1
|
Thawornpan P, Salsabila ZZ, Kochayoo P, Khunsri T, Malee C, Wangriatisak K, Leepiyasakulchai C, Ntumngia FB, Adams JH, Chootong P. Polarization toward Tfh2 cell involved in development of MBC and antibody responses against Plasmodium vivax infection. PLoS Negl Trop Dis 2024; 18:e0012625. [PMID: 39475899 PMCID: PMC11524495 DOI: 10.1371/journal.pntd.0012625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 10/11/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Plasmodium vivax is the dominant Plasmodium spp. causing malaria throughout tropical and sub-tropical countries. Humoral immunity is induced during P. vivax infection. However, data on longevity of antibody and memory B cell (MBC) responses is lacking. Follicular helper T cells (Tfh) are drivers of high-affinity and long-lived antibody responses. Understanding of Tfh-mediated immunity against malaria is valuable for vaccine development. METHODOLOGY/PRINCIPAL FINDINGS We enrolled 31 acutely infected P. vivax patients in low malaria transmission areas of Thailand to detect frequencies, phenotypes and kinetics of different subsets of circulating Tfh (cTfh) and MBCs, and to evaluate their association with humoral immunity following natural P. vivax infection. Expansion of cTfh2 cells, activated and atypical MBCs were shown during acute malaria. To relate increased cTfh2 cells to humoral immunity, P. vivax-specific MBCs and antibodies were assessed. High anti-PvCSP and -PvDBPII seropositivity was detected and most subjects produced MBCs specific to these antigens. The increased cTfh2 cells were positively related to atypical MBCs, plasmablasts/plasma cells, and anti-PvDBPII IgM and IgG levels. Distributions of memory cTfh cell subsets were altered from central memory (CM) to effector memory (EM) during infection. The highest ratios of cTfh-EM/cTfh-CM were represented in cTfh2 cells. Positive correlation of cTfh17-EM with activated and atypical MBCs was observed, while cTfh2-CM and cTfh17-CM cells were positively related to PvDBPII-specific MBCs and IgM levels. CONCLUSIONS/SIGNIFICANCE Present study demonstrated that P. vivax infection induced cTfh polarization into cTfh2 subset, and alteration of memory cTfh2 phenotype from CM to EM phase. These P. vivax-induced cTfh responses significantly associated with generation of MBCs and antibody responses. Therefore, cTfh2 cells might possibly influence humoral immunity by inducing expansion of activated and atypical MBCs, and by generating P. vivax-specific MBCs and antibody responses following natural infection.
Collapse
Affiliation(s)
- Pongsakorn Thawornpan
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Zulfa Zahra Salsabila
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Piyawan Kochayoo
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Tipanan Khunsri
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Chayapat Malee
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Kittikorn Wangriatisak
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Chaniya Leepiyasakulchai
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Francis Babila Ntumngia
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - John H. Adams
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - Patchanee Chootong
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| |
Collapse
|
2
|
Bravo M, Dileepan T, Dolan M, Hildebrand J, Wolford J, Hanson ID, Hamilton SE, Frosch AE, Burrack KS. IL-15 Complex-Induced IL-10 Enhances Plasmodium-specific CD4+ T Follicular Helper Differentiation and Antibody Production. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:992-1001. [PMID: 38305633 PMCID: PMC10932862 DOI: 10.4049/jimmunol.2300525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 01/09/2024] [Indexed: 02/03/2024]
Abstract
Malaria, which results from infection with Plasmodium parasites, remains a major public health problem. Although humans do not develop long-lived, sterilizing immunity, protection against symptomatic disease develops after repeated exposure to Plasmodium parasites and correlates with the acquisition of humoral immunity. Despite the established role Abs play in protection from malaria disease, dysregulated inflammation is thought to contribute to the suboptimal immune response to Plasmodium infection. Plasmodium berghei ANKA (PbA) infection results in a fatal severe malaria disease in mice. We previously demonstrated that treatment of mice with IL-15 complex (IL-15C; IL-15 bound to an IL-15Rα-Fc fusion protein) induces IL-10 expression in NK cells, which protects mice from PbA-induced death. Using a novel MHC class II tetramer to identify PbA-specific CD4+ T cells, in this study we demonstrate that IL-15C treatment enhances T follicular helper (Tfh) differentiation and modulates cytokine production by CD4+ T cells. Moreover, genetic deletion of NK cell-derived IL-10 or IL-10R expression on T cells prevents IL-15C-induced Tfh differentiation. Additionally, IL-15C treatment results in increased anti-PbA IgG Ab levels and improves survival following reinfection. Overall, these data demonstrate that IL-15C treatment, via its induction of IL-10 from NK cells, modulates the dysregulated inflammation during Plasmodium infection to promote Tfh differentiation and Ab generation, correlating with improved survival from reinfection. These findings will facilitate improved control of malaria infection and protection from disease by informing therapeutic strategies and vaccine design.
