1
|
Henry JM, Carter AR, Wu SL, Smith DL. A Probabilistic Synthesis of Malaria Epidemiology: Exposure, Infection, Parasite Densities, and Detection. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.24.25324561. [PMID: 40196243 PMCID: PMC11974775 DOI: 10.1101/2025.03.24.25324561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
The epidemiology of Plasmodium falciparum malaria presents a unique set of challenges due to the complicated dynamics of infection, immunity, disease, and detection. Studies of malaria epidemiology commonly measure malaria parasite densities or prevalence, but since malaria is so complex with so many factors to consider, a complete mathematical synthesis of malaria epidemiology has been elusive. Here, we take a new approach. From a simple model of malaria exposure and infection in human cohorts as they age, we develop random variables describing the multiplicity of infection (MoI) and the age of infection (AoI). Next, using the MoI and AoI distributions, we develop random variables describing parasite densities, parasite counts, and detection. We also derived a random variable describing the age of the youngest infection (AoY), which can be used to compute approximate parasite densities in complex infections. Finally, we derive a simple system of differential equations with hybrid variables that track the mean MoI, AoI and AoY, and we show it matches the complex probabilistic system with reasonable accuracy. We can thus compute the state of any individual chosen at random from the population in two ways. The same approach - pairing random variables and hybrid models - can be extended to model other features of malaria epidemiology, including disease, malaria immunity, treatment and chemoprotection, and infectiousness. The computational simplicity of hybrid models has some advantages over compartmental models and stochastic individual-based models, and with the supporting probabilistic framework, provide a sound basis for a synthesis of observational malaria epidemiology.
Collapse
Affiliation(s)
- John M. Henry
- College of the Environment, University of Washington, Seattle, WA, USA
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Austin R. Carter
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Sean L. Wu
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - David L. Smith
- Department of Health Metrics Sciences, School of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
2
|
Souffou M, Dechavanne C, Kammoun Z, Viwami F, Gaugué I, Beldjoudi N, Dechavanne S, Sare N, Garcia A, Dambrun M, Migot-Nabias F. Functionality of Toxoplasma gondii antibodies in a population of Beninese pregnant women exposed to malaria. Sci Rep 2025; 15:9303. [PMID: 40102442 PMCID: PMC11920409 DOI: 10.1038/s41598-025-91803-5] [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: 10/24/2024] [Accepted: 02/24/2025] [Indexed: 03/20/2025] Open
Abstract
Plasmodium falciparum and Toxoplasma gondii are two apicomplexan parasites that can lead to severe complications for the newborn when contracted during pregnancy. This study explores the cross-reactivity of antibodies specific to both pathogens in pregnant women, exposed or not to malaria. The antibody response against full-length recombinant antigens from P. falciparum (PfAMA1, Pfs48/45) and T. gondii (TgAMA1, TgSAG1, TgGRA7), selected for their strong immunogenicity, was analysed on 150 plasma samples from women residing in Benin or France. The antibody functionality was assessed using P. falciparum in vitro Growth Inhibition Assay (GIA). As the main results, toxoplasmosis seropositive women with an ongoing P. falciparum infection better inhibited P. falciparum invasion compared to toxoplasmosis seronegative women (34.6% vs. 17.2%, p ≤ 0.01). Women with positive serologies for both parasites presented a significantly higher inhibition of P. falciparum invasion compared to those only seropositive for malaria (coef = 6.27, p = 0.076) in reference with double-negative women (coef = 11.35, p = 0.001). These data suggest that plasma samples containing anti-T. gondii IgG may contribute reducing the development of P. falciparum parasites. This study provides insight into the immune dynamics of the co-infection by these two apicomplexans with potential implications for developing cross-protective vaccines and therapies.
Collapse
Grants
- PhD scholarship Conseil Départemental de Mayotte
- IdEx 2019 "Dynamique Recherche", PlasDCty Université Paris Cité, France
- DHU 'Risks and pregnancy", PRIDE 2016, TOXODIAG AP-HP Nord et Université Paris Cité
- DHU 'Risks and pregnancy", PRIDE 2016, TOXODIAG AP-HP Nord et Université Paris Cité
- grant 0602DIRmba, 2018, CoaLa Institut de Médecine et d'Epidémiologie Appliquée
- grant 0602DIRmba, 2018, CoaLa Institut de Médecine et d'Epidémiologie Appliquée
- ANR-19-CE44-0004, IgName Agence Nationale de la Recherche
Collapse
Affiliation(s)
- Mariama Souffou
- Université Paris Cité, MERIT, IRD, Inserm, Paris, Paris, F-75006, France
| | - Célia Dechavanne
- Université Paris Cité, MERIT, IRD, Inserm, Paris, Paris, F-75006, France
| | - Zaineb Kammoun
- Université Paris Cité, Cibles Thérapeutiques et Conception de Médicaments (CiTCoM), CNRS, Paris, France
| | - Firmine Viwami
- Université Paris Cité, MERIT, IRD, Inserm, Paris, Paris, F-75006, France
| | - Isabelle Gaugué
- Université Paris Cité, MERIT, IRD, Inserm, Paris, Paris, F-75006, France
- Genetics and Developmental Biology, Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Paris, 75005, France
| | - Naima Beldjoudi
- Epidemiology and Clinical Research Department, GH Paris Nord Val de Seine, Assistance Publique - Hôpitaux de Paris, Paris, France
| | | | - Nawal Sare
- Centre d'Etude et de Recherche sur les Pathologies Associées à la Grossesse et à L'Enfance, Cotonou, Bénin
| | - André Garcia
- Université Paris Cité, MERIT, IRD, Inserm, Paris, Paris, F-75006, France
| | - Magalie Dambrun
- Université Paris Cité, MERIT, IRD, Inserm, Paris, Paris, F-75006, France.
| | | |
Collapse
|
3
|
David-Vieira C, Carpinter BA, Bezerra-Bellei J, Machado L, Raimundo FO, Rodolphi CM, Renhe DC, Guedes IR, Gonçalves FMM, Pereira LPC, Ferreira MV, Nascimento HLDS, Neto AF, Gomes FR, Rocha VN, Castro JMA, Scopel KKG. Lung Damage Induced by Plasmodium berghei ANKA in Murine Model of Malarial Infection is Mitigated by Dietary Supplementation with DHA-Rich Omega-3. ACS Infect Dis 2024; 10:3607-3617. [PMID: 39303151 PMCID: PMC11474944 DOI: 10.1021/acsinfecdis.4c00482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/30/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024]
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are severe complications that can occur in infections caused by any Plasmodium species. Due to the high lethality rate and the lack of specific treatment for ALI/ARDS, studies aimed at understanding and searching for treatment strategies for such complications have been fundamental. Here, we investigated the protective role of dietary supplementation with DHA-rich fish oil against lung damage induced by Plasmodium berghei ANKA in a murine model. Our results demonstrated that alveolar vascular damage, lung edema, and histopathological alterations were significantly reduced in mice that received dietary supplementation compared to those that did not receive the supplementation. Furthermore, a significant reduction in the number of CD8+ T lymphocytes, in addition to reduced infiltration of inflammatory cells in the bronchoalveolar lavage fluid was also observed. High levels of IL-10, but not of TNF-α and IFN-γ, were also observed in infected mice that received the supplementation, along with a reduction in local oxidative stress. Together, the data suggest that dietary supplementation with DHA-rich fish oil in malarial endemic areas may help reduce lung damage resulting from the infection, thus preventing worsening of the condition.
Collapse
Affiliation(s)
- Carolina David-Vieira
- Research
Centre of Parasitology, Department of Parasitology, Microbiology and
Immunology and Post-Graduate Program in Biological Science, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | - Barbara Albuquerque Carpinter
- Research
Centre of Parasitology, Department of Parasitology, Microbiology and
Immunology and Post-Graduate Program in Biological Science, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | - Jéssica
Correia Bezerra-Bellei
- Research
Centre of Parasitology, Department of Parasitology, Microbiology and
Immunology and Post-Graduate Program in Biological Science, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | - Letícia
Ferreira Machado
- Research
Centre of Parasitology, Department of Parasitology, Microbiology and
Immunology and Post-Graduate Program in Biological Science, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | - Felipe Oliveira Raimundo
- Research
Centre of Parasitology, Department of Parasitology, Microbiology and
Immunology and Post-Graduate Program in Biological Science, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | - Cinthia Magalhães Rodolphi
- Research
Centre of Parasitology, Department of Parasitology, Microbiology and
Immunology and Post-Graduate Program in Biological Science, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | - Daniela Chaves Renhe
- Research
Centre of Parasitology, Department of Parasitology, Microbiology and
Immunology and Post-Graduate Program in Biological Science, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | - Isabella Rodrigues
Nogueira Guedes
- Research
Centre of Parasitology, Department of Parasitology, Microbiology and
Immunology and Post-Graduate Program in Biological Science, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | - Fernanda Mikaela Moreira Gonçalves
- Research
Centre of Parasitology, Department of Parasitology, Microbiology and
Immunology and Post-Graduate Program in Biological Science, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | - Ludmila Ponce
Monken Custódio Pereira
- Research
Centre of Parasitology, Department of Parasitology, Microbiology and
Immunology and Post-Graduate Program in Biological Science, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | | | - Haroldo Lobo dos Santos Nascimento
- Research
Centre of Pathology and Veterinary Histology, Department of Veterinary
Medicine, Federal University of Juiz de
Fora, Juiz de
Fora 36036-900, Brazil
| | - Adolfo Firmino Neto
- Research
Centre of Pathology and Veterinary Histology, Department of Veterinary
Medicine, Federal University of Juiz de
Fora, Juiz de
Fora 36036-900, Brazil
| | | | - Vinicius Novaes Rocha
- Research
Centre of Pathology and Veterinary Histology, Department of Veterinary
Medicine, Federal University of Juiz de
Fora, Juiz de
Fora 36036-900, Brazil
| | - Juciane Maria
de Andrade Castro
- Research
Centre of Parasitology, Department of Parasitology, Microbiology and
Immunology and Post-Graduate Program in Biological Science, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | - Kézia Katiani Gorza Scopel
- Research
Centre of Parasitology, Department of Parasitology, Microbiology and
Immunology and Post-Graduate Program in Biological Science, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| |
Collapse
|
4
|
Tebben K, Yirampo S, Coulibaly D, Koné AK, Laurens MB, Stucke EM, Dembélé A, Tolo Y, Traoré K, Niangaly A, Berry AA, Kouriba B, Plowe CV, Doumbo OK, Lyke KE, Takala-Harrison S, Thera MA, Travassos MA, Serre D. Gene expression analyses reveal differences in children's response to malaria according to their age. Nat Commun 2024; 15:2021. [PMID: 38448421 PMCID: PMC10918175 DOI: 10.1038/s41467-024-46416-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 02/26/2024] [Indexed: 03/08/2024] Open
Abstract
In Bandiagara, Mali, children experience on average two clinical malaria episodes per year. However, even in the same transmission area, the number of uncomplicated symptomatic infections, and their parasitemia, can vary dramatically among children. We simultaneously characterize host and parasite gene expression profiles from 136 Malian children with symptomatic falciparum malaria and examine differences in the relative proportion of immune cells and parasite stages, as well as in gene expression, associated with infection and or patient characteristics. Parasitemia explains much of the variation in host and parasite gene expression, and infections with higher parasitemia display proportionally more neutrophils and fewer T cells, suggesting parasitemia-dependent neutrophil recruitment and/or T cell extravasation to secondary lymphoid organs. The child's age also strongly correlates with variations in gene expression: Plasmodium falciparum genes associated with age suggest that older children carry more male gametocytes, while variations in host gene expression indicate a stronger innate response in younger children and stronger adaptive response in older children. These analyses highlight the variability in host responses and parasite regulation during P. falciparum symptomatic infections and emphasize the importance of considering the children's age when studying and treating malaria infections.
Collapse
Affiliation(s)
- Kieran Tebben
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Salif Yirampo
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Drissa Coulibaly
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Abdoulaye K Koné
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Matthew B Laurens
- Malaria Research Program, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Emily M Stucke
- Malaria Research Program, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ahmadou Dembélé
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Youssouf Tolo
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Karim Traoré
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Amadou Niangaly
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Andrea A Berry
- Malaria Research Program, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bourema Kouriba
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Christopher V Plowe
- Malaria Research Program, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ogobara K Doumbo
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Kirsten E Lyke
- Malaria Research Program, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Shannon Takala-Harrison
- Malaria Research Program, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mahamadou A Thera
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Mark A Travassos
- Malaria Research Program, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - David Serre
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA.
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
5
|
Macalinao ML, Inoue SI, Tsogtsaikhan S, Matsumoto H, Bayarsaikhan G, Jian JY, Kimura K, Yasumizu Y, Inoue T, Yoshida H, Hafalla J, Kimura D, Yui K. IL-27 produced during acute malaria infection regulates Plasmodium-specific memory CD4 + T cells. EMBO Mol Med 2023; 15:e17713. [PMID: 37855243 DOI: 10.15252/emmm.202317713] [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: 03/14/2023] [Revised: 09/27/2023] [Accepted: 09/28/2023] [Indexed: 10/20/2023] Open
Abstract
Malaria infection elicits both protective and pathogenic immune responses, and IL-27 is a critical cytokine that regulate effector responses during infection. Here, we identified a critical window of CD4+ T cell responses that is targeted by IL-27. Neutralization of IL-27 during acute infection with Plasmodium chabaudi expanded specific CD4+ T cells, which were maintained at high levels thereafter. In the chronic phase, Plasmodium-specific CD4+ T cells in IL-27-neutralized mice consisted mainly of CD127+ KLRG1- and CD127- KLRG1+ subpopulations that displayed distinct cytokine production, proliferative capacity, and are maintained in a manner independent of active infection. Single-cell RNA-seq analysis revealed that these CD4+ T cell subsets formed independent clusters that express unique Th1-type genes. These IL-27-neutralized mice exhibited enhanced cellular and humoral immune responses and protection. These findings demonstrate that IL-27, which is produced during the acute phase of malaria infection, inhibits the development of unique Th1 memory precursor CD4+ T cells, suggesting potential implications for the development of vaccines and other strategic interventions.
Collapse
Affiliation(s)
- Maria Lourdes Macalinao
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Shin-Ichi Inoue
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Sanjaadorj Tsogtsaikhan
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Hirotaka Matsumoto
- School of Information and Data Sciences, Nagasaki University, Nagasaki, Japan
| | - Ganchimeg Bayarsaikhan
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Jiun-Yu Jian
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Kazumi Kimura
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Yoshiaki Yasumizu
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, Japan
| | - Tsuyoshi Inoue
- Department of Physiology of Visceral Function and Body Fluid, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Hiroki Yoshida
- Division of Molecular and Cellular Immunoscience, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, Saga, Japan
| | - Julius Hafalla
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Daisuke Kimura
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Katsuyuki Yui
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
- Shionogi Global Infectious Diseases Division, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
6
|
Tebben K, Yirampo S, Coulibaly D, Koné AK, Laurens MB, Stucke EM, Dembélé A, Tolo Y, Traoré K, Niangaly A, Berry AA, Kouriba B, Plowe CV, Doumbo OK, Lyke KE, Takala-Harrison S, Thera MA, Travassos MA, Serre D. Gene expression analyses reveal differences in children's response to malaria according to their age. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.24.563751. [PMID: 37961701 PMCID: PMC10634788 DOI: 10.1101/2023.10.24.563751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
In Bandiagara, Mali, children experience on average two clinical malaria episodes per season. However, even in the same transmission area, the number of uncomplicated symptomatic infections, and their parasitemia, vary dramatically among children. To examine the factors contributing to these variations, we simultaneously characterized the host and parasite gene expression profiles from 136 children with symptomatic falciparum malaria and analyzed the expression of 9,205 human and 2,484 Plasmodium genes. We used gene expression deconvolution to estimate the relative proportion of immune cells and parasite stages in each sample and to adjust the differential gene expression analyses. Parasitemia explained much of the variation in both host and parasite gene expression and revealed that infections with higher parasitemia had more neutrophils and fewer T cells, suggesting parasitemia-dependent neutrophil recruitment and/or T cell extravasation to secondary lymphoid organs. The child's age was also strongly correlated with gene expression variations. Plasmodium falciparum genes associated with age suggested that older children carried more male gametocytes, while host genes associated with age indicated a stronger innate response (through TLR and NLR signaling) in younger children and stronger adaptive immunity (through TCR and BCR signaling) in older children. These analyses highlight the variability in host responses and parasite regulation during P. falciparum symptomatic infections and emphasize the importance of considering the children's age when studying and treating malaria infections.
