodies were associated with the antibody-dependent respiratory burst (ADRB) activity in a functional assay of merozoite phagocytosis by polymorphonuclear cells. Importantly, high antibody levels against each of the three MSP4 constructs at the end of the dry season were associated with reduced morbidity during the subsequent transmission season in an age-adjusted Poisson regression model (IRR = 0.65 [0.50-0.83], P<0.001 for responses over the median values). These data are consistent with a protective role for the naturally acquired anti-MSP4 antibodies and support further development of MSP4 as a candidate component of malaria vaccine.
Collapse
Affiliation(s)
- Ronald Perraut
- Unité d'Immunologie, Institut Pasteur de Dakar, Senegal.
| | | | | | - Babacar Diouf
- Unité d'Immunologie, Institut Pasteur de Dakar, Senegal
| | - Cheikh Sokhna
- Institut de Recherche pour le Développement (IRD), URMITE, UMR 198, Dakar, Senegal
| | | | - Adama Tall
- Unité d'Epidémiologie, Institut Pasteur de Dakar, Senegal
| | - Cheikh Loucoubar
- Institut Pasteur de Dakar, G4 Biostatistiques Bioinformatique et Modélisation, Dakar, Senegal
| | | | - Odile Mercereau-Puijalon
- Institut Pasteur, Département Parasites et Insectes Vecteurs, 25 Rue du Dr. Roux, 75015 Paris, France
| |
Collapse
|
49
|
Mother-Newborn Pairs in Malawi Have Similar Antibody Repertoires to Diverse Malaria Antigens. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:CVI.00136-17. [PMID: 28835359 DOI: 10.1128/cvi.00136-17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 07/31/2017] [Indexed: 11/20/2022]
Abstract
Maternal antibodies may play a role in protecting newborns against malaria disease. Plasmodium falciparum parasite surface antigens are diverse, and protection from infection requires allele-specific immunity. Although malaria-specific antibodies have been shown to cross the placenta, the extent to which antibodies that respond to the full repertoire of diverse antigens are transferred from the mother to the infant has not been explored. Understanding the breadth of maternal antibody responses and to what extent these antibodies are transferred to the child can inform vaccine design and evaluation. We probed plasma from cord blood and serum from mothers at delivery using a customized protein microarray that included variants of malaria vaccine target antigens to assess the intensity and breadth of seroreactivity to three malaria vaccine candidate antigens in mother-newborn pairs in Malawi. Among the 33 paired specimens that were assessed, mothers and newborns had similar intensity and repertoire of seroreactivity. Maternal antibody levels against vaccine candidate antigens were the strongest predictors of infant antibody levels. Placental malaria did not significantly impair transplacental antibody transfer. However, mothers with placental malaria had significantly higher antibody levels against these blood-stage antigens than mothers without placental malaria. The repertoire and levels of infant antibodies against a wide range of malaria vaccine candidate antigen variants closely mirror maternal levels in breadth and magnitude regardless of evidence of placental malaria. Vaccinating mothers with an effective malaria vaccine during pregnancy may induce high and potentially protective antibody repertoires in newborns.
Collapse
|
50
|
Tuju J, Kamuyu G, Murungi LM, Osier FHA. Vaccine candidate discovery for the next generation of malaria vaccines. Immunology 2017; 152:195-206. [PMID: 28646586 PMCID: PMC5588761 DOI: 10.1111/imm.12780] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 06/12/2017] [Accepted: 06/19/2017] [Indexed: 12/21/2022] Open
Abstract
Although epidemiological observations, IgG passive transfer studies and experimental infections in humans all support the feasibility of developing highly effective malaria vaccines, the precise antigens that induce protective immunity remain uncertain. Here, we review the methodologies applied to vaccine candidate discovery for Plasmodium falciparum malaria from the pre- to post-genomic era. Probing of genomic and cDNA libraries with antibodies of defined specificities or functional activity predominated the former, whereas reverse vaccinology encompassing high throughput in silico analyses of genomic, transcriptomic or proteomic parasite data sets is the mainstay of the latter. Antibody-guided vaccine design spanned both eras but currently benefits from technological advances facilitating high-throughput screening and downstream applications. We make the case that although we have exponentially increased our ability to identify numerous potential vaccine candidates in a relatively short space of time, a significant bottleneck remains in their validation and prioritization for evaluation in clinical trials. Longitudinal cohort studies provide supportive evidence but results are often conflicting between studies. Demonstration of antigen-specific antibody function is valuable but the relative importance of one mechanism over another with regards to protection remains undetermined. Animal models offer useful insights but may not accurately reflect human disease. Challenge studies in humans are preferable but prohibitively expensive. In the absence of reliable correlates of protection, suitable animal models or a better understanding of the mechanisms underlying protective immunity in humans, vaccine candidate discovery per se may not be sufficient to provide the paradigm shift necessary to develop the next generation of highly effective subunit malaria vaccines.
Collapse
Affiliation(s)
- James Tuju
- KEMRI‐Wellcome Trust Research ProgrammeCentre for Geographic Medicine CoastKilifiKenya
- Department of BiochemistryPwani UniversityKilifiKenya
| | - Gathoni Kamuyu
- KEMRI‐Wellcome Trust Research ProgrammeCentre for Geographic Medicine CoastKilifiKenya
| | - Linda M. Murungi
- KEMRI‐Wellcome Trust Research ProgrammeCentre for Geographic Medicine CoastKilifiKenya
| | - Faith H. A. Osier
- KEMRI‐Wellcome Trust Research ProgrammeCentre for Geographic Medicine CoastKilifiKenya
- Centre for Infectious DiseasesHeidelberg University HospitalHeidelbergGermany
- Department of Biomedical SciencesPwani UniversityKilifiKenya
| |
Collapse
|