Collapse
Affiliation(s)
| | | | | | - Jacob Hildebrand
- Center for Immunology, University of Minnesota
- Department of Laboratory Medicine and Pathology, University of Minnesota
| | | | | | - Sara E. Hamilton
- Center for Immunology, University of Minnesota
- Department of Laboratory Medicine and Pathology, University of Minnesota
| | - Anne E. Frosch
- Hennepin Healthcare Research Institute
- Center for Immunology, University of Minnesota
| | - Kristina S. Burrack
- Hennepin Healthcare Research Institute
- Center for Immunology, University of Minnesota
| |
Collapse
|
3
|
Ren Z, Shi Q, Xu S, Xu J, Yin Y, Lin Z, Xu S, Ma X, Liu Y, Zhu G, He X, Lu J, Li Y, Zhang W, Liu J, Yang Y, Han ET, Cao J, Lu F. Elicitation of T-cell-derived IFN-γ-dependent immunity by highly conserved Plasmodium ovale curtisi Duffy binding protein domain region II (PocDBP-RII). Parasit Vectors 2023; 16:269. [PMID: 37553591 PMCID: PMC10410920 DOI: 10.1186/s13071-023-05897-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/27/2023] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND Infections with Plasmodium ovale are widely distributed but rarely investigated, and the resulting burden of disease has been underestimated. Plasmodium ovale curtisi Duffy binding protein domain region II (PocDBP-RII) is an essential ligand for reticulocyte recognition and host cell invasion by P. ovale curtisi. However, the genomic variation, antigenicity and immunogenicity of PocDBP-RII remain major knowledge gaps. METHODS A total of 93 P. ovale curtisi samples were collected from migrant workers who returned to China from 17 countries in Africa between 2012 and 2016. The genetic polymorphism, natural selection and copy number variation (CNV) were investigated by sequencing and real-time PCR. The antigenicity and immunogenicity of the recombinant PocDBP-RII (rPocDBP-RII) protein were further examined, and the humoral and cellular responses of immunized mice were assessed using protein microarrays and flow cytometry. RESULTS Efficiently expressed and purified rPocDBP-RII (39 kDa) was successfully used as an antigen for immunization in mice. The haplotype diversity (Hd) of PocDBP-RII gene was 0.105, and the nucleotide diversity index (π) was 0.00011. No increased copy number was found among the collected isolates of P. ovale curtisi. Furthermore, rPocDBP-RII induced persistent antigen-specific antibody production with a serum IgG antibody titer of 1: 16,000. IFN-γ-producing T cells, rather than IL-10-producing cells, were activated in response to the stimulation of rPocDBP-RII. Compared to PBS-immunized mice (negative control), there was a higher percentage of CD4+CD44highCD62L- T cells (effector memory T cells) and CD8+CD44highCD62L+ T cells (central memory T cells) in rPocDBP-RII‑immunized mice. CONCLUSIONS PocDBP-RII sequences were highly conserved in clinical isolates of P. ovale curtisi. rPocDBP-RII protein could mediate protective blood-stage immunity through IFN-γ-producing CD4+ and CD8+ T cells and memory T cells, in addition to inducing specific antibodies. Our results suggested that rPocDBP-RII protein has potential as a vaccine candidate to provide assessment and guidance for malaria control and elimination activities.