Collapse
Affiliation(s)
- Kieran Tebben
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine; Baltimore, USA
| | - Salif Yirampo
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies; Bamako, Mali
| | - Drissa Coulibaly
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies; Bamako, Mali
| | - Abdoulaye K. Koné
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies; Bamako, Mali
| | - Matthew B. Laurens
- Malaria Research Program, Center for Vaccine Development and Global Health, University of Maryland School of Medicine; Baltimore, USA
| | - Emily M. Stucke
- Malaria Research Program, Center for Vaccine Development and Global Health, University of Maryland School of Medicine; Baltimore, USA
| | - Ahmadou Dembélé
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies; Bamako, Mali
| | - Youssouf Tolo
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies; Bamako, Mali
| | - Karim Traoré
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies; Bamako, Mali
| | - Amadou Niangaly
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies; Bamako, Mali
| | - Andrea A. Berry
- Malaria Research Program, Center for Vaccine Development and Global Health, University of Maryland School of Medicine; Baltimore, USA
| | - Bourema Kouriba
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies; Bamako, Mali
| | - Christopher V. Plowe
- Malaria Research Program, Center for Vaccine Development and Global Health, University of Maryland School of Medicine; Baltimore, USA
| | - Ogobara K Doumbo
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies; Bamako, Mali
| | - Kirsten E. Lyke
- Malaria Research Program, Center for Vaccine Development and Global Health, University of Maryland School of Medicine; Baltimore, USA
| | - Shannon Takala-Harrison
- Malaria Research Program, Center for Vaccine Development and Global Health, University of Maryland School of Medicine; Baltimore, USA
| | - Mahamadou A. Thera
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies; Bamako, Mali
| | - Mark A. Travassos
- Malaria Research Program, Center for Vaccine Development and Global Health, University of Maryland School of Medicine; Baltimore, USA
| | - David Serre
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine; Baltimore, USA
| |
Collapse
|
7
|
de Assis GMP, de Alvarenga DAM, Souza LBE, Sánchez-Arcila JC, Silva EFE, de Pina-Costa A, Gonçalves GHP, Souza JCDJ, Nunes AJD, Pissinatti A, Moreira SB, Torres LDM, Costa HL, Tinoco HDP, Pereira VDS, Soares IDS, de Sousa TN, Ntumngia FB, Adams JH, Kano FS, Hirano ZMB, Pratt-Riccio LR, Daniel-Ribeiro CT, Ferreira JO, Carvalho LH, Alves de Brito CF. IgM antibody responses against Plasmodium antigens in neotropical primates in the Brazilian Atlantic Forest. Front Cell Infect Microbiol 2023; 13:1169552. [PMID: 37829607 PMCID: PMC10565664 DOI: 10.3389/fcimb.2023.1169552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 08/11/2023] [Indexed: 10/14/2023] Open
Abstract
Introduction Zoonotic transmission is a challenge for the control and elimination of malaria. It has been recorded in the Atlantic Forest, outside the Amazon which is the endemic region in Brazil. However, only very few studies have assessed the antibody response, especially of IgM antibodies, in Neotropical primates (NP). Therefore, in order to contribute to a better understanding of the immune response in different hosts and facilitate the identification of potential reservoirs, in this study, naturally acquired IgM antibody responses against Plasmodium antigens were evaluated, for the first time, in NP from the Atlantic Forest. Methods The study was carried out using 154 NP samples from three different areas of the Atlantic Forest. IgM antibodies against peptides of the circumsporozoite protein (CSP) from different Plasmodium species and different erythrocytic stage antigens were detected by ELISA. Results Fifty-nine percent of NP had IgM antibodies against at least one CSP peptide and 87% against at least one Plasmodium vivax erythrocytic stage antigen. Levels of antibodies against PvAMA-1 were the highest compared to the other antigens. All families of NP showed IgM antibodies against CSP peptides, and, most strikingly, against erythrocytic stage antigens. Generalized linear models demonstrated that IgM positivity against PvCSP and PvAMA-1 was associated with PCR-detectable blood-stage malaria infection and the host being free-living. Interestingly, animals with IgM against both PvCSP and PvAMA-1 were 4.7 times more likely to be PCR positive than animals that did not have IgM for these two antigens simultaneously. Discussion IgM antibodies against different Plasmodium spp. antigens are present in NP from the Atlantic Forest. High seroprevalence and antibody levels against blood-stage antigens were observed, which had a significant association with molecular evidence of infection. IgM antibodies against CSP and AMA-1 may be used as a potential marker for the identification of NP infected with Plasmodium, which are reservoirs of malaria in the Brazilian Atlantic Forest.
Collapse
Affiliation(s)
- Gabriela Maíra Pereira de Assis
- Grupo de Pesquisa em Biologia Molecular e Imunologia da malária, Instituto René Rachou/Fiocruz Minas, Belo Horizonte, Brazil
| | | | - Luisa Braga e Souza
- Grupo de Pesquisa em Biologia Molecular e Imunologia da malária, Instituto René Rachou/Fiocruz Minas, Belo Horizonte, Brazil
| | - Juan Camilo Sánchez-Arcila
- School of Natural Sciences, Molecular and Cell Biology Department, University of California, Merced, Merced, CA, United States
| | | | - Anielle de Pina-Costa
- Laboratório de Doenças Febris Agudas, Instituto Nacional de Infectologia Evandro Chagas (INI), Fiocruz, Rio de Janeiro, Brazil
- Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal), Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz (IOC), Fiocruz, Rio de Janeiro, Brazil
- Escola de Enfermagem Aurora de Afonso Costa, Departamento de Doenças infecciosas e Parasitárias, Universidade Federal Fluminense, Niterói, Brazil
| | | | | | - Ana Julia Dutra Nunes
- Fundação Universidade Regional de Blumenau (FURB), Blumenau, Brazil
- Centro de Pesquisas Biológicas de Indaial, Indaial, Brazil
- Programa de conservação do Bugio Ruivo, Perini Business Park, Joinville, Brazil
| | - Alcides Pissinatti
- Centro de Primatologia do Rio de Janeiro (CPRJ), Instituto Estadual do Ambiente (INEA), Guapimirim, Brazil
- Centro Universitário Serra dos Órgãos (Unifeso), Teresópolis, Brazil
| | - Silvia Bahadian Moreira
- Centro de Primatologia do Rio de Janeiro (CPRJ), Instituto Estadual do Ambiente (INEA), Guapimirim, Brazil
| | - Leticia de Menezes Torres
- Grupo de Pesquisa em Biologia Molecular e Imunologia da malária, Instituto René Rachou/Fiocruz Minas, Belo Horizonte, Brazil
| | - Helena Lott Costa
- Grupo de Pesquisa em Biologia Molecular e Imunologia da malária, Instituto René Rachou/Fiocruz Minas, Belo Horizonte, Brazil
| | | | | | - Irene da Silva Soares
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Taís Nóbrega de Sousa
- Grupo de Pesquisa em Biologia Molecular e Imunologia da malária, Instituto René Rachou/Fiocruz Minas, Belo Horizonte, Brazil
| | - Francis Babila Ntumngia
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, FL, United States
| | - John H. Adams
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, FL, United States
| | - Flora Satiko Kano
- Grupo de Pesquisa em Biologia Molecular e Imunologia da malária, Instituto René Rachou/Fiocruz Minas, Belo Horizonte, Brazil
| | - Zelinda Maria Braga Hirano
- Fundação Universidade Regional de Blumenau (FURB), Blumenau, Brazil
- Centro de Pesquisas Biológicas de Indaial, Indaial, Brazil
- Programa de conservação do Bugio Ruivo, Perini Business Park, Joinville, Brazil
| | - Lilian Rose Pratt-Riccio
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz (IOC), Fiocruz, Rio de Janeiro, Brazil
| | - Cláudio Tadeu Daniel-Ribeiro
- Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal), Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz (IOC), Fiocruz, Rio de Janeiro, Brazil
| | - Joseli Oliveira Ferreira
- Laboratório de Imunoparasitologia, Instituto Oswaldo Cruz (IOC), Fiocruz, Rio de Janeiro, Brazil
| | - Luzia Helena Carvalho
- Grupo de Pesquisa em Biologia Molecular e Imunologia da malária, Instituto René Rachou/Fiocruz Minas, Belo Horizonte, Brazil
| | | |
Collapse
|
8
|
Markwalter CF, Petersen JEV, Zeno EE, Sumner KM, Freedman E, Mangeni JN, Abel L, Obala AA, Prudhomme-O’Meara W, Taylor SM. Symptomatic malaria enhances protection from reinfection with homologous Plasmodium falciparum parasites. PLoS Pathog 2023; 19:e1011442. [PMID: 37307293 PMCID: PMC10289385 DOI: 10.1371/journal.ppat.1011442] [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: 01/20/2023] [Revised: 06/23/2023] [Accepted: 05/24/2023] [Indexed: 06/14/2023] Open
Abstract
A signature remains elusive of naturally-acquired immunity against Plasmodium falciparum. We identified P. falciparum in a 14-month cohort of 239 people in Kenya, genotyped at immunogenic parasite targets expressed in the pre-erythrocytic (circumsporozoite protein, CSP) and blood (apical membrane antigen 1, AMA-1) stages, and classified into epitope type based on variants in the DV10, Th2R, and Th3R epitopes in CSP and the c1L region of AMA-1. Compared to asymptomatic index infections, symptomatic malaria was associated with reduced reinfection by parasites bearing homologous CSP-Th2R (adjusted hazard ratio [aHR]:0.63; 95% CI:0.45-0.89; p = 0.008) CSP-Th3R (aHR:0.71; 95% CI:0.52-0.97; p = 0.033), and AMA-1 c1L (aHR:0.63; 95% CI:0.43-0.94; p = 0.022) epitope types. The association of symptomatic malaria with reduced hazard of homologous reinfection was strongest for rare epitope types. Symptomatic malaria provides more durable protection against reinfection with parasites bearing homologous epitope types. The phenotype represents a legible molecular epidemiologic signature of naturally-acquired immunity by which to identify new antigen targets.
Collapse
Affiliation(s)
- Christine F. Markwalter
- Duke Global Health Institute, Duke University, Durham, North Carolina, United States of America
| | - Jens E. V. Petersen
- Division of Infectious Diseases, School of Medicine, Duke University, Durham, North Carolina, United States of America
| | - Erica E. Zeno
- Division of Infectious Diseases, School of Medicine, Duke University, Durham, North Carolina, United States of America
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Kelsey M. Sumner
- Division of Infectious Diseases, School of Medicine, Duke University, Durham, North Carolina, United States of America
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Elizabeth Freedman
- Division of Infectious Diseases, School of Medicine, Duke University, Durham, North Carolina, United States of America
| | - Judith N. Mangeni
- School of Public Health, College of Health Sciences, Moi University, Eldoret, Kenya
| | - Lucy Abel
- Academic Model Providing Access to Healthcare, Moi Teaching and Referral Hospital, Eldoret, Kenya
| | - Andrew A. Obala
- School of Medicine, College of Health Sciences, Moi University, Eldoret, Kenya
| | - Wendy Prudhomme-O’Meara
- Duke Global Health Institute, Duke University, Durham, North Carolina, United States of America
- Division of Infectious Diseases, School of Medicine, Duke University, Durham, North Carolina, United States of America
- School of Public Health, College of Health Sciences, Moi University, Eldoret, Kenya
| | - Steve M. Taylor
- Duke Global Health Institute, Duke University, Durham, North Carolina, United States of America
- Division of Infectious Diseases, School of Medicine, Duke University, Durham, North Carolina, United States of America
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
9
|
Ibitokou SA, Gbedande K, Opata MM, Carpio VH, Marshall KM, Stephens R. Effects of Low-Level Persistent Infection on Maintenance of Immunity by CD4 T Cell Subsets and Th1 Cytokines. Infect Immun 2023; 91:e0053122. [PMID: 36920200 PMCID: PMC10016079 DOI: 10.1128/iai.00531-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
Abstract
CD4 T cells are required, along with antibodies, for complete protection from blood-stage infection with Plasmodium spp., which cause malaria. Without continuous exposure, as on emigration of people from endemic areas, protection from malaria decays. As in other persistent infections, low-level Plasmodium chabaudi infection protects the host from reinfection at 2 months postinfection, a phenomenon termed premunition. Premunition is correlated with T cell responses, rather than antibody levels. We previously showed that while both effector T cells (Teff) and memory T cells (Tmem) are present after infection, Teff protect better than Tmem. Here, we studied T cell kinetics post-infection by labeling dividing Ifng+ T cells with 5-bromo-2'-deoxyuridine (BrdU) in infected Ifng reporter mice. Large drops in specific T cell numbers and Ifng+ cells upon clearance of parasites suggest a mechanism for decay of protection. Although protection decays, CD4 Tmem persist, including a highly differentiated CD27- effector memory (Tem) subset that maintains some Ifng expression. In addition, pretreatment of chronically infected animals with neutralizing antibody to interferon gamma (IFN-γ) or with clodronate liposomes before reinfection decreases premunition, supporting a role for Th1-type immunity to reinfection. A pulse-chase experiment comparing chronically infected to treated animals showed that recently divided Ifng+ T cells, particularly IFN-γ+ TNF+ IL-2- T cells, are promoted by persistent infection. These data suggest that low-level persistent infection reduces CD4+ Tmem and multifunctional Teff survival, but promotes IFN-γ+ TNF+ IL-2- T cells and Ifng+ terminally differentiated effector T cells, and prolongs immunity.
Collapse
Affiliation(s)
- Samad A. Ibitokou
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, USA
| | - Komi Gbedande
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, USA
| | - Michael M. Opata
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, USA
| | - Victor H. Carpio
- Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Karis M. Marshall
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, USA
| | - Robin Stephens
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, USA
- Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
10
|
Stadler E, Cromer D, Ogunlade S, Ongoiba A, Doumbo S, Kayentao K, Traore B, Crompton PD, Portugal S, Davenport MP, Khoury DS. Evidence for exposure dependent carriage of malaria parasites across the dry season: modelling analysis of longitudinal data. Malar J 2023; 22:42. [PMID: 36737743 PMCID: PMC9898990 DOI: 10.1186/s12936-023-04461-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 01/17/2023] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND In malaria endemic regions, transmission of Plasmodium falciparum parasites is often seasonal with very low transmission during the dry season and high transmission in the wet season. Parasites survive the dry season within some individuals who experience prolonged carriage of parasites and are thought to 'seed' infection in the next transmission season. METHODS Dry season carriers and their role in the subsequent transmission season are characterized using a combination of mathematical simulations and data analysis of previously described data from a longitudinal study in Mali of individuals aged 3 months-12 years (n = 579). RESULTS Simulating the life-history of individuals experiencing repeated exposure to infection predicts that dry season carriage is more likely in the oldest, most exposed and most immune individuals. This hypothesis is supported by the data from Mali, which shows that carriers are significantly older, experience a higher biting rate at the beginning of the transmission season and develop clinical malaria later than non-carriers. Further, since the most exposed individuals in a community are most likely to be dry season carriers, this is predicted to enable a more than twofold faster spread of parasites into the mosquito population at the start of the subsequent wet season. CONCLUSIONS Carriage of malaria parasites over the months-long dry season in Mali is most likely in the older, more exposed and more immune children. These children may act as super-spreaders facilitating the fast spread of parasites at the beginning of the next transmission season.