Collapse
Affiliation(s)
- Zhenyu Ren
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Qiyang Shi
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory On Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China
| | - Simin Xu
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
- Changshu Second People's Hospital, Suzhou, 215500, Jiangsu, People's Republic of China
| | - Jiahui Xu
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Yi Yin
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Zhijie Lin
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Sui Xu
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory On Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China
| | - Xiaoqin Ma
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory On Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China
| | - Yaobao Liu
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory On Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China
| | - Guoding Zhu
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory On Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China
| | - Xinlong He
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Jingyuan Lu
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Yinyue Li
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Wenwen Zhang
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Jiali Liu
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Yun Yang
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 24341, Republic of Korea
| | - Jun Cao
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory On Parasite and Vector Control Technology, Jiangsu Provincial Medical Key Laboratory, Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China.
| | - Feng Lu
- Department of Pathogenic Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.
- Affiliated Hospital of Yangzhou University, Yangzhou, 225000, People's Republic of China.
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, School of Medicine, Yangzhou University, Yangzhou, 225009, People's Republic of China.
| |
Collapse
|
4
|
Ferreira NS, Lima NF, Sulczewski FB, Soares IS, Ferreira MU, Boscardin SB. Plasmodium vivax infection alters the peripheral immunoregulatory network of CD4 T follicular cells and B cells. Eur J Immunol 2023; 53:e2350372. [PMID: 37160134 DOI: 10.1002/eji.202350372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 04/10/2023] [Accepted: 05/08/2023] [Indexed: 05/11/2023]
Abstract
Regulatory and effector cell responses to Plasmodium vivax, the most common human malaria parasite outside Africa, remain understudied in naturally infected populations. Here, we describe peripheral CD4+ T- and B-cell populations during and shortly after an uncomplicated P. vivax infection in 38 continuously exposed adult Amazonians. Consistent with previous observations, we found an increased frequency in CD4+ CD45RA- CD25+ FoxP3+ T regulatory cells that express the inhibitory molecule CTLA-4 during the acute infection, with a sustained expansion of CD21- CD27- atypical memory cells within the CD19+ B-cell compartment. Both Th1- and Th2-type subsets of CXCR5+ ICOShi PD-1+ circulating T follicular helper (cTfh) cells, which are thought to contribute to antibody production, were induced during P. vivax infection, with a positive correlation between overall cTfh cell frequency and IgG antibody titers to the P. vivax blood-stage antigen MSP119 . We identified significant changes in cell populations that had not been described in human malaria, such as an increased frequency of CTLA-4+ T follicular regulatory cells that antagonize Tfh cells, and a decreased frequency of circulating CD24hi CD27+ B regulatory cells in response to acute infection. In conclusion, we disclose a complex immunoregulatory network that is critical to understand how naturally acquired immunity develops in P. vivax malaria.
Collapse
Affiliation(s)
- Natália S Ferreira
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Nathália F Lima
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Fernando B Sulczewski
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Irene S Soares
- Department of Clinical and Toxicological Analyses, University of São Paulo, São Paulo, Brazil
| | - Marcelo U Ferreira
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Global Health and Tropical Medicine, Institute of Hygiene and Tropical Medicine, NOVA University of Lisbon, Lisbon, Portugal
| | - Silvia B Boscardin
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
5
|
Ibraheem Y, Bayarsaikhan G, Inoue SI. Host immunity to Plasmodium infection: Contribution of Plasmodium berghei to our understanding of T cell-related immune response to blood-stage malaria. Parasitol Int 2022; 92:102646. [PMID: 35998816 DOI: 10.1016/j.parint.2022.102646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 10/15/2022]
Abstract
Malaria is a life-threatening disease caused by infection with Plasmodium parasites. The goal of developing an effective malaria vaccine is yet to be reached despite decades of massive research efforts. CD4+ helper T cells, CD8+ cytotoxic T cells, and γδ T cells are associated with immune responses to both liver-stage and blood-stage Plasmodium infection. The immune responses of T cell-lineages to Plasmodium infection are associated with both protection and immunopathology. Studies with mouse model of malaria contribute to our understanding of host immune response. In this paper, we focus primarily on mouse malaria model with blood-stage Plasmodium berghei infection and review our knowledge of T cell immune responses against Plasmodium infection. Moreover, we also discuss findings of experimental human studies. Uncovering the precise mechanisms of T cell-mediated immunity to Plasmodium infection can be accomplished through further investigations using mouse models of malaria with rodent Plasmodium parasites. Those findings would be invaluable to advance the efforts for development of an effective malaria vaccine.