Collapse
Affiliation(s)
- Eva Stadler
- grid.1005.40000 0004 4902 0432The Kirby Institute, UNSW Sydney, Sydney, NSW 2052 Australia
| | - Deborah Cromer
- grid.1005.40000 0004 4902 0432The Kirby Institute, UNSW Sydney, Sydney, NSW 2052 Australia
| | - Samson Ogunlade
- grid.1005.40000 0004 4902 0432The Kirby Institute, UNSW Sydney, Sydney, NSW 2052 Australia
| | - Aissata Ongoiba
- grid.461088.30000 0004 0567 336XMalaria Research and Training Centre, Department of Epidemiology of Parasitic Diseases, International Center of Excellence in Research, University of Sciences, Technique, and Technology of Bamako, 91094 Bamako, Mali
| | - Safiatou Doumbo
- grid.461088.30000 0004 0567 336XMalaria Research and Training Centre, Department of Epidemiology of Parasitic Diseases, International Center of Excellence in Research, University of Sciences, Technique, and Technology of Bamako, 91094 Bamako, Mali
| | - Kassoum Kayentao
- grid.461088.30000 0004 0567 336XMalaria Research and Training Centre, Department of Epidemiology of Parasitic Diseases, International Center of Excellence in Research, University of Sciences, Technique, and Technology of Bamako, 91094 Bamako, Mali
| | - Boubacar Traore
- grid.461088.30000 0004 0567 336XMalaria Research and Training Centre, Department of Epidemiology of Parasitic Diseases, International Center of Excellence in Research, University of Sciences, Technique, and Technology of Bamako, 91094 Bamako, Mali
| | - Peter D. Crompton
- grid.419681.30000 0001 2164 9667Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, USA
| | - Silvia Portugal
- grid.419681.30000 0001 2164 9667Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, USA
| | - Miles P. Davenport
- grid.1005.40000 0004 4902 0432The Kirby Institute, UNSW Sydney, Sydney, NSW 2052 Australia
| | - David S. Khoury
- grid.1005.40000 0004 4902 0432The Kirby Institute, UNSW Sydney, Sydney, NSW 2052 Australia
| |
Collapse
|
11
|
Fontana MF, Ollmann Saphire E, Pepper M. Plasmodium infection disrupts the T follicular helper cell response to heterologous immunization. eLife 2023; 12:83330. [PMID: 36715223 PMCID: PMC9886276 DOI: 10.7554/elife.83330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 01/12/2023] [Indexed: 01/31/2023] Open
Abstract
Naturally acquired immunity to malaria develops only after many years and repeated exposures, raising the question of whether Plasmodium parasites, the etiological agents of malaria, suppress the ability of dendritic cells (DCs) to activate optimal T cell responses. We demonstrated recently that B cells, rather than DCs, are the principal activators of CD4+ T cells in murine malaria. In the present study, we further investigated factors that might prevent DCs from priming Plasmodium-specific T helper cell responses. We found that DCs were significantly less efficient at taking up infected red blood cells (iRBCs) compared to soluble antigen, whereas B cells more readily bound iRBCs. To assess whether DCs retained the capacity to present soluble antigen during malaria, we measured responses to a heterologous protein immunization administered to naïve mice or mice infected with P. chabaudi. Antigen uptake, DC activation, and expansion of immunogen-specific T cells were intact in infected mice, indicating DCs remained functional. However, polarization of the immunogen-specific response was dramatically altered, with a near-complete loss of germinal center T follicular helper cells specific for the immunogen, accompanied by significant reductions in antigen-specific B cells and antibody. Our results indicate that DCs remain competent to activate T cells during Plasmodium infection, but that T cell polarization and humoral responses are severely disrupted. This study provides mechanistic insight into the development of both Plasmodium-specific and heterologous adaptive responses in hosts with malaria.
Collapse
Affiliation(s)
- Mary F Fontana
- Department of Immunology, University of Washington School of MedicineSeattleUnited States
| | - Erica Ollmann Saphire
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for ImmunologyLa JollaUnited States
| | - Marion Pepper
- Department of Immunology, University of Washington School of MedicineSeattleUnited States
| |
Collapse
|
12
|
Brown SL, Bauer JJ, Lee J, Ntirandekura E, Stumhofer JS. IgM + and IgM - memory B cells represent heterogeneous populations capable of producing class-switched antibodies and germinal center B cells upon rechallenge with P. yoelii. J Leukoc Biol 2022; 112:1115-1135. [PMID: 35657097 PMCID: PMC9613510 DOI: 10.1002/jlb.4a0921-523r] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 03/29/2022] [Accepted: 04/15/2022] [Indexed: 12/24/2022] Open
Abstract
Memory B cells (MBCs) are essential for maintaining long-term humoral immunity to infectious organisms, including Plasmodium. MBCs are a heterogeneous population whose function can be dictated by isotype or expression of particular surface proteins. Here, aided by antigen-specific B-cell tetramers, MBC populations were evaluated to discern their phenotype and function in response to infection with a nonlethal strain of P. yoelii. Infection of mice with P. yoelii 17X resulted in 2 predominant MBC populations: somatically hypermutated isotype-switched (IgM- ) and IgM+ MBCs that coexpressed CD73 and CD80 that produced antigen-specific antibodies in response to secondary infection. Rechallenge experiments indicated that IgG-producing cells dominated the recall response over the induction of IgM-secreting cells, with both populations expanding with similar timing during the secondary response. Furthermore, using ZsGreen1 expression as a surrogate for activation-induced cytidine deaminase expression alongside CD73 and CD80 coexpression, ZsGreen1+ CD73+ CD80+ IgM+ , and IgM- MBCs gave rise to plasmablasts that secreted Ag-specific Abs after adoptive transfer and infection with P. yoelii. Moreover, ZsGreen1+ CD73+ CD80+ IgM+ and IgM- MBCs could differentiate into B cells with a germinal center phenotype after adoptive transfer. A third population of B cells (ZsGreen1- CD73- CD80- IgM- ) that is apparent after infection responded poorly to reactivation in vitro and in vivo, indicating that these cells do not represent a canonical population of MBCs. Together these data indicated that MBC function is not defined by immunoglobulin isotype, nor does coexpression of key surface markers limit the potential fate of MBCs after recall.
Collapse
Affiliation(s)
- Susie L Brown
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Jonathan J Bauer
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Juhyung Lee
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Enatha Ntirandekura
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Jason S Stumhofer
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
13
|
Abad P, Marín-García P, Heras M, Fobil JN, Hutchful AG, Diez A, Puyet A, Reyes-Palomares A, Azcárate IG, Bautista JM. Microscopic and submicroscopic infection by Plasmodium falciparum: Immunoglobulin M and A profiles as markers of intensity and exposure. Front Cell Infect Microbiol 2022; 12:934321. [PMID: 36118030 PMCID: PMC9478039 DOI: 10.3389/fcimb.2022.934321] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 08/04/2022] [Indexed: 11/15/2022] Open
Abstract
Assessment of serological Plasmodium falciparum–specific antibodies in highly endemic areas provides valuable information about malaria status and parasite exposure in the population. Although serological evidence of Plasmodium exposure is commonly determined by Plasmodium-specific immunoglobulin G (IgG) levels; IgM and IgA are likely markers of malaria status that remain relatively unexplored. Previous studies on IgM and IgA responses have been based on their affinity for single antigens with shortage of immune responses analysis against the whole Plasmodium proteome. Here, we provide evidence of how P. falciparum infection triggers the production of specific IgM and IgA in plasma and its relationship with parasite density and changes in hematological parameters. A total of 201 individuals attending a hospital in Breman Asikuma, Ghana, were recruited into this study. Total and P. falciparum–specific IgM, IgA, and IgG were assessed by ELISA and examined in relation to age (0–5, 14–49, and ≥50 age ranges); infection (submicroscopic vs. microscopic malaria); pregnancy and hematological parameters. Well-known IgG response was used as baseline control. P. falciparum–specific IgM and IgA levels increased in the population with the age, similarly to IgG. These data confirm that acquired humoral immunity develops by repeated infections through the years endorsing IgM and IgA as exposure markers in endemic malaria regions. High levels of specific IgA and IgM in children were associated with microscopic malaria and worse prognosis, because most of them showed severe anemia. This new finding shows that IgM and IgA may be used as diagnostic markers in this age group. We also found an extremely high prevalence of submicroscopic malaria (46.27% on average) accompanied by IgM and IgA levels indistinguishable from those of uninfected individuals. These data, together with the observed lack of sensitivity of rapid diagnostic tests (RDTs) compared to PCR, invoke the urgent need to implement diagnostic markers for submicroscopic malaria. Overall, this study opens the potential use of P. falciparum–specific IgM and IgA as new serological markers to predict malaria status in children and parasite exposure in endemic populations. The difficulties in finding markers of submicroscopic malaria are highlighted, emphasizing the need to explore this field in depth.
Collapse
Affiliation(s)
- Paloma Abad
- Department of Biochemistry and Molecular Biology and Research Institute Hospital 12 de Octubre (Imas12), Universidad Complutense de Madrid, Madrid, Spain
| | | | - Marcos Heras
- Department of Biochemistry and Molecular Biology and Research Institute Hospital 12 de Octubre (Imas12), Universidad Complutense de Madrid, Madrid, Spain
| | - Julius N. Fobil
- Department of Biological, Environmental and Occupational Health Sciences, School of Public Health, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | - Alfred G. Hutchful
- Laboratory of Hematology and Infectious Diseases, Our Lady of Grace Hospital, Breman-Asikuma, Ghana
| | - Amalia Diez
- Department of Biochemistry and Molecular Biology and Research Institute Hospital 12 de Octubre (Imas12), Universidad Complutense de Madrid, Madrid, Spain
| | - Antonio Puyet
- Department of Biochemistry and Molecular Biology and Research Institute Hospital 12 de Octubre (Imas12), Universidad Complutense de Madrid, Madrid, Spain
| | - Armando Reyes-Palomares
- Department of Biochemistry and Molecular Biology and Research Institute Hospital 12 de Octubre (Imas12), Universidad Complutense de Madrid, Madrid, Spain
| | - Isabel G. Azcárate
- Faculty of Health Sciences, Rey Juan Carlos University, Alcorcón, Spain
- *Correspondence: Isabel G. Azcárate, ; José M. Bautista,
| | - José M. Bautista
- Department of Biochemistry and Molecular Biology and Research Institute Hospital 12 de Octubre (Imas12), Universidad Complutense de Madrid, Madrid, Spain
- *Correspondence: Isabel G. Azcárate, ; José M. Bautista,
| |
Collapse
|
14
|
Lugaajju A, Tijani MK, Persson KEM. Flow Cytometry Assay of Plasmodium Falciparum-Specific B-Cell Proportions. Methods Mol Biol 2022; 2470:681-688. [PMID: 35881383 DOI: 10.1007/978-1-0716-2189-9_51] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Detection of P. falciparum-specific subpopulations of B-cells is important for studies of immunity in malaria. This protocol relies on the photostability and protein loading capacity of carboxylated quantum dots to detect a broad range of different P. falciparum-specific B-cells. Infected red blood cell ghosts, obtained by permeabilization of infected cells with Streptolysin O, are coupled with carboxyl quantum dots using N-ethyl-N-dimethylaminopropyl-carbodiimide condensation. Immunophenotyping of P. falciparum-specific B-cells is performed by flow cytometry using Fc-receptor block, quantum dot-infected red blood cell ghost conjugates, and fluorochrome-conjugated anti-human CD19 mouse monoclonal antibodies.
Collapse
Affiliation(s)
- Allan Lugaajju
- School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
- Department of Laboratory Medicine, Lund University, Skåne University Hospital Lund, Lund, Sweden
| | - Muyideen Kolapo Tijani
- Department of Laboratory Medicine, Lund University, Skåne University Hospital Lund, Lund, Sweden
- Cellular Parasitology Programme, Cell Biology and Genetics Unit, Department of Zoology, University of Ibadan, Ibadan, Nigeria
| | | |
Collapse
|
15
|
Live attenuated vaccines, a favorable strategy to provide long-term immunity against protozoan diseases. Trends Parasitol 2021; 38:316-334. [PMID: 34896016 DOI: 10.1016/j.pt.2021.11.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 11/12/2021] [Accepted: 11/12/2021] [Indexed: 12/25/2022]
Abstract
The control of diseases caused by protozoan parasites is one of the United Nations' Sustainable Development Goals. In recent years much research effort has gone into developing a new generation of live attenuated vaccines (LAVs) against malaria, Chagas disease and leishmaniasis. However, there is a bottleneck related to their biosafety, production, and distribution that slows downs further development. The success of irradiated or genetically attenuated sporozoites against malaria, added to the first LAV against leishmaniasis to be evaluated in clinical trials, is indicative that the drawbacks of LAVs are gradually being overcome. However, whether persistence of LAVs is a prerequisite for sustained long-term immunity remains to be clarified, and the procedures necessary for clinical evaluation of vaccine candidates need to be standardized.
Collapse
|
16
|
Daniyan MO. Heat Shock Proteins as Targets for Novel Antimalarial Drug Discovery. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1340:205-236. [PMID: 34569027 DOI: 10.1007/978-3-030-78397-6_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Plasmodium falciparum, the parasitic agent that is responsible for a severe and dangerous form of human malaria, has a history of long years of cohabitation with human beings with attendant negative consequences. While there have been some gains in the fight against malaria through the application of various control measures and the use of chemotherapeutic agents, and despite the global decline in malaria cases and associated deaths, the continual search for new and effective therapeutic agents is key to achieving sustainable development goals. An important parasite survival strategy, which is also of serious concern to the scientific community, is the rate at which the parasites continually develop resistance to drugs. Among the key players in the parasite's ability to develop resistance, maintain cellular integrity, and survives within an unusual environment of the red blood cells are the molecular chaperones of the heat shock proteins (HSP) family. HSPs constitute a novel avenue for antimalarial drug discovery and by exploring their ubiquitous nature and multifunctional activities, they may be suitable targets for the discovery of multi-targets antimalarial drugs, needed to fight incessant drug resistance. In this chapter, features of selected families of plasmodial HSPs that can be exploited in drug discovery are presented. Also, known applications of HSPs in small molecule screening, their potential usefulness in high throughput drug screening, as well as possible challenges are highlighted.
Collapse
Affiliation(s)
- Michael Oluwatoyin Daniyan
- Department of Pharmacology, Faculty of Pharmacy, Obafemi Awolowo University, Ile-Ife, Osun State, Nigeria.
| |
Collapse
|
17
|
Reddy SB, Nagy N, Rönnberg C, Chiodi F, Lugaajju A, Heuts F, Szekely L, Wahlgren M, Persson KEM. Direct contact between Plasmodium falciparum and human B-cells in a novel co-culture increases parasite growth and affects B-cell growth. Malar J 2021; 20:303. [PMID: 34225761 PMCID: PMC8256226 DOI: 10.1186/s12936-021-03831-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 06/24/2021] [Indexed: 11/17/2022] Open
Abstract
Background Plasmodium falciparum parasites cause malaria and co-exist in humans together with B-cells for long periods of time. Immunity is only achieved after repeated exposure. There has been a lack of methods to mimic the in vivo co-occurrence, where cells and parasites can be grown together for many days, and it has been difficult with long time in vitro studies. Methods and results A new method for growing P. falciparum in 5% CO2 with a specially formulated culture medium is described. This knowledge was used to establish the co-culture of live P. falciparum together with human B-cells in vitro for 10 days. The presence of B-cells clearly enhanced parasite growth, but less so when Transwell inserts were used (not allowing passage of cells or merozoites), showing that direct contact is advantageous. B-cells also proliferated more in presence of parasites. Symbiotic parasitic growth was verified using CESS cell-line and it showed similar results, indicating that B-cells are indeed the cells responsible for the effect. In malaria endemic areas, people often have increased levels of atypical memory B-cells in the blood, and in this assay it was demonstrated that when parasites were present there was an increase in the proportion of CD19 + CD20 + CD27 − FCRL4 + B-cells, and a contraction of classical memory B-cells. This effect was most clearly seen when direct contact between B-cells and parasites was allowed. Conclusions These results demonstrate that P. falciparum and B-cells undoubtedly can affect each other when allowed to multiply together, which is valuable information for future vaccine studies.
Collapse
Affiliation(s)
| | - Noemi Nagy
- Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Caroline Rönnberg
- Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.,Department of Clinical Microbiology, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Francesca Chiodi
- Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Allan Lugaajju
- Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.,Makerere University, Kampala, Uganda
| | - Frank Heuts
- Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Laszlo Szekely
- Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Mats Wahlgren
- Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Kristina E M Persson
- Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden. .,Department of Laboratory Medicine, Skåne University Hospital, Lund University, Lund, Sweden.
| |
Collapse
|
18
|
Mejia P, Treviño-Villarreal JH, De Niz M, Meibalan E, Longchamp A, Reynolds JS, Turnbull LB, Opoka RO, Roussilhon C, Spielmann T, Ozaki CK, Heussler VT, Seydel KB, Taylor TE, John CC, Milner DA, Marti M, Mitchell JR. Adipose tissue parasite sequestration drives leptin production in mice and correlates with human cerebral malaria. SCIENCE ADVANCES 2021; 7:7/13/eabe2484. [PMID: 33762334 PMCID: PMC7990332 DOI: 10.1126/sciadv.abe2484] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 02/04/2021] [Indexed: 05/03/2023]
Abstract
Circulating levels of the adipokine leptin are linked to neuropathology in experimental cerebral malaria (ECM), but its source and regulation mechanism remain unknown. Here, we show that sequestration of infected red blood cells (iRBCs) in white adipose tissue (WAT) microvasculature increased local vascular permeability and leptin production. Mice infected with parasite strains that fail to sequester in WAT displayed reduced leptin production and protection from ECM. WAT sequestration and leptin induction were lost in CD36KO mice; however, ECM susceptibility revealed sexual dimorphism. Adipocyte leptin was regulated by the mechanistic target of rapamycin complex 1 (mTORC1) and blocked by rapamycin. In humans, although Plasmodium falciparum infection did not increase circulating leptin levels, iRBC sequestration, tissue leptin production, and mTORC1 activity were positively correlated with CM in pediatric postmortem WAT. These data identify WAT sequestration as a trigger for leptin production with potential implications for pathogenesis of malaria infection, prognosis, and treatment.
Collapse
Affiliation(s)
- Pedro Mejia
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA.
| | | | - Mariana De Niz
- Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
- Institute of Cell Biology, University of Bern, Bern, Switzerland
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Elamaran Meibalan
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Alban Longchamp
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Surgery and the Heart and Vascular Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Justin S Reynolds
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Lindsey B Turnbull
- Department of Pediatric Infectious Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Robert O Opoka
- Department of Pediatrics and Child Health, Makerere University, Kampala, Uganda
| | | | - Tobias Spielmann
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - C Keith Ozaki
- Department of Surgery and the Heart and Vascular Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Karl B Seydel
- Department of Osteopathic Medical Specialties, Michigan State University, East Lansing, MI, USA
| | - Terrie E Taylor
- Department of Osteopathic Medical Specialties, Michigan State University, East Lansing, MI, USA
| | - Chandy C John
- Department of Pediatric Infectious Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Danny A Milner
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- American Society for Clinical Pathology, Chicago, IL, USA
| | - Matthias Marti
- Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - James R Mitchell
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
19
|
Antibody Dynamics for Plasmodium vivax Malaria: A Mathematical Model. Bull Math Biol 2021; 83:6. [PMID: 33387082 DOI: 10.1007/s11538-020-00837-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 11/13/2020] [Indexed: 10/22/2022]
Abstract
Malaria is a mosquito-borne disease that, despite intensive control and mitigation initiatives, continues to pose an enormous public health burden. Plasmodium vivax is one of the principal causes of malaria in humans. Antibodies, which play a fundamental role in the host response to P. vivax, are acquired through exposure to the parasite. Here, we introduce a stochastic, within-host model of antibody responses to P. vivax for an individual in a general transmission setting. We begin by developing an epidemiological framework accounting for P. vivax infections resulting from new mosquito bites (primary infections), as well as the activation of dormant-liver stages known as hypnozoites (relapses). By constructing an infinite server queue, we obtain analytic results for the distribution of relapses in a general transmission setting. We then consider a simple model of antibody kinetics, whereby antibodies are boosted with each infection, but are subject to decay over time. By embedding this model for antibody kinetics in the epidemiological framework using a generalised shot noise process, we derive analytic expressions governing the distribution of antibody levels for a single individual in a general transmission setting. Our work provides a means to explore exposure-dependent antibody dynamics for P. vivax, with the potential to address key questions in the context of serological surveillance and acquired immunity.
Collapse
|
20
|
Costa EMF, Amador ECC, Silva ES, Alvarenga CO, Pereira PE, Póvoa MM, Cunha MG. Malaria transmission and individual variability of the naturally acquired IgG antibody against the Plasmodium vivax blood-stage antigen in an endemic area in Brazil. Acta Trop 2020; 209:105537. [PMID: 32454033 DOI: 10.1016/j.actatropica.2020.105537] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/11/2020] [Accepted: 05/11/2020] [Indexed: 11/20/2022]
Abstract
Plasmodium vivax remains an important cause of malaria in South America and Asia, and analyses of the antibody immune response are being used to identify biomarker of parasite exposure. The IgG antibody naturally acquired predominantly occurs against targets on blood-stage parasites, including C-terminal of the merozoite surface protein 1 (MSP1-19). Epidemiological and immunological evidence has been showed that antibodies to malaria parasite antigens are lost in the absence of ongoing exposure. We describe the IgG antibody response in individuals living in an unstable malaria transmission area in Pará state, Amazon region, Brazil, where an epidemic of P. vivax malaria was recorded and monitored over time. As indicated by epidemiological data, the number of P. vivax-caused malaria cases decreased by approximately 90% after three years and the prevalence of IgG positive to PvMSP1-19 decreased significantly over time, in 2010 (93.4%), 2012 (78.3%), and 2013 (85.1%). Acquisition and decay of the IgG antibody against P. vivax MSP1-19 showed variability among individuals living in areas with recent circulating parasites, where the malaria epidemic was being monitored until transmission had been completely controlled. We also found that previous malaria episodes were associated with an increased in the IgG positivity . Our results showed epidemiological, spatial, temporal and individual variability. The understanding on dynamics of antibodies may have implications for the design of serosurveillance tools for monitoring parasite circulation, especially in a context with spatial and temporal changes in P. vivax malaria transmission.
Collapse
Affiliation(s)
- Edna Maria F Costa
- Universidade Federal do Pará, Instituto de Ciências Biológicas, CEP: 66075-110, Belém, Pará, Brazil
| | | | - Eliane S Silva
- Fundação Centro de Hemoterapia e Hematologia do Pará, CEP: 660033-000, Belém, Pará, Brazil
| | - Cassiana O Alvarenga
- Universidade Federal do Pará, Instituto de Ciências Biológicas, CEP: 66075-110, Belém, Pará, Brazil
| | - Pedro Elias Pereira
- Fundação Centro de Hemoterapia e Hematologia do Pará, CEP: 660033-000, Belém, Pará, Brazil
| | - Marinete M Póvoa
- Instituto Evandro Chagas, CEP: 66087-082, Ananindeua, Pará, Brazil
| | - Maristela G Cunha
- Universidade Federal do Pará, Instituto de Ciências Biológicas, CEP: 66075-110, Belém, Pará, Brazil.
| |
Collapse
|
21
|
Alonso D, Dobson A, Pascual M. Critical transitions in malaria transmission models are consistently generated by superinfection. Philos Trans R Soc Lond B Biol Sci 2020; 374:20180275. [PMID: 31056048 DOI: 10.1098/rstb.2018.0275] [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] [Indexed: 01/17/2023] Open
Abstract
The history of modelling vector-borne infections essentially begins with the papers by Ross on malaria. His models assume that the dynamics of malaria can most simply be characterized by two equations that describe the prevalence of malaria in the human and mosquito hosts. This structure has formed the central core of models for malaria and most other vector-borne diseases for the past century, with additions acknowledging important aetiological details. We partially add to this tradition by describing a malaria model that provides for vital dynamics in the vector and the possibility of super-infection in the human host: reinfection of asymptomatic hosts before they have cleared a prior infection. These key features of malaria aetiology create the potential for break points in the prevalence of infected hosts, sudden transitions that seem to characterize malaria's response to control in different locations. We show that this potential for critical transitions is a general and underappreciated feature of any model for vector-borne diseases with incomplete immunity, including the canonical Ross-McDonald model. Ignoring these details of the host's immune response to infection can potentially lead to serious misunderstanding in the interpretation of malaria distribution patterns and the design of control schemes for other vector-borne diseases. This article is part of the theme issue 'Modelling infectious disease outbreaks in humans, animals and plants: approaches and important themes'. This issue is linked with the subsequent theme issue 'Modelling infectious disease outbreaks in humans, animals and plants: epidemic forecasting and control'.
Collapse
Affiliation(s)
- David Alonso
- 1 Theoretical and Computational Ecology, Center for Advanced Studies (CEAB-CSIC) , Blanes , Spain
| | - Andy Dobson
- 2 Ecology and Evolutionary Biology, Eno Hall, Princeton University , NJ 08540 , USA.,3 Santa Fe Institute , Hyde Park Road, Santa Fe, NM , USA
| | - Mercedes Pascual
- 3 Santa Fe Institute , Hyde Park Road, Santa Fe, NM , USA.,4 Ecology and Evolutionary Biology, University of Chicago , Chicago, IL , USA
| |
Collapse
|
22
|
Soares RR, Nakaie CR, Rodrigues-da-Silva RN, da Silva RL, Lima-Junior JDC, Scopel KKG. Main B-cell epitopes of PvAMA-1 and PvMSP-9 are targeted by naturally acquired antibodies and epitope-specific memory cells in acute and convalescent phases of vivax malaria. Parasite Immunol 2020; 42:e12705. [PMID: 32096238 DOI: 10.1111/pim.12705] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 01/30/2020] [Accepted: 02/11/2020] [Indexed: 01/11/2023]
Abstract
Although antibodies are considered critical for malaria protection, little is known about the mechanisms/factors that maintain humoral immunity, especially regarding the induction and maintenance of memory B cells over time. In Brazilian endemic areas, this is the first time that the profile of antibody responses and the occurrence of antigen-specific memory B cells (MBC) against P vivax were investigated during acute malaria and up to six months after parasite clearance. For this, we selected two peptides, PvAMA-1(S290-K307) and PvMSP-9(E795-A808) , which represent the apical membrane antigen-1 and merozoite surface protein-9 of P vivax, respectively. Both peptides were previously described as containing linear B-cell epitopes. Our findings were as follows: 1-both peptides were recognized by IgG antibodies at a high frequency (between 24% and 81%) in all study groups; 2-in the absence of infection, the IgG levels remained stable throughout 6 months of follow-up; and 3-PvAMA-1(S290-K307) and PvMSP-9(E795-A808) -specific MBCs were detected in all individual groups in the absence of reinfection throughout the follow-up period, suggesting long-lived MBC. However, no positive association was observed between malaria-specific antibody levels and frequency of MBCs over time. Taken together, these results suggest that peptides can be, in the future, an alternative strategy to polypeptidic vaccine formulation.
Collapse
Affiliation(s)
- Roberta Reis Soares
- Department of Parasitology, Microbiology and Immunology, Institute of Biological Sciences, Federal University of Juiz de For a, Juiz de For a, Brazil
| | - Clovis Ryuichi Nakaie
- Departament of Biophysics, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | | | - Rogério Lauria da Silva
- Departament of Biophysics, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | | | - Kézia Katiani Gorza Scopel
- Department of Parasitology, Microbiology and Immunology, Institute of Biological Sciences, Federal University of Juiz de For a, Juiz de For a, Brazil
| |
Collapse
|
23
|
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: 48] [Impact Index Per Article: 9.6] [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
|
24
|
Pan Y, Sun X, Li D, Zhao Y, Jin F, Cao Y. PD-1 blockade promotes immune memory following Plasmodium berghei ANKA reinfection. Int Immunopharmacol 2020; 80:106186. [PMID: 31931371 DOI: 10.1016/j.intimp.2020.106186] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 12/27/2019] [Accepted: 01/01/2020] [Indexed: 01/22/2023]
Abstract
The establishment of malaria immune memory is slow, incomplete, and short-lived. The mechanisms underpinning the generation and maintenance of anti-malarial immune memory remain unclear. This study evaluated the possible role of programmed cell death-1 (PD-1) in the establishment of malaria immune memory. Following infection by Plasmodium berghei ANKA (Pb ANKA) we compared natural immunity, acquired immunity, and immune memory between WT and mice lacking PD-1 via monoclonal antibody treatment. We found that parasitemia levels were significantly lower in the PD-1 knockdown group. After PD-1 elimination, dendritic cells, Th1, and T-follicular helper cells increased significantly. In addition, memory T, long-lived plasma cells, and serum antibody production also increased significantly. Therefore, PD-1 elimination induced stronger natural and acquired immune responses and enhanced immune memory against the parasite.
Collapse
Affiliation(s)
- Yanyan Pan
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang 110013, China; Department of Central Laboratory, Dalian Municipal Central Hospital, Dalian 116033, China
| | - Xiaodan Sun
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang 110013, China
| | - Danni Li
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang 110013, China
| | - Yan Zhao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang 110013, China
| | - Feng Jin
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang 110001,China
| | - Yaming Cao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang 110013, China.
| |
Collapse
|
25
|
Jensen AR, Adams Y, Hviid L. Cerebral Plasmodium falciparum malaria: The role of PfEMP1 in its pathogenesis and immunity, and PfEMP1-based vaccines to prevent it. Immunol Rev 2020; 293:230-252. [PMID: 31562653 PMCID: PMC6972667 DOI: 10.1111/imr.12807] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 09/04/2019] [Accepted: 09/09/2019] [Indexed: 12/13/2022]
Abstract
Malaria, a mosquito-borne infectious disease caused by parasites of the genus Plasmodium continues to be a major health problem worldwide. The unicellular Plasmodium-parasites have the unique capacity to infect and replicate within host erythrocytes. By expressing variant surface antigens Plasmodium falciparum has evolved to avoid protective immune responses; as a result in endemic areas anti-malaria immunity develops gradually over many years of multiple and repeated infections. We are studying the role of Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) expressed by asexual stages of P. falciparum responsible for the pathogenicity of severe malaria. The immunopathology of falciparum malaria has been linked to cyto-adhesion of infected erythrocytes to specific host receptors. A greater appreciation of the PfEMP1 molecules important for the development of protective immunity and immunopathology is a prerequisite for the rational discovery and development of a safe and protective anti-disease malaria vaccine. Here we review the role of ICAM-1 and EPCR receptor adhering falciparum-parasites in the development of severe malaria; we discuss our current research to understand the factors involved in the pathogenesis of cerebral malaria and the feasibility of developing a vaccine targeted specifically to prevent this disease.