Collapse
Affiliation(s)
- Yarob Ibraheem
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki-city, Nagasaki 852-8523, Japan
| | - Ganchimeg Bayarsaikhan
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki-city, 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-city, Nagasaki 852-8523, Japan.
| |
Collapse
|
6
|
Recent Advances in Understanding the Inflammatory Response in Malaria: A Review of the Dual Role of Cytokines. J Immunol Res 2021; 2021:7785180. [PMID: 34790829 PMCID: PMC8592744 DOI: 10.1155/2021/7785180] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/01/2021] [Accepted: 10/15/2021] [Indexed: 11/25/2022] Open
Abstract
Malaria is a serious and, in some unfortunate cases, fatal disease caused by a parasite of the Plasmodium genus. It predominantly occurs in tropical areas where it is transmitted through the bite of an infected Anopheles mosquito. The pathogenesis of malaria is complex and incompletely elucidated. During blood-stage infection, in response to the presence of the parasite, the host's immune system produces proinflammatory cytokines including IL-6, IL-8, IFN-γ, and TNF, cytokines which play a pivotal role in controlling the growth of the parasite and its elimination. Regulatory cytokines such as transforming growth factor- (TGF-) β and IL-10 maintain the balance between the proinflammatory and anti-inflammatory responses. However, in many cases, cytokines have a double role. On the one hand, they contribute to parasitic clearance, and on the other, they are responsible for pathological changes encountered in malaria. Cytokine-modulating strategies may represent a promising modern approach in disease management. In this review, we discuss the host immune response in malaria, analyzing the latest studies on the roles of pro- and anti-inflammatory cytokines.
Collapse
|
7
|
Immunosuppression in Malaria: Do Plasmodium falciparum Parasites Hijack the Host? Pathogens 2021; 10:pathogens10101277. [PMID: 34684226 PMCID: PMC8536967 DOI: 10.3390/pathogens10101277] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/13/2022] Open
Abstract
Malaria reflects not only a state of immune activation, but also a state of general immune defect or immunosuppression, of complex etiology that can last longer than the actual episode. Inhabitants of malaria-endemic regions with lifelong exposure to the parasite show an exhausted or immune regulatory profile compared to non- or minimally exposed subjects. Several studies and experiments to identify and characterize the cause of this malaria-related immunosuppression have shown that malaria suppresses humoral and cellular responses to both homologous (Plasmodium) and heterologous antigens (e.g., vaccines). However, neither the underlying mechanisms nor the relative involvement of different types of immune cells in immunosuppression during malaria is well understood. Moreover, the implication of the parasite during the different stages of the modulation of immunity has not been addressed in detail. There is growing evidence of a role of immune regulators and cellular components in malaria that may lead to immunosuppression that needs further research. In this review, we summarize the current evidence on how malaria parasites may directly and indirectly induce immunosuppression and investigate the potential role of specific cell types, effector molecules and other immunoregulatory factors.
Collapse
|
8
|
Bonam SR, Rénia L, Tadepalli G, Bayry J, Kumar HMS. Plasmodium falciparum Malaria Vaccines and Vaccine Adjuvants. Vaccines (Basel) 2021; 9:1072. [PMID: 34696180 PMCID: PMC8541031 DOI: 10.3390/vaccines9101072] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/10/2021] [Accepted: 09/22/2021] [Indexed: 12/02/2022] Open
Abstract
Malaria-a parasite vector-borne disease-is a global health problem, and Plasmodium falciparum has proven to be the deadliest among Plasmodium spp., which causes malaria in humans. Symptoms of the disease range from mild fever and shivering to hemolytic anemia and neurological dysfunctions. The spread of drug resistance and the absence of effective vaccines has made malaria disease an ever-emerging problem. Although progress has been made in understanding the host response to the parasite, various aspects of its biology in its mammalian host are still unclear. In this context, there is a pressing demand for the development of effective preventive and therapeutic strategies, including new drugs and novel adjuvanted vaccines that elicit protective immunity. The present article provides an overview of the current knowledge of anti-malarial immunity against P. falciparum and different options of vaccine candidates in development. A special emphasis has been made on the mechanism of action of clinically used vaccine adjuvants.
Collapse
Affiliation(s)
- Srinivasa Reddy Bonam
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Equipe-Immunopathologie et Immunointervention Thérapeutique, Sorbonne Université, Université de Paris, F-75006 Paris, France;
| | - Laurent Rénia
- A*STAR Infectious Diseases Labs, 8A Biomedical Grove, Singapore 138648, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore 308232, Singapore
| | - Ganesh Tadepalli
- Vaccine Immunology Laboratory, Organic Synthesis and Process Chemistry Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India;
| | - Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Equipe-Immunopathologie et Immunointervention Thérapeutique, Sorbonne Université, Université de Paris, F-75006 Paris, France;
- Biological Sciences & Engineering, Indian Institute of Technology Palakkad, Palakkad 678623, India
| | - Halmuthur Mahabalarao Sampath Kumar
- Vaccine Immunology Laboratory, Organic Synthesis and Process Chemistry Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India;
| |
Collapse
|
9
|
Lamsfus Calle C, Fendel R, Singh A, Richie TL, Hoffman SL, Kremsner PG, Mordmüller B. Expansion of Functional Myeloid-Derived Suppressor Cells in Controlled Human Malaria Infection. Front Immunol 2021; 12:625712. [PMID: 33815377 PMCID: PMC8017236 DOI: 10.3389/fimmu.2021.625712] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 02/22/2021] [Indexed: 12/12/2022] Open
Abstract
Malaria can cause life-threatening complications which are often associated with inflammatory reactions. More subtle, but also contributing to the burden of disease are chronic, often subclinical infections, which result in conditions like anemia and immunologic hyporesponsiveness. Although very frequent, such infections are difficult to study in endemic regions because of interaction with concurrent infections and immune responses. In particular, knowledge about mechanisms of malaria-induced immunosuppression is scarce. We measured circulating immune cells by cytometry in healthy, malaria-naïve, adult volunteers undergoing controlled human malaria infection (CHMI) with a focus on potentially immunosuppressive cells. Infectious Plasmodium falciparum (Pf) sporozoites (SPZ) (PfSPZ Challenge) were inoculated during two independent studies to assess malaria vaccine efficacy. Volunteers were followed daily until parasites were detected in the circulation by RT-qPCR. This allowed us to analyze immune responses during pre-patency and at very low parasite densities in malaria-naïve healthy adults. We observed a consistent increase in circulating polymorphonuclear myeloid-derived suppressor cells (PMN-MDSC) in volunteers who developed P. falciparum blood stage parasitemia. The increase was independent of preceding vaccination with a pre-erythrocytic malaria vaccine. PMN-MDSC were functional, they suppressed CD4+ and CD8+ T cell proliferation as shown by ex-vivo co-cultivation with stimulated T cells. PMN-MDSC reduced T cell proliferation upon stimulation by about 50%. Interestingly, high circulating PMN-MDSC numbers were associated with lymphocytopenia. The number of circulating regulatory T cells (Treg) and monocytic MDSC (M-MDSC) showed no significant parasitemia-dependent variation. These results highlight PMN-MDSC in the peripheral circulation as an early indicator of infection during malaria. They suppress CD4+ and CD8+ T cell proliferation in vitro. Their contribution to immunosuppression in vivo in subclinical and uncomplicated malaria will be the subject of further research. Pre-emptive antimalarial pre-treatment of vaccinees to reverse malaria-associated PMN-MDSC immunosuppression could improve vaccine response in exposed individuals.