Collapse
Affiliation(s)
- Anja Ramstedt Jensen
- Centre for Medical Parasitology at Department of Immunology and MicrobiologyFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Yvonne Adams
- Centre for Medical Parasitology at Department of Immunology and MicrobiologyFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Lars Hviid
- Centre for Medical Parasitology at Department of Immunology and MicrobiologyFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Department of Infectious DiseasesRigshospitaletCopenhagenDenmark
| |
Collapse
|
26
|
Olsen RW, Ecklu-Mensah G, Bengtsson A, Ofori MF, Kusi KA, Koram KA, Hviid L, Adams Y, Jensen ATR. Acquisition of IgG to ICAM-1-Binding DBLβ Domains in the Plasmodium falciparum Erythrocyte Membrane Protein 1 Antigen Family Varies between Groups A, B, and C. Infect Immun 2019; 87:e00224-19. [PMID: 31308082 PMCID: PMC6759304 DOI: 10.1128/iai.00224-19] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 07/03/2019] [Indexed: 12/31/2022] Open
Abstract
Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) is an important malaria virulence factor. The protein family can be divided into clinically relevant subfamilies. ICAM-1-binding group A PfEMP1 proteins also bind endothelial protein C receptor and have been associated with cerebral malaria in children. IgG to these PfEMP1 proteins is acquired later in life than that to group A PfEMP1 not binding ICAM-1. The kinetics of acquisition of IgG to group B and C PfEMP1 proteins binding ICAM-1 is unclear and was studied here. Gene sequences encoding group B and C PfEMP1 with DBLβ domains known to bind ICAM-1 were used to identify additional binders. Levels of IgG specific for DBLβ domains from group A, B, and C PfEMP1 binding or not binding ICAM-1 were measured in plasma from Ghanaian children with or without malaria. Seven new ICAM-1-binding DBLβ domains from group B and C PfEMP1 were identified. Healthy children had higher levels of IgG specific for ICAM-1-binding DBLβ domains from group A than from groups B and C. However, the opposite pattern was found in children with malaria, particularly among young patients. Acquisition of IgG specific for DBLβ domains binding ICAM-1 differs between PfEMP1 groups.
Collapse
MESH Headings
- Antibodies, Protozoan/biosynthesis
- Child
- Child, Preschool
- Erythrocytes/immunology
- Erythrocytes/parasitology
- Female
- Gene Expression
- Ghana
- Humans
- Immunoglobulin G/biosynthesis
- Infant
- Intercellular Adhesion Molecule-1/genetics
- Intercellular Adhesion Molecule-1/immunology
- Malaria, Cerebral/genetics
- Malaria, Cerebral/immunology
- Malaria, Cerebral/parasitology
- Malaria, Cerebral/pathology
- Malaria, Falciparum/genetics
- Malaria, Falciparum/immunology
- Malaria, Falciparum/parasitology
- Malaria, Falciparum/pathology
- Male
- Plasmodium falciparum/immunology
- Plasmodium falciparum/pathogenicity
- Polymorphism, Genetic
- Protein Binding
- Protein Domains
- Protozoan Proteins/classification
- Protozoan Proteins/genetics
- Protozoan Proteins/immunology
- Seasons
- Severity of Illness Index
Collapse
Affiliation(s)
- Rebecca W Olsen
- Centre for Medical Parasitology at Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gertrude Ecklu-Mensah
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Anja Bengtsson
- Centre for Medical Parasitology at Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael F Ofori
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Kwadwo A Kusi
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Kwadwo A Koram
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Lars Hviid
- Centre for Medical Parasitology at Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Yvonne Adams
- Centre for Medical Parasitology at Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anja T R Jensen
- Centre for Medical Parasitology at Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
27
|
Soares RR, Cunha CF, Ferraz‐Nogueira R, Marins‐dos‐Santos A, Rodrigues‐da‐Silva RN, Soares I, Lima‐Junior J, Bertho AL, Ferreira MU, Scopel KKG. Apical membrane protein 1‐specific antibody profile and temporal changes in peripheral blood B‐cell populations in
Plasmodium vivax
malaria. Parasite Immunol 2019; 41:e12662. [DOI: 10.1111/pim.12662] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 06/07/2019] [Accepted: 07/01/2019] [Indexed: 02/03/2023]
Affiliation(s)
- Roberta Reis Soares
- Department of Parasitology, Microbiology and Immunology Institute of Biological Sciences, Federal University of Juiz de Fora Juiz de Fora Brazil
| | - Clarissa F. Cunha
- Laboratory of Immunoparasitology Oswaldo Cruz Institute, FIOCRUZ Rio de Janeiro Brazil
| | - Raquel Ferraz‐Nogueira
- Laboratory of Immunoparasitology Oswaldo Cruz Institute, FIOCRUZ Rio de Janeiro Brazil
- Flow Cytometry Cell Sorting Core Facility Oswaldo Cruz Institute, FIOCRUZ Rio de Janeiro Brazil
| | | | | | - Irene Soares
- School of Pharmaceutical Sciences, Department of Clinical Analyses and Toxicology University of São Paulo Sao Paulo Brazil
| | - Josué Lima‐Junior
- Laboratory of Immunoparasitology Oswaldo Cruz Institute, FIOCRUZ Rio de Janeiro Brazil
| | - Alvaro Luiz Bertho
- Laboratory of Immunoparasitology Oswaldo Cruz Institute, FIOCRUZ Rio de Janeiro Brazil
- Flow Cytometry Cell Sorting Core Facility Oswaldo Cruz Institute, FIOCRUZ Rio de Janeiro Brazil
| | - Marcelo Urbano Ferreira
- Department of Parasitology, Institute of Biomedical Sciences University of São Paulo Sao Paulo Brazil
| | - Kézia Katiani Gorza Scopel
- Department of Parasitology, Microbiology and Immunology Institute of Biological Sciences, Federal University of Juiz de Fora Juiz de Fora Brazil
| |
Collapse
|
28
|
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: 3.8] [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
|
29
|
Shaw TN, Inkson CA, Villegas-Mendez A, Pattinson DJ, Strangward P, Else KJ, Draper SJ, Zeef LAH, Couper KN. Infection-Induced Resistance to Experimental Cerebral Malaria Is Dependent Upon Secreted Antibody-Mediated Inhibition of Pathogenic CD8 + T Cell Responses. Front Immunol 2019; 10:248. [PMID: 30846985 PMCID: PMC6394254 DOI: 10.3389/fimmu.2019.00248] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 01/29/2019] [Indexed: 12/27/2022] Open
Abstract
Cerebral malaria (CM) is one of the most severe complications of Plasmodium falciparum infection. There is evidence that repeated parasite exposure promotes resistance against CM. However, the immunological basis of this infection-induced resistance remains poorly understood. Here, utilizing the Plasmodium berghei ANKA (PbA) model of experimental cerebral malaria (ECM), we show that three rounds of infection and drug-cure protects against the development of ECM during a subsequent fourth (4X) infection. Exposure-induced resistance was associated with specific suppression of CD8+ T cell activation and CTL-related pathways, which corresponded with the development of heterogeneous atypical B cell populations as well as the gradual infection-induced generation and maintenance of high levels of anti-parasite IgG. Mechanistically, transfer of high-titer anti-parasite IgG did not protect 1X infected mice against ECM and depletion of atypical and regulatory B cells during 4X infection failed to abrogate infection-induced resistance to ECM. However, IgMi mice that were unable to produce secreted antibody, or undergo class switching, during the repeated rounds of infection failed to develop resistance against ECM. The failure of infection-induced protection in IgMi mice was associated with impaired development of atypical B cell populations and the inability to suppress pathogenic CD8+ T cell responses. Our results, therefore, suggest the importance of anti-parasite antibody responses, gradually acquired, and maintained through repeated Plasmodium infections, for modulating the B cell compartment and eventually suppressing memory CD8+ T cell reactivation to establish infection-induced resistance to ECM.
Collapse
Affiliation(s)
- Tovah N. Shaw
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Colette A. Inkson
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Ana Villegas-Mendez
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | | | - Patrick Strangward
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Kathryn J. Else
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Simon J. Draper
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Leo A. H. Zeef
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Kevin N. Couper
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
30
|
Nakamae S, Kimura D, Miyakoda M, Sukhbaatar O, Inoue SI, Yui K. Role of IL-10 in inhibiting protective immune responses against infection with heterologous Plasmodium parasites. Parasitol Int 2019; 70:5-15. [PMID: 30639137 DOI: 10.1016/j.parint.2019.01.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 01/04/2019] [Accepted: 01/07/2019] [Indexed: 11/25/2022]
Abstract
Malaria is induced by infection with Plasmodium parasites, which are genetically diverse, and the immune response to Plasmodium infection has both allele-specific and cross-reactive components. To determine the role of the cross-reactive immune response in the protection and disease manifestation in heterologous Plasmodium infection, we used infection models of P. chabaudi chabaudi (Pcc) and P. berghei ANKA (PbA). CD4+ T cells primed with Pcc infection exhibited strong cross-reactivity to PbA antigens. We infected C57BL/6 mice with Pcc and subsequently treated them with an anti-Plasmodium drug. The Pcc-primed mice exhibited reduced parasitemia and showed no signs of experimental cerebral malaria after infection with PbA. CD4+ T cells from the Pcc-primed mice produced high levels of IFN-γ and IL-10 in response to PbA early after PbA infection. The blockade of IL-10 signaling with anti-IL-10 receptor antibody increased the proportion of activated CD4+ and γδ T cells and the IFN-γ production by CD4+ T cells in response to PbA antigens, while markedly reducing the levels of parasitemia. In contrast, IL-10 blockade did not have a significant effect on parasitemia levels in unprimed mice after PbA infection. These data suggest a potent regulatory role of IL-10 in the cross-reactive memory response to the infection with heterologous Plasmodium parasites leading to the inhibition of the protective immunity and pathogenesis.
Collapse
Affiliation(s)
- Sayuri Nakamae
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Program for Nurturing Global Leaders in Tropical and Emerging Infectious Diseases, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Daisuke Kimura
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Department of Immunology, School of Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Department of Health, Sports, and Nutrition, Faculty of Health and Welfare, Kobe Women's University, 4-7-2 Minatojima-nakamachi, Chuo-ku, Kobe 650-0046, Japan
| | - Mana Miyakoda
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Department of Immunology, School of Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Research and Education Center for Drug Fostering and Evolution, School of Pharmaceutical Sciences, Nagasaki University, 1-14 Bunkyomachi, Nagasaki 852-8521, Japan
| | - Odsuren Sukhbaatar
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Program for Nurturing Global Leaders in Tropical and Emerging Infectious Diseases, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Shin-Ichi Inoue
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Department of Immunology, School of Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Katsuyuki Yui
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Program for Nurturing Global Leaders in Tropical and Emerging Infectious Diseases, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Department of Immunology, School of Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Graduate School of Tropical Medicine and Global Health, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan.
| |
Collapse
|
31
|
Gowda DC, Wu X. Parasite Recognition and Signaling Mechanisms in Innate Immune Responses to Malaria. Front Immunol 2018; 9:3006. [PMID: 30619355 PMCID: PMC6305727 DOI: 10.3389/fimmu.2018.03006] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 12/05/2018] [Indexed: 12/20/2022] Open
Abstract
Malaria caused by the Plasmodium family of parasites, especially P.falciparum and P. vivax, is a major health problem in many countries in the tropical and subtropical regions of the world. The disease presents a wide array of systemic clinical conditions and several life-threatening organ pathologies, including the dreaded cerebral malaria. Like many other infectious diseases, malaria is an inflammatory response-driven disease, and positive outcomes to infection depend on finely tuned regulation of immune responses that efficiently clear parasites and allow protective immunity to develop. Immune responses initiated by the innate immune system in response to parasites play key roles both in protective immunity development and pathogenesis. Initial pro-inflammatory responses are essential for clearing infection by promoting appropriate cell-mediated and humoral immunity. However, elevated and prolonged pro-inflammatory responses owing to inappropriate cellular programming contribute to disease conditions. A comprehensive knowledge of the molecular and cellular mechanisms that initiate immune responses and how these responses contribute to protective immunity development or pathogenesis is important for developing effective therapeutics and/or a vaccine. Historically, in efforts to develop a vaccine, immunity to malaria was extensively studied in the context of identifying protective humoral responses, targeting proteins involved in parasite invasion or clearance. The innate immune response was thought to be non-specific. However, during the past two decades, there has been a significant progress in understanding the molecular and cellular mechanisms of host-parasite interactions and the associated signaling in immune responses to malaria. Malaria infection occurs at two stages, initially in the liver through the bite of a mosquito, carrying sporozoites, and subsequently, in the blood through the invasion of red blood cells by merozoites released from the infected hepatocytes. Soon after infection, both the liver and blood stage parasites are sensed by various receptors of the host innate immune system resulting in the activation of signaling pathways and production of cytokines and chemokines. These immune responses play crucial roles in clearing parasites and regulating adaptive immunity. Here, we summarize the knowledge on molecular mechanisms that underlie the innate immune responses to malaria infection.
Collapse
Affiliation(s)
- D Channe Gowda
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Xianzhu Wu
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
32
|
Miyakoda M, Bayarsaikhan G, Kimura D, Akbari M, Udono H, Yui K. Metformin Promotes the Protection of Mice Infected With Plasmodium yoelii Independently of γδ T Cell Expansion. Front Immunol 2018; 9:2942. [PMID: 30619302 PMCID: PMC6300485 DOI: 10.3389/fimmu.2018.02942] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 11/30/2018] [Indexed: 01/05/2023] Open
Abstract
Adaptive immune responses are critical for protection against infection with Plasmodium parasites. The metabolic state dramatically changes in T cells during activation and the memory phase. Recent findings suggest that metformin, a medication for treating type-II diabetes, enhances T-cell immune responses by modulating lymphocyte metabolism. In this study, we investigated whether metformin could enhance anti-malaria immunity. Mice were infected with Plasmodium yoelii and administered metformin. Levels of parasitemia were reduced in treated mice compared with those in untreated mice, starting at ~2 weeks post-infection. The number of γδ T cells dramatically increased in the spleens of treated mice compared with that in untreated mice during the later phase of infection, while that of αβ T cells did not. The proportions of Vγ1+ and Vγ2+ γδ T cells increased, suggesting that activated cells were selectively expanded. However, these γδ T cells expressed inhibitory receptors and had severe defects in cytokine production, suggesting that they were in a state of exhaustion. Metformin was unable to rescue the cells from exhaustion at this stage. Depletion of γδ T cells with antibody treatment did not affect the reduction of parasitemia in metformin-treated mice, suggesting that the effect of metformin on the reduction of parasitemia was independent of γδ T cells.
Collapse
Affiliation(s)
- Mana Miyakoda
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan.,Research and Education Center for Drug Fostering and Evolution, School of Pharmaceutical Sciences, Nagasaki University, Nagasaki, Japan
| | - Ganchimeg Bayarsaikhan
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Daisuke Kimura
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan.,Department of Health, Sports, and Nutrition, Faculty of Health and Welfare, Kobe Women's University, Kobe, Japan
| | - Masoud Akbari
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Heiichiro Udono
- Department of Immunology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Katsuyuki Yui
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan.,Graduate School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
33
|
Howard J, Zaidi I, Loizon S, Mercereau-Puijalon O, Déchanet-Merville J, Mamani-Matsuda M. Human Vγ9Vδ2 T Lymphocytes in the Immune Response to P. falciparum Infection. Front Immunol 2018; 9:2760. [PMID: 30538708 PMCID: PMC6277687 DOI: 10.3389/fimmu.2018.02760] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 11/09/2018] [Indexed: 01/02/2023] Open
Abstract
Malaria is an infectious disease caused by the protozoan parasite Plasmodium sp, the most lethal being Plasmodium falciparum. Clinical malaria is associated with the asexual replication cycle of Plasmodium parasites inside the red blood cells (RBCs) and a dysregulated immune response. Although the mechanisms of immune responses to blood—or liver-stage parasites have been extensively studied, this has not led to satisfactory leads for vaccine design. Among innate immune cells responding to infection are the non-conventional gamma-delta T-cells. The Vγ9Vδ2 T-cell subset, found only in primates, is activated in response to non-peptidic phosphoantigens produced by stressed mammalian cells or by microorganisms such as Mycobacteria, E.coli, and Plasmodium. The potential protective role of Vγ9Vδ2 T-cells against infections and cancer progression is of current research interest. Vγ9Vδ2 T-cells have been shown to play a role in the early control of P. falciparum parasitemia and to influence malaria adaptive immunity via cytokine release and antigen presentation. They are activated and expanded during a primary P. falciparum infection in response to malaria phosphoantigens and their activity is modulated upon subsequent infections. Here, we review the wide range of functions by which Vγ9Vδ2 T-cells could both contribute to and protect from malaria pathology, with a particular focus on their ability to induce both innate and adaptive responses. We discuss how the multifunctional roles of these T-cells could open new perspectives on gamma-delta T-cell-based interventions to prevent or cure malaria.
Collapse
Affiliation(s)
- Jennifer Howard
- Division of Intramural Research (DIR), National Institutes of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - Irfan Zaidi
- Division of Intramural Research (DIR), National Institutes of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - Séverine Loizon
- Univ. Bordeaux, CNRS ImmunoConcEpT UMR 5164, Bordeaux, France
| | | | | | | |
Collapse
|
34
|
Summerauer AM, Colombo L, Ogwang R, Berger C, Fehr J, Bürgler S. High purity high yield tandem B and T helper cell isolation for qRT-PCR analysis suitable for basically equipped laboratories. Malar J 2018; 17:395. [PMID: 30373593 PMCID: PMC6206625 DOI: 10.1186/s12936-018-2547-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 10/24/2018] [Indexed: 11/17/2022] Open
Abstract
Background Malaria is still a major health problem in sub-Saharan Africa and south-east Asia, but research on malaria in low-income countries can be a challenge due to the lack of laboratory equipment. In addition, severe malaria mainly affects very young children, which limits the amount of blood available for research purposes. Thus, there is a need for protocols that yield a maximum of information from a minimum amount of blood, which are operable in basically equipped laboratories. Results A protocol for tandem B and T helper (Th) cell isolation directly from whole blood, and a freezer-independent sample preservation method compatible with the warm and humid climate of malaria regions was established and validated. The protocol thereby circumvents the need of high-technology centrifuges and unimpeachable power supply for peripheral blood mononuclear cell isolation. Both purity and yield are excellent. Depending on the expression level of the genes of interest, between 2 and 5 ml of blood are adequate for reliable qRT-PCR results from both B and Th cells of healthy paediatric donors as well as paediatric malaria patients. Conclusion This protocol for high purity high yield B cell and Th cell isolation and sample storage for subsequent qRT-PCR analysis from a minimal amount of blood is contrivable with basic equipment and independent of continuous power supply. Thus, it is likely to be of avail for many scientists performing malaria research in rural institutes or hospitals, and thus in countries where malaria is most prevalent.
Collapse
Affiliation(s)
- Andrea Maria Summerauer
- Experimental Infectious Diseases and Cancer Research, Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Lorenzo Colombo
- Experimental Infectious Diseases and Cancer Research, Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Rodney Ogwang
- Makerere University, College of Health Sciences, Kampala, Uganda.,Centre of Tropical Neuroscience, Kitgum Site, Uganda
| | - Christoph Berger
- Experimental Infectious Diseases and Cancer Research, Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Jan Fehr
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.,Department of Public Health, Epidemiology Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Simone Bürgler
- Experimental Infectious Diseases and Cancer Research, Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
35
|
Rodriguez-Barraquer I, Arinaitwe E, Jagannathan P, Kamya MR, Rosenthal PJ, Rek J, Dorsey G, Nankabirwa J, Staedke SG, Kilama M, Drakeley C, Ssewanyana I, Smith DL, Greenhouse B. Quantification of anti-parasite and anti-disease immunity to malaria as a function of age and exposure. eLife 2018; 7:35832. [PMID: 30044224 PMCID: PMC6103767 DOI: 10.7554/elife.35832] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Accepted: 07/15/2018] [Indexed: 01/13/2023] Open
Abstract
Fundamental gaps remain in our understanding of how immunity to malaria develops. We used detailed clinical and entomological data from parallel cohort studies conducted across the malaria transmission spectrum in Uganda to quantify the development of immunity against symptomatic P. falciparum as a function of age and transmission intensity. We focus on: anti-parasite immunity (i.e. ability to control parasite densities) and anti-disease immunity (i.e. ability to tolerate higher parasite densities without fever). Our findings suggest a strong effect of age on both types of immunity, not explained by cumulative-exposure. They also show an independent effect of exposure, where children living in moderate/high transmission settings develop immunity faster as transmission increases. Surprisingly, children in the lowest transmission setting appear to develop immunity more efficiently than those living in moderate transmission settings. Anti-parasite and anti-disease immunity develop in parallel, reducing the probability of experiencing symptomatic malaria upon each subsequent P. falciparum infection.
Collapse
Affiliation(s)
- Isabel Rodriguez-Barraquer
- Department of Medicine, San Francisco General Hospital, University of California, San Francisco, San Francisco, United States
| | | | | | - Moses R Kamya
- Department of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
| | - Phillip J Rosenthal
- Department of Medicine, San Francisco General Hospital, University of California, San Francisco, San Francisco, United States
| | - John Rek
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Grant Dorsey
- Department of Medicine, San Francisco General Hospital, University of California, San Francisco, San Francisco, United States
| | | | - Sarah G Staedke
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Maxwell Kilama
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Chris Drakeley
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | | | - David L Smith
- Institute of Health Metrics and Evaluation, University of Washington, Seattle, United States
| | - Bryan Greenhouse
- Department of Medicine, San Francisco General Hospital, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
36
|
Olsen RW, Ecklu-Mensah G, Bengtsson A, Ofori MF, Lusingu JPA, Castberg FC, Hviid L, Adams Y, Jensen ATR. Natural and Vaccine-Induced Acquisition of Cross-Reactive IgG-Inhibiting ICAM-1-Specific Binding of a Plasmodium falciparum PfEMP1 Subtype Associated Specifically with Cerebral Malaria. Infect Immun 2018; 86:e00622-17. [PMID: 29426042 PMCID: PMC5865037 DOI: 10.1128/iai.00622-17] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 01/25/2018] [Indexed: 12/17/2022] Open
Abstract
Cerebral malaria (CM) is a potentially deadly outcome of Plasmodium falciparum malaria that is precipitated by sequestration of infected erythrocytes (IEs) in the brain. The adhesion of IEs to brain endothelial cells is mediated by a subtype of parasite-encoded erythrocyte membrane protein 1 (PfEMP1) that facilitates dual binding to host intercellular adhesion molecule 1 (ICAM-1) and endothelial protein receptor C (EPCR). The PfEMP1 subtype is characterized by the presence of a particular motif (DBLβ_motif) in the constituent ICAM-1-binding DBLβ domain. The rate of natural acquisition of DBLβ_motif-specific IgG antibodies and the ability to induce such antibodies by vaccination are unknown, and the aim of this study was to provide such data. We used an enzyme-linked immunosorbent assay (ELISA) to measure DBLβ-specific IgG in plasma from Ghanaian children with malaria. The ability of human immune plasma and DBLβ-specific rat antisera to inhibit the interaction between ICAM-1 and DBLβ was assessed using ELISA and in vitro assays of IE adhesion under flow. The acquisition of DBLβ_motif-specific IgG coincided with age-specific susceptibility to CM. Broadly cross-reactive antibodies inhibiting the interaction between ICAM-1 and DBLβ_motif domains were detectable in immune plasma and in sera of rats immunized with specific DBLβ_motif antigens. Importantly, antibodies against the DBLβ_motif inhibited ICAM-1-specific in vitro adhesion of erythrocytes infected by four of five P. falciparum isolates from cerebral malaria patients. We conclude that natural exposure to P. falciparum as well as immunization with specific DBLβ_motif antigens can induce cross-reactive antibodies that inhibit the interaction between ICAM-1 and a broad range of DBLβ_motif domains. These findings raise hope that a vaccine designed specifically to prevent CM is feasible.
Collapse
MESH Headings
- Adolescent
- Amino Acid Motifs
- Antibodies, Neutralizing/immunology
- Antibodies, Protozoan/immunology
- Antigens, Protozoan/immunology
- Binding Sites
- Child
- Child, Preschool
- Cross Reactions/immunology
- Ghana
- Humans
- Immunoglobulin G/immunology
- Immunoglobulin G/metabolism
- Infant
- Intercellular Adhesion Molecule-1/metabolism
- Malaria Vaccines/immunology
- Malaria, Cerebral/immunology
- Malaria, Cerebral/metabolism
- Malaria, Cerebral/parasitology
- Malaria, Falciparum/immunology
- Malaria, Falciparum/metabolism
- Malaria, Falciparum/parasitology
- Plasmodium falciparum/immunology
- Protein Binding/immunology
- Protein Interaction Domains and Motifs
- Protozoan Proteins/chemistry
- Protozoan Proteins/immunology
- Protozoan Proteins/metabolism
- Tanzania
Collapse
Affiliation(s)
- Rebecca W Olsen
- Centre for Medical Parasitology at Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gertrude Ecklu-Mensah
- Centre for Medical Parasitology at Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Anja Bengtsson
- Centre for Medical Parasitology at Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael F Ofori
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - John P A Lusingu
- National Institute for Medical Research, Tanga Centre, Tanga City, Tanzania
| | - Filip C Castberg
- Centre for Medical Parasitology at Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Microbiology, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Lars Hviid
- Centre for Medical Parasitology at Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Yvonne Adams
- Centre for Medical Parasitology at Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anja T R Jensen
- Centre for Medical Parasitology at Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
37
|
Martin TCS, Vinetz JM. Asymptomatic Plasmodium vivax parasitaemia in the low-transmission setting: the role for a population-based transmission-blocking vaccine for malaria elimination. Malar J 2018; 17:89. [PMID: 29466991 PMCID: PMC5822557 DOI: 10.1186/s12936-018-2243-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 02/17/2018] [Indexed: 12/21/2022] Open
Abstract
Plasmodium vivax remains an important cause of morbidity and mortality across the Americas, Horn of Africa, East and South East Asia. Control of transmission has been hampered by emergence of chloroquine resistance and several intrinsic characteristics of infection including asymptomatic carriage, challenges with diagnosis, difficulty eradicating the carrier state and early gametocyte appearance. Complex human-parasite-vector immunological interactions may facilitate onward infection of mosquitoes. Given these challenges, new therapies are being explored including the development of transmission to mosquito blocking vaccines. Herein, the case supporting the need for transmission-blocking vaccines to augment control of P. vivax parasite transmission and explore factors that are limiting eradication efforts is discussed.
Collapse
Affiliation(s)
- Thomas C S Martin
- Division of Infectious Diseases, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Joseph M Vinetz
- Division of Infectious Diseases, Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
38
|
Menezes RADO, Gomes MDSM, Mendes AM, Couto ÁARDA, Nacher M, Pimenta TS, de Sousa ACP, Baptista ARDS, de Jesus MI, Enk MJ, Cunha MG, Machado RLD. Enteroparasite and vivax malaria co-infection on the Brazil-French Guiana border: Epidemiological, haematological and immunological aspects. PLoS One 2018; 13:e0189958. [PMID: 29293589 PMCID: PMC5749708 DOI: 10.1371/journal.pone.0189958] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 12/05/2017] [Indexed: 11/30/2022] Open
Abstract
Malaria-enteroparasitic co-infections are known for their endemicity. Although they are prevalent, little is known about their epidemiology and effect on the immune response. This study evaluated the effect of enteroparasite co-infections with malaria caused by Plasmodium vivax in a border area between Brazil and French Guiana. The cross sectional study took place in Oiapoque, a municipality of Amapá, on the Amazon border. Malaria was diagnosed using thick blood smears, haemoglobin dosage by an automated method and coproparasitology by the Hoffman and Faust methods. The anti-PvMSP-119 IgG antibodies in the plasma were evaluated using ELISA and Th1 (IFN-γ, TNF-α and IL-2), and Th2 (IL-4, IL-5 and IL-10) cytokine counts were performed by flow cytometry. The participants were grouped into those that were monoinfected with vivax malaria (M), vivax malaria-enteroparasite co-infected (CI), monoinfected with enteroparasite (E) and endemic controls (EC), who were negative for both diseases. 441 individuals were included and grouped according to their infection status: [M 6.9% (30/441)], [Cl 26.5% (117/441)], [E 32.4% (143/441)] and [EC 34.2% (151/441)]. Males prevailed among the (M) 77% (23/30) and (CI) 60% (70/117) groups. There was a difference in haemoglobin levels among the different groups under study for [EC-E], [EC-Cl], [E-M] and [Cl-M], with (p < 0.01). Anaemia was expressed as a percentage between individuals [CI-EC (p < 0.05)]. In terms of parasitaemia, there were differences for the groups [CI-M (p < 0.05)]. Anti-PvMSP-119 antibodies were detected in 51.2% (226/441) of the population. The level of cytokines evaluation revealed a large variation in TNF-α and IL-10 concentrations in the co-infected group. In this study we did not observe any influence of coinfection on the acquisition of IgG antibodies against PvMSP119, as well as on the profile of the cytokines that characterize the Th1 and Th2 patterns. However, co-infection increased TNF-α and IL-10 levels.