Collapse
Affiliation(s)
| | - Rolf Fendel
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany.,Centre de Recherches Médicales de Lambaréné (CERMEL), Lambaréné, Gabon
| | - Anurag Singh
- Department of Pediatrics 1, University Children's Hospital Tübingen, Tübingen, Germany.,Institute for Clinical and Experimental Transfusion Medicine, University Hospital Tübingen, Tübingen, Germany
| | | | | | - Peter G Kremsner
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany.,Centre de Recherches Médicales de Lambaréné (CERMEL), Lambaréné, Gabon
| | - Benjamin Mordmüller
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany.,German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany.,Centre de Recherches Médicales de Lambaréné (CERMEL), Lambaréné, Gabon
| |
Collapse
|
10
|
Braddom AE, Batugedara G, Bol S, Bunnik EM. Potential functions of atypical memory B cells in Plasmodium-exposed individuals. Int J Parasitol 2020; 50:1033-1042. [PMID: 32987039 PMCID: PMC7666103 DOI: 10.1016/j.ijpara.2020.08.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/28/2020] [Accepted: 08/01/2020] [Indexed: 02/08/2023]
Abstract
Naturally acquired iummunity against clinical malaria is slow to develop, taking years of repeated exposure to parasites to acquire sufficiently broad and potent antibody responses. Increasing evidence suggests that Plasmodium infection and the resulting immune stimulation contribute to changes in the B cell compartment. In particular, accumulation of atypical memory B cells (atMBCs) is common in Plasmodium-exposed individuals. Similarities to B cell subsets present in other acute and chronic disease settings have provided insight into the development and potential function of these cells; however, their contribution to protection against malaria is still poorly understood. Here, we discuss recent findings that have increased our understanding of atMBCs and outline outstanding questions related to their function and development in the protective immune response to malaria.
Collapse
Affiliation(s)
- Ashley E Braddom
- Department of Microbiology, Immunology and Molecular Genetics, Lozano Long School of Medicine, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Gayani Batugedara
- Department of Microbiology, Immunology and Molecular Genetics, Lozano Long School of Medicine, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Sebastiaan Bol
- Department of Microbiology, Immunology and Molecular Genetics, Lozano Long School of Medicine, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Evelien M Bunnik
- Department of Microbiology, Immunology and Molecular Genetics, Lozano Long School of Medicine, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.
| |
Collapse
|
11
|
Levack RC, Newell KL, Popescu M, Cabrera-Martinez B, Winslow GM. CD11c + T-bet + B Cells Require IL-21 and IFN-γ from Type 1 T Follicular Helper Cells and Intrinsic Bcl-6 Expression but Develop Normally in the Absence of T-bet. THE JOURNAL OF IMMUNOLOGY 2020; 205:1050-1058. [PMID: 32680956 DOI: 10.4049/jimmunol.2000206] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 06/16/2020] [Indexed: 12/11/2022]
Abstract
CD11c+ T-bet+ B cells generated during ehrlichial infection require CD4+ T cell help and IL-21 signaling for their development, but the exact T cell subset required had not been known. In this study, we show in a mouse model of Ehrlichia muris that type 1 T follicular helper (TFH1) cells provide help to CD11c+ T-bet+ B cells via the dual secretion of IL-21 and IFN-γ in a CD40/CD40L-dependent manner. TFH1 cell help was delivered in two phases: IFN-γ signals were provided early in infection, whereas CD40/CD40L help was provided late in infection. In contrast to T-bet+ T cells, T-bet+ B cells did not develop in the absence of B cell-intrinsic Bcl-6 but were generated in the absence of T-bet. T-bet-deficient memory B cells were largely indistinguishable from their wild-type counterparts, although they no longer underwent switching to IgG2c. These data suggest that a primary function of T-bet in B cells during ehrlichial infection is to promote appropriate class switching, not lineage specification. Thus, CD11c+ memory B cells develop normally without T-bet but require Bcl-6 and specialized help from dual cytokine-producing TFH1 cells.