Collapse
Affiliation(s)
- Rubens Alex de Oliveira Menezes
- Postgraduate Program in the Biology of Infectious and Parasitic Agents, Federal University of Pará (UFPA), Belém, Pará State, Brazil
- Laboratory of morphofunctional and parasitic studies with impact on health (LEMPIS), Federal University of Amapá (UNIFAP), Macapa, Amapá State, Brazil
- * E-mail:
| | | | - Anapaula Martins Mendes
- UNIFAP/Oiapoque Binational Campus, Federal University of Amapá, Oiapoque, Amapá State, Brazil
| | | | - Mathieu Nacher
- Centre d’Investigation Clinique, CIC INSERM 1424, Centre Hospitalier de Cayenne, Cayenne, French Guiana
| | - Tamirys Simão Pimenta
- Postgraduate Program in Neuroscience and Cell Biology, UFPA, Belém, Pará State, Brazil
- Evandro Chagas Institute/Brazilian Secretariat of Health Surveillance (SVS)/Brazilian Ministry of Health (MS), Ananindeua, Pará State, Brazil
| | - Aline Collares Pinheiro de Sousa
- Evandro Chagas Institute/Brazilian Secretariat of Health Surveillance (SVS)/Brazilian Ministry of Health (MS), Ananindeua, Pará State, Brazil
| | | | - Maria Izabel de Jesus
- Evandro Chagas Institute/Brazilian Secretariat of Health Surveillance (SVS)/Brazilian Ministry of Health (MS), Ananindeua, Pará State, Brazil
| | - Martin Johannes Enk
- Evandro Chagas Institute/Brazilian Secretariat of Health Surveillance (SVS)/Brazilian Ministry of Health (MS), Ananindeua, Pará State, Brazil
| | - Maristela Gomes Cunha
- Postgraduate Program in the Biology of Infectious and Parasitic Agents, Federal University of Pará (UFPA), Belém, Pará State, Brazil
- Laboratory of Microbiology and Immunology, Federal University of Pará (UFPA), Belém, Pará State, Brazil
| | - Ricardo Luiz Dantas Machado
- Postgraduate Program in the Biology of Infectious and Parasitic Agents, Federal University of Pará (UFPA), Belém, Pará State, Brazil
- Evandro Chagas Institute/Brazilian Secretariat of Health Surveillance (SVS)/Brazilian Ministry of Health (MS), Ananindeua, Pará State, Brazil
- Fluminense Federal University, Niterói, Rio de Janeiro State, Brazil
| |
Collapse
|
39
|
Natural Parasite Exposure Induces Protective Human Anti-Malarial Antibodies. Immunity 2017; 47:1197-1209.e10. [PMID: 29195810 DOI: 10.1016/j.immuni.2017.11.007] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 08/22/2017] [Accepted: 11/04/2017] [Indexed: 12/21/2022]
Abstract
Antibodies against the NANP repeat of circumsporozoite protein (CSP), the major surface antigen of Plasmodium falciparum (Pf) sporozoites, can protect from malaria in animal models but protective humoral immunity is difficult to induce in humans. Here we cloned and characterized rare affinity-matured human NANP-reactive memory B cell antibodies elicited by natural Pf exposure that potently inhibited parasite transmission and development in vivo. We unveiled the molecular details of antibody binding to two distinct protective epitopes within the NANP repeat. NANP repeat recognition was largely mediated by germline encoded and immunoglobulin (Ig) heavy-chain complementarity determining region 3 (HCDR3) residues, whereas affinity maturation contributed predominantly to stabilizing the antigen-binding site conformation. Combined, our findings illustrate the power of exploring human anti-CSP antibody responses to develop tools for malaria control in the mammalian and the mosquito vector and provide a molecular basis for the structure-based design of next-generation CSP malaria vaccines.
Collapse
|
40
|
Van Braeckel-Budimir N, Gras S, Ladell K, Josephs TM, Pewe L, Urban SL, Miners KL, Farenc C, Price DA, Rossjohn J, Harty JT. A T Cell Receptor Locus Harbors a Malaria-Specific Immune Response Gene. Immunity 2017; 47:835-847.e4. [PMID: 29150238 DOI: 10.1016/j.immuni.2017.10.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 08/02/2017] [Accepted: 10/26/2017] [Indexed: 01/05/2023]
Abstract
Immune response (Ir) genes, originally proposed by Baruj Benacerraf to explain differential antigen-specific responses in animal models, have become synonymous with the major histocompatibility complex (MHC). We discovered a non-MHC-linked Ir gene in a T cell receptor (TCR) locus that was required for CD8+ T cell responses to the Plasmodium berghei GAP5040-48 epitope in mice expressing the MHC class I allele H-2Db. GAP5040-48-specific CD8+ T cell responses emerged from a very large pool of naive Vβ8.1+ precursors, which dictated susceptibility to cerebral malaria and conferred protection against recombinant Listeria monocytogenes infection. Structural analysis of a prototypical Vβ8.1+ TCR-H-2Db-GAP5040-48 ternary complex revealed that germline-encoded complementarity-determining region 1β residues present exclusively in the Vβ8.1 segment mediated essential interactions with the GAP5040-48 peptide. Collectively, these findings demonstrated that Vβ8.1 functioned as an Ir gene that was indispensable for immune reactivity against the malaria GAP5040-48 epitope.
Collapse
Affiliation(s)
| | - Stephanie Gras
- Australian Research Council Centre of Excellence for Advanced Molecular Imaging, Monash University, Clayton, VIC 3800, Australia; Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Kristin Ladell
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, UK
| | - Tracy M Josephs
- Australian Research Council Centre of Excellence for Advanced Molecular Imaging, Monash University, Clayton, VIC 3800, Australia; Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Lecia Pewe
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA
| | - Stina L Urban
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA
| | - Kelly L Miners
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, UK
| | - Carine Farenc
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - David A Price
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, UK; Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA.
| | - Jamie Rossjohn
- Australian Research Council Centre of Excellence for Advanced Molecular Imaging, Monash University, Clayton, VIC 3800, Australia; Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, UK.
| | - John T Harty
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA; Department of Pathology, University of Iowa, Iowa City, IA 52242, USA; Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
41
|
Abstract
The two ligands B cell-activating factor of the tumor necrosis factor family (BAFF) and a proliferation-inducing ligand (APRIL) and the three receptors BAFF receptor (BAFF-R), transmembrane activator and calcium-modulating cyclophilin ligand interactor (TACI), and B cell maturation antigen (BCMA) are members of the "BAFF system molecules." BAFF system molecules are primarily involved in B cell homeostasis. The relevance of BAFF system molecules in host responses to microbial assaults has been investigated in clinical studies and in mice deficient for each of these molecules. Many microbial products modulate the expression of these molecules. Data from clinical studies suggest a correlation between increased expression levels of BAFF system molecules and elevated B cell responses. Depending on the pathogen, heightened B cell responses may strengthen the host response or promote susceptibility. Whereas pathogen-mediated increases in the expression levels of the ligands and/or the receptors appear to promote microbial clearance, certain pathogens have evolved to ablate B cell responses by suppressing the expression of TACI and/or BAFF-R on B cells. Other than its well-established role in B cell responses, the TACI-mediated activation of macrophages is also implicated in resistance to intracellular pathogens. An improved understanding of the role that BAFF system molecules play in infection may assist in devising novel strategies for vaccine development.
Collapse
Affiliation(s)
- Jiro Sakai
- Laboratory of Bacterial Polysaccharides, Division of Bacterial Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Mustafa Akkoyunlu
- Laboratory of Bacterial Polysaccharides, Division of Bacterial Parasitic and Allergenic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
42
|
Simmons RA, Mboera L, Miranda ML, Morris A, Stresman G, Turner EL, Kramer R, Drakeley C, O'Meara WP. A longitudinal cohort study of malaria exposure and changing serostatus in a malaria endemic area of rural Tanzania. Malar J 2017; 16:309. [PMID: 28764717 PMCID: PMC5539976 DOI: 10.1186/s12936-017-1945-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 07/18/2017] [Indexed: 01/16/2023] Open
Abstract
Background Measurements of anti-malarial antibodies are increasingly used as a proxy of transmission intensity. Most serological surveys are based on the use of cross-sectional data that, when age-stratified, approximates historical patterns of transmission within a population. Comparatively few studies leverage longitudinal data to explicitly relate individual infection events with subsequent antibody responses. Methods The occurrence of seroconversion and seroreversion events for two Plasmodium falciparum asexual stage antigens (MSP-1 and AMA-1) was examined using three annual measurements of 691 individuals from a cohort of individuals in a malaria-endemic area of rural east-central Tanzania. Mixed-effect logistic regression models were employed to determine factors associated with changes in serostatus over time. Results While the expected population-level relationship between seroprevalence and disease incidence was observed, on an individual level the relationship between individual infections and the antibody response was complex. MSP-1 antibody responses were more dynamic in response to the occurrence and resolution of infection events than AMA-1, while the latter was more correlated with consecutive infections. The MSP-1 antibody response to an observed infection seemed to decay faster over time than the corresponding AMA-1 response. Surprisingly, there was no evidence of an age effect on the occurrence of a conversion or reversion event. Conclusions While the population-level results concur with previously published sero-epidemiological surveys, the individual-level results highlight the more complex relationship between detected infections and antibody dynamics than can be analysed using cross-sectional data. The longitudinal analysis of serological data may provide a powerful tool for teasing apart the complex relationship between infection events and the corresponding immune response, thereby improving the ability to rapidly assess the success or failure of malaria control programmes.
Collapse
Affiliation(s)
- Ryan A Simmons
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, 2721, Durham, NC, 27701, USA. .,Duke Global Health Institute, Duke University, 310 Trent Drive, Durham, NC, 27701, USA.
| | - Leonard Mboera
- National Institute for Medical Research, 3 Barack Obama Drive, P.O. Box 9653, 11101, Dar es Salaam, United Republic of Tanzania
| | | | - Alison Morris
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Gillian Stresman
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Elizabeth L Turner
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, 2721, Durham, NC, 27701, USA.,Duke Global Health Institute, Duke University, 310 Trent Drive, Durham, NC, 27701, USA
| | - Randall Kramer
- Duke Global Health Institute, Duke University, 310 Trent Drive, Durham, NC, 27701, USA
| | - Chris Drakeley
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Wendy P O'Meara
- Duke Global Health Institute, Duke University, 310 Trent Drive, Durham, NC, 27701, USA
| |
Collapse
|
43
|
Niang M, Niass O, Diagne N, Sarr FD, Faye MM, Diop F, Diouf B, Faye J, Badiane A, Perraut R, Sokhna C, Trape JF, Tall A, Toure-Balde A. Temporal analysis of IgG antibody responses to Plasmodium falciparum antigens in relation to changing malaria epidemiology in a West African setting. Malar J 2017; 16:283. [PMID: 28693608 PMCID: PMC5504683 DOI: 10.1186/s12936-017-1928-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 06/30/2017] [Indexed: 12/02/2022] Open
Abstract
Background Coordinated scaled-up malaria control interventions have substantially contributed to the dramatic decrease of malaria-related morbidity and mortality in several endemic countries, including Senegal. However, the impacts of a given malaria control intervention on vector and parasite populations, acquired immunity, and disease burden remain very poorly documented largely due to the lack of continuous surveys. This study took advantage of the sera bank established as part of the Dielmo longitudinal project to investigate the dynamics of IgG antibody responses that accompanied the epidemiological changes resulting from malaria control interventions. Schizonts crude extract of a local strain of Plasmodium falciparum (Pfsch07/03) was used in ELISA to measure and compare seroprevalence and magnitude of IgG antibody responses from 2000 to 2012. Results The prevalence of Pfsch07/03 IgG antibody responses progressively decreased from 97.25% in 2000 to 57.3% in 2012. The prevalence of Pfsch07/03 antibodies categorized between three different age groups (<7, 7–15, and >15 years) revealed increased seroprevalence with age ranging from 47.19 to 62.67 and 89.45%, respectively in (<7, 7–15, and >15 years) old age groups. A marked drop in seroprevalence was observed after 2008 and was significant in the younger (<7 years) and intermediate (7–15 years) age groups, unlike older individuals aged >15 years (p = 1.00). Conclusions The study revealed a substantial contribution of all malaria control interventions to the decrease of IgG antibodies responses to Pfsch07/03 throughout prevention of human-mosquitos contacts, or reduction of parasite biomass. The present study demonstrates the wider potential of sero-epidemiological analysis in monitoring changes in malaria transmission resulting from a given malaria control intervention.
Collapse
Affiliation(s)
- Makhtar Niang
- Immunology Unit, Institut Pasteur Dakar, 36 Avenue Pasteur, BP 220, Dakar, Senegal
| | - Oumy Niass
- Immunology Unit, Institut Pasteur Dakar, 36 Avenue Pasteur, BP 220, Dakar, Senegal
| | - Nafissatou Diagne
- Institut de Recherche pour le développement, BP 1386, Dakar, Senegal
| | - Fatoumata Diene Sarr
- Epidemiology of Infectious Diseases Unit, Institut Pasteur Dakar, 36 Avenue Pasteur, BP 220, Dakar, Senegal
| | - Michel Matar Faye
- Immunology Unit, Institut Pasteur Dakar, 36 Avenue Pasteur, BP 220, Dakar, Senegal
| | - Fode Diop
- Immunology Unit, Institut Pasteur Dakar, 36 Avenue Pasteur, BP 220, Dakar, Senegal
| | - Babacar Diouf
- Immunology Unit, Institut Pasteur Dakar, 36 Avenue Pasteur, BP 220, Dakar, Senegal
| | - Joseph Faye
- Epidemiology of Infectious Diseases Unit, Institut Pasteur Dakar, 36 Avenue Pasteur, BP 220, Dakar, Senegal
| | - Abdoulaye Badiane
- Epidemiology of Infectious Diseases Unit, Institut Pasteur Dakar, 36 Avenue Pasteur, BP 220, Dakar, Senegal
| | - Ronald Perraut
- Immunology Unit, Institut Pasteur Dakar, 36 Avenue Pasteur, BP 220, Dakar, Senegal
| | - Cheikh Sokhna
- Institut de Recherche pour le développement, BP 1386, Dakar, Senegal
| | | | - Adama Tall
- Epidemiology of Infectious Diseases Unit, Institut Pasteur Dakar, 36 Avenue Pasteur, BP 220, Dakar, Senegal
| | - Aissatou Toure-Balde
- Immunology Unit, Institut Pasteur Dakar, 36 Avenue Pasteur, BP 220, Dakar, Senegal.
| |
Collapse
|
44
|
Solana JC, Ramírez L, Corvo L, de Oliveira CI, Barral-Netto M, Requena JM, Iborra S, Soto M. Vaccination with a Leishmania infantum HSP70-II null mutant confers long-term protective immunity against Leishmania major infection in two mice models. PLoS Negl Trop Dis 2017; 11:e0005644. [PMID: 28558043 PMCID: PMC5466331 DOI: 10.1371/journal.pntd.0005644] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 06/09/2017] [Accepted: 05/16/2017] [Indexed: 12/24/2022] Open
Abstract
Background The immunization with genetically attenuated Leishmania cell lines has been associated to the induction of memory and effector T cell responses against Leishmania able to control subsequent challenges. A Leishmania infantum null mutant for the HSP70-II genes has been described, possessing a non-virulent phenotype. Methodology/Principal findings The L. infantum attenuated parasites (LiΔHSP70-II) were inoculated in BALB/c (intravenously and subcutaneously) and C57BL/6 (subcutaneously) mice. An asymptomatic infection was generated and parasites diminished progressively to become undetectable in most of the analyzed organs. However, inoculation resulted in the long-term induction of parasite specific IFN-γ responses able to control the disease caused by a challenge of L. major infective promastigotes. BALB/c susceptible mice showed very low lesion development and a drastic decrease in parasite burdens in the lymph nodes draining the site of infection and internal organs. C57BL/6 mice did not show clinical manifestation of disease, correlated to the rapid migration of Leishmania specific IFN-γ producing T cells to the site of infection. Conclusion/Significance Inoculation of the LiΔHSP70-II attenuated line activates mammalian immune system for inducing moderate pro-inflammatory responses. These responses are able to confer long-term protection in mice against the infection of L. major virulent parasites. Despite numerous efforts made, a vaccine against leishmaniasis for humans is not available. Attempts based on parasite fractions or selected antigens failed to confer long lasting protection. On the other side, leishmanization, which consists in the inoculation of live virulent parasites in hidden parts of the body, is effective against cutaneous leishmaniasis in humans but objectionable in terms of biosafety. Some efforts have been made to design live vaccines to make leishmanization safer. A promising strategy is the development of genetically attenuated parasites, able to confer immunity without undesirable side effects. Here, we have employed an attenuated L. infantum line (LiΔHSP70-II) as a vaccine against heterologous challenge with L. major in two experimental models. Infection with LiΔHSP70-II parasites does not cause pathology and induces long-term protection based on the induction of IFN-γ producing T cells that are recruited rapidly and specifically to the site of challenge with the virulent parasites. These results support the idea of using attenuated parasites for vaccination.
Collapse
Affiliation(s)
- José Carlos Solana
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Departamento de Biología Molecular. Nicolás Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
| | - Laura Ramírez
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Departamento de Biología Molecular. Nicolás Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
| | - Laura Corvo
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Departamento de Biología Molecular. Nicolás Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
| | | | - Manoel Barral-Netto
- Centro de Pesquisas Gonçalo Moniz (Fundação Oswaldo Cruz-FIOCRUZ). Salvador, Bahia, Brazil
| | - José María Requena
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Departamento de Biología Molecular. Nicolás Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
| | - Salvador Iborra
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Departamento de Biología Molecular. Nicolás Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
- * E-mail: (SI); (MS)
| | - Manuel Soto
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Departamento de Biología Molecular. Nicolás Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
- * E-mail: (SI); (MS)
| |
Collapse
|
45
|
Tomasini N, Ragone PG, Gourbière S, Aparicio JP, Diosque P. Epidemiological modeling of Trypanosoma cruzi: Low stercorarian transmission and failure of host adaptive immunity explain the frequency of mixed infections in humans. PLoS Comput Biol 2017; 13:e1005532. [PMID: 28481887 PMCID: PMC5440054 DOI: 10.1371/journal.pcbi.1005532] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 05/22/2017] [Accepted: 04/24/2017] [Indexed: 01/27/2023] Open
Abstract
People living in areas with active vector-borne transmission of Chagas disease have multiple contacts with its causative agent, Trypanosoma cruzi. Reinfections by T. cruzi are possible at least in animal models leading to lower or even hardly detectable parasitaemia. In humans, although reinfections are thought to have major public health implications by increasing the risk of chronic manifestations of the disease, there is little quantitative knowledge about their frequency and the timing of parasite re-inoculation in the course of the disease. Here, we implemented stochastic agent-based models i) to estimate the rate of re-inoculation in humans and ii) to assess how frequent are reinfections during the acute and chronic stages of the disease according to alternative hypotheses on the adaptive immune response following a primary infection. By using a hybrid genetic algorithm, the models were fitted to epidemiological data of Argentinean rural villages where mixed infections by different genotypes of T. cruzi reach 56% in humans. To explain this percentage, the best model predicted 0.032 (0.008-0.042) annual reinfections per individual with 98.4% of them occurring in the chronic phase. In addition, the parasite escapes to the adaptive immune response mounted after the primary infection in at least 20% of the events of re-inoculation. With these low annual rates, the risks of reinfection during the typically long chronic stage of the disease stand around 14% (4%-18%) and 60% (21%-70%) after 5 and 30 years, with most individuals being re-infected 1-3 times overall. These low rates are better explained by the weak efficiency of the stercorarian mode of transmission than a highly efficient adaptive immune response. Those estimates are of particular interest for vaccine development and for our understanding of the higher risk of chronic disease manifestations suffered by infected people living in endemic areas.
Collapse
Affiliation(s)
- Nicolás Tomasini
- Instituto de Patología Experimental, Facultad de Ciencias de la Salud, CONICET, Universidad Nacional de Salta, Salta, Argentina
| | - Paula Gabriela Ragone
- Instituto de Patología Experimental, Facultad de Ciencias de la Salud, CONICET, Universidad Nacional de Salta, Salta, Argentina
| | - Sébastien Gourbière
- UMR 228 ESPACE-DEV-IMAGES, ‘Institut de Modélisation et d'Analyses en Géo-Environnement et Santé’, Université de Perpignan Via Domitia, Perpignan, France
| | - Juan Pablo Aparicio
- Instituto de Investigaciones en Energía no Convencional, CONICET, Universidad Nacional de Salta, Salta, Argentina
| | - Patricio Diosque
- Instituto de Patología Experimental, Facultad de Ciencias de la Salud, CONICET, Universidad Nacional de Salta, Salta, Argentina
| |
Collapse
|
46
|
Strangward P, Haley MJ, Shaw TN, Schwartz JM, Greig R, Mironov A, de Souza JB, Cruickshank SM, Craig AG, Milner DA, Allan SM, Couper KN. A quantitative brain map of experimental cerebral malaria pathology. PLoS Pathog 2017; 13:e1006267. [PMID: 28273147 PMCID: PMC5358898 DOI: 10.1371/journal.ppat.1006267] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 03/20/2017] [Accepted: 03/01/2017] [Indexed: 11/19/2022] Open
Abstract
The murine model of experimental cerebral malaria (ECM) has been utilised extensively in recent years to study the pathogenesis of human cerebral malaria (HCM). However, it has been proposed that the aetiologies of ECM and HCM are distinct, and, consequently, no useful mechanistic insights into the pathogenesis of HCM can be obtained from studying the ECM model. Therefore, in order to determine the similarities and differences in the pathology of ECM and HCM, we have performed the first spatial and quantitative histopathological assessment of the ECM syndrome. We demonstrate that the accumulation of parasitised red blood cells (pRBCs) in brain capillaries is a specific feature of ECM that is not observed during mild murine malaria infections. Critically, we show that individual pRBCs appear to occlude murine brain capillaries during ECM. As pRBC-mediated congestion of brain microvessels is a hallmark of HCM, this suggests that the impact of parasite accumulation on cerebral blood flow may ultimately be similar in mice and humans during ECM and HCM, respectively. Additionally, we demonstrate that cerebrovascular CD8+ T-cells appear to co-localise with accumulated pRBCs, an event that corresponds with development of widespread vascular leakage. As in HCM, we show that vascular leakage is not dependent on extensive vascular destruction. Instead, we show that vascular leakage is associated with alterations in transcellular and paracellular transport mechanisms. Finally, as in HCM, we observed axonal injury and demyelination in ECM adjacent to diverse vasculopathies. Collectively, our data therefore shows that, despite very different presentation, and apparently distinct mechanisms, of parasite accumulation, there appear to be a number of comparable features of cerebral pathology in mice and in humans during ECM and HCM, respectively. Thus, when used appropriately, the ECM model may be useful for studying specific pathological features of HCM.
Collapse
Affiliation(s)
- Patrick Strangward
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Michael J. Haley
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Tovah N. Shaw
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Jean-Marc Schwartz
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Rachel Greig
- Immunology Unit, Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Aleksandr Mironov
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - J. Brian de Souza
- Immunology Unit, Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Sheena M. Cruickshank
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Alister G. Craig
- Department of Molecular and Biochemical Parasitology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Danny A. Milner
- Department of Pathology, The Brigham & Women’s Hospital, Boston, Massachusetts, United States of America
| | - Stuart M. Allan
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Kevin N. Couper
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
- * E-mail:
| |
Collapse
|
47
|
Safety and efficacy of PfSPZ Vaccine against Plasmodium falciparum via direct venous inoculation in healthy malaria-exposed adults in Mali: a randomised, double-blind phase 1 trial. THE LANCET. INFECTIOUS DISEASES 2017; 17:498-509. [PMID: 28216244 DOI: 10.1016/s1473-3099(17)30104-4] [Citation(s) in RCA: 235] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 01/04/2017] [Accepted: 01/12/2017] [Indexed: 12/28/2022]
Abstract
BACKGROUND Plasmodium falciparum sporozite (PfSPZ) Vaccine is a metabolically active, non-replicating, whole malaria sporozoite vaccine that has been reported to be safe and protective against P falciparum controlled human malaria infection in malaria-naive individuals. We aimed to assess the safety and protective efficacy of PfSPZ Vaccine against naturally acquired P falciparum in malaria-experienced adults in Mali. METHODS After an open-label dose-escalation study in a pilot safety cohort, we did a double-blind, randomised, placebo-controlled trial based in Donéguébougou and surrounding villages in Mali. We recruited 18-35-year-old healthy adults who were randomly assigned (1:1) in a double-blind manner, with stratification by village and block randomisation, to receive either five doses of 2·7 × 105 PfSPZ or normal saline at days 0, 28, 56, 84, and 140 during the dry season (January to July inclusive). Participants and investigators were masked to group assignments, which were unmasked at the final study visit, 6 months after receipt of the last vaccination. Participants received combined artemether and lumefantrine (four tablets, each containing 20 mg artemether and 120 mg lumefantrine, given twice per day over 3 days for a total of six doses) to eliminate P falciparum before the first and last vaccinations. We collected blood smears every 2 weeks and during any illness for 24 weeks after the fifth vaccination. The primary outcome was the safety and tolerability of the vaccine, assessed as local and systemic reactogenicity and adverse events. The sample size was calculated for the exploratory efficacy endpoint of time to first P falciparum infection beginning 28 days after the fifth vaccination. The safety analysis included all participants who received at least one dose of investigational product, whereas the efficacy analyses included only participants who received all five vaccinations. This trial is registered at ClinicalTrials.gov, number NCT01988636. FINDINGS Between Jan 18 and Feb 24, 2014, we enrolled 93 participants into the main study cohort with 46 participants assigned PfSPZ Vaccine and 47 assigned placebo, all of whom were evaluable for safety. We detected no significant differences in local or systemic adverse events or laboratory abnormalities between the PfSPZ Vaccine and placebo groups, and only grade 1 (mild) local or systemic adverse events occurred in both groups. The most common solicited systemic adverse event in the vaccine and placebo groups was headache (three [7%] people in the vaccine group vs four [9%] in the placebo group) followed by fatigue (one [2%] person in the placebo group), fever (one [2%] person in the placebo group), and myalgia (one [2%] person in each group). The exploratory efficacy analysis included 41 participants from the vaccine group and 40 from the placebo group. Of these participants, 37 (93%) from the placebo group and 27 (66%) from the vaccine group developed P falciparum infection. The hazard ratio for vaccine efficacy was 0·517 (95% CI 0·313-0·856) by time-to-infection analysis (log-rank p=0·01), and 0·712 (0·528-0·918) by proportional analysis (p=0·006). INTERPRETATION PfSPZ Vaccine was well tolerated and safe. PfSPZ Vaccine showed significant protection in African adults against P falciparum infection throughout an entire malaria season. FUNDING US National Institutes of Health Intramural Research Program, Sanaria.
Collapse
|
48
|
Hojo-Souza NS, Pereira DB, de Souza FSH, de Oliveira Mendes TA, Cardoso MS, Tada MS, Zanini GM, Bartholomeu DC, Fujiwara RT, Bueno LL. On the cytokine/chemokine network during Plasmodium vivax malaria: new insights to understand the disease. Malar J 2017; 16:42. [PMID: 28118834 PMCID: PMC5260126 DOI: 10.1186/s12936-017-1683-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 01/05/2017] [Indexed: 11/10/2022] Open
Abstract
Background The clinical outcome of malaria depends on the delicate balance between pro-inflammatory and immunomodulatory cytokine responses triggered during infection. Despite the numerous reports on characterization of plasma levels of cytokines/chemokines, there is no consensus on the profile of these mediators during blood stage malaria. The identification of acute phase biomarkers might contribute to a better understanding of the disease, allowing the use of more effective therapeutic approaches to prevent the progression towards severe disease. In the present study, the plasma levels of cytokines and chemokines and their association with parasitaemia and number of previous malaria episodes were evaluated in Plasmodium vivax-infected patients during acute and convalescence phase, as well as in healthy donors. Methods Samples of plasma were obtained from peripheral blood samples from four different groups: P. vivax-infected, P. vivax-treated, endemic control and malaria-naïve control. The cytokine (IL-6, IL-10, IL-17, IL-27, TGF-β, IFN-γ and TNF) and chemokine (MCP-1/CCL2, IP-10/CXCL10 and RANTES/CCL5) plasma levels were measured by CBA or ELISA. The network analysis was performed using Spearman correlation coefficient. Results Plasmodium vivax infection induced a pro-inflammatory response driven by IL-6 and IL-17 associated with an immunomodulatory profile mediated by IL-10 and TGF-β. In addition, a reduction was observed of IFN-γ plasma levels in P. vivax group. A lower level of IL-27 was observed in endemic control group in comparison to malaria-naïve control group. No significant results were found for IL-12p40 and TNF. It was also observed that P. vivax infection promoted higher levels of MCP-1/CCL2 and IP-10/CXCL10 and lower levels of RANTES/CCL5. The plasma level of IL-10 was elevated in patients with high parasitaemia and with more than five previous malaria episodes. Furthermore, association profile between cytokine and chemokine levels were observed by correlation network analysis indicating signature patterns associated with different parasitaemia levels. Conclusions The P. vivax infection triggers a balanced immune response mediated by IL-6 and MCP-1/CCL2, which is modulated by IL-10. In addition, the results indicated that IL-10 plasma levels are influenced by parasitaemia and number of previous malaria episodes.
Collapse
Affiliation(s)
- Natália Satchiko Hojo-Souza
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Fernanda Sumika Hojo de Souza
- Departamento de Ciência da Computação, Universidade Federal de São João del-Rei, São João del-Rei, Minas Gerais, Brazil
| | | | - Mariana Santos Cardoso
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Graziela Maria Zanini
- Instituto de Pesquisa Clínica Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Daniella Castanheira Bartholomeu
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ricardo Toshio Fujiwara
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Lilian Lacerda Bueno
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
49
|
Lugaajju A, Reddy SB, Wahlgren M, Kironde F, Persson KEM. Development of Plasmodium falciparum specific naïve, atypical, memory and plasma B cells during infancy and in adults in an endemic area. Malar J 2017; 16:37. [PMID: 28109284 PMCID: PMC5251336 DOI: 10.1186/s12936-017-1697-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 01/13/2017] [Indexed: 11/12/2022] Open
Abstract
Background B-cells are essential in immunity against malaria, but which sub-sets of B-cells specifically recognize Plasmodium falciparum and when they appear is still largely unknown. Results Using the flow cytometry technique for detection of P. falciparum specific (Pf+) B-cells, this study for the first time measured the development of Pf+ B cell (CD19+) phenotypes in Ugandan babies from birth up to nine months, and in their mothers. The babies showed increases in Pf+ IgG memory B-cells (MBCs), atypical MBCs, and plasma cells/blasts over time, but the proportion of these cells were still lower than in the mothers who displayed stable levels (5, 18, and 3%, respectively). Pf+ non-IgG+ MBCs and naïve B-cells binding to P. falciparum antigens were higher in the babies compared to the mothers (12 and 50%). In ELISA there was an increase in IgG and IgM antibodies over time in babies, and stable levels in mothers. At baby delivery, multigravidae mothers had a higher proportion of Pf+ IgG MBCs and less Pf+ naïve B-cells than primigravidae mothers. Conclusions In newborns, naïve B-cells are a major player in recognizing P. falciparum. In adults, the high proportion of Pf+ atypical MBCs suggests a major role for these cells. Both in infants and adults, non-IgG+ MBCs were higher than IgG MBCs, indicating that these cells deserve more focus in future. Electronic supplementary material The online version of this article (doi:10.1186/s12936-017-1697-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Allan Lugaajju
- School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda.,Department of Microbiology, Tumor, and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Sreenivasulu B Reddy
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Mats Wahlgren
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Fred Kironde
- School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda.,Habib Medical School, Islamic University in Uganda (IUIU), Kampala, Uganda
| | - Kristina E M Persson
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institutet, Stockholm, Sweden. .,Department of Laboratory Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
50
|
Montes de Oca M, Good MF, McCarthy JS, Engwerda CR. The Impact of Established Immunoregulatory Networks on Vaccine Efficacy and the Development of Immunity to Malaria. THE JOURNAL OF IMMUNOLOGY 2016; 197:4518-4526. [DOI: 10.4049/jimmunol.1600619] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 08/26/2016] [Indexed: 02/07/2023]
|