Collapse
Affiliation(s)
- Russell C Levack
- Department of Microbiology and Immunology, Upstate Medical University, Syracuse, NY 13210
| | - Krista L Newell
- Department of Microbiology and Immunology, Upstate Medical University, Syracuse, NY 13210
| | - Maria Popescu
- Department of Microbiology and Immunology, Upstate Medical University, Syracuse, NY 13210
| | | | - Gary M Winslow
- Department of Microbiology and Immunology, Upstate Medical University, Syracuse, NY 13210
| |
Collapse
|
12
|
Yap XZ, McCall MBB, Sauerwein RW. Fast and fierce versus slow and smooth: Heterogeneity in immune responses to Plasmodium in the controlled human malaria infection model. Immunol Rev 2020; 293:253-269. [PMID: 31605396 PMCID: PMC6973142 DOI: 10.1111/imr.12811] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/11/2019] [Accepted: 09/13/2019] [Indexed: 12/19/2022]
Abstract
Controlled human malaria infection (CHMI) is an established model in clinical malaria research. Upon exposure to Plasmodium falciparum parasites, malaria-naive volunteers differ in dynamics and composition of their immune profiles and subsequent capacity to generate protective immunity. CHMI volunteers are either inflammatory responders who have prominent cellular IFN-γ production primarily driven by adaptive T cells, or tempered responders who skew toward antibody-mediated humoral immunity. When exposed to consecutive CHMIs under antimalarial chemoprophylaxis, individuals who can control parasitemia after a single immunization (fast responders) are more likely to be protected against a subsequent challenge infection. Fast responders tend to be inflammatory responders who can rapidly induce long-lived IFN-γ+ T cell responses. Slow responders or even non-responders can also be protected, but via a more diverse range of responses that take a longer time to reach full protective efficacy, in part due to their tempered phenotype. The latter group can be identified at baseline before CHMI by higher expression of inhibitory ligands CTLA-4 and TIM-3 on CD4+ T cells. Delineating heterogeneity in human immune responses to P. falciparum will facilitate rational design and strategy towards effective malaria vaccines.
Collapse
Affiliation(s)
- Xi Zen Yap
- Department of Medical MicrobiologyRadboud University Medical CenterNijmegenThe Netherlands
- Radboud Center for Infectious DiseasesRadboud University Medical CenterNijmegenThe Netherlands
| | - Matthew B. B. McCall
- Department of Medical MicrobiologyRadboud University Medical CenterNijmegenThe Netherlands
- Radboud Center for Infectious DiseasesRadboud University Medical CenterNijmegenThe Netherlands
| | - Robert W. Sauerwein
- Department of Medical MicrobiologyRadboud University Medical CenterNijmegenThe Netherlands
- Radboud Center for Infectious DiseasesRadboud University Medical CenterNijmegenThe Netherlands
| |
Collapse
|
13
|
Powell MD, Read KA, Sreekumar BK, Jones DM, Oestreich KJ. IL-12 signaling drives the differentiation and function of a T H1-derived T FH1-like cell population. Sci Rep 2019; 9:13991. [PMID: 31570752 PMCID: PMC6769002 DOI: 10.1038/s41598-019-50614-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 09/13/2019] [Indexed: 12/15/2022] Open
Abstract
CD4+ T follicular helper (TFH) cells provide help to B cells and promote antibody-mediated immune responses. Increasing evidence supports the existence of TFH populations that secrete cytokines typically associated with the effector functions of other CD4+ T cell subsets. These include T helper 1 (TH1)-biased TFH (TFH1) cells that have recognized roles in both immune responses to pathogens and also the pathogenesis of autoimmune disease. Given their apparent importance to human health, there is interest in understanding the mechanisms that regulate TFH1 cell formation and function. However, their origin and the molecular requirements for their differentiation are unclear. Here, we describe a population of murine TH1-derived, TFH1-like cells that express the chemokine receptor Cxcr3 and produce both the TH1 cytokine interferon-γ and the TFH-associated cytokine interleukin-21 (IL-21). Furthermore, these TFH1-like cells promote B cell activation and antibody production at levels indistinguishable from conventional IL-6-derived TFH-like cells. Regarding their regulatory requirements, we find that IL-12 signaling is necessary for the differentiation and function of this TFH1-like cell population. Specifically, IL-12-dependent activation of STAT4, and unexpectedly STAT3, promotes increased expression of IL-21 and the TFH lineage-defining transcription factor Bcl-6 in TFH1-like cells. Taken together, these findings provide insight into the potential origin and differentiation requirements of TFH1 cells.
Collapse
Affiliation(s)
- Michael D Powell
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Kaitlin A Read
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA.,Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA.,Biomedical and Veterinary Sciences Graduate Program, Virginia Tech, Virginia, USA
| | - Bharath K Sreekumar
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA.,Translational Biology, Medicine, and Health Graduate Program, Virginia Tech, Virginia, USA
| | - Devin M Jones
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA.,Translational Biology, Medicine, and Health Graduate Program, Virginia Tech, Virginia, USA
| | - Kenneth J Oestreich
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA. .,Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA. .,Virginia Tech Carilion School of Medicine, Roanoke, VA, USA.
| |
Collapse
|