1
|
Rollason S, Riley E, Lello J. Stage specific immune responses to schistosomes may explain conflicting results in malaria-schistosome coinfection studies. Infect Dis Model 2025; 10:1003-1018. [PMID: 40520249 PMCID: PMC12159226 DOI: 10.1016/j.idm.2025.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 04/16/2025] [Accepted: 05/19/2025] [Indexed: 06/18/2025] Open
Abstract
Malaria and schistosomiasis are two of the most clinically important human parasitic diseases in terms of morbidity and mortality, collectively causing approximately 800,000 deaths annually. Coinfection with their causative parasites, Plasmodium spp. and Schistosoma spp., is common, particularly in sub-Saharan Africa. These parasites may interact with each other via their effects on the host immune system, but studies to date report conflicting consequences of such interactions, some suggesting that schistosomes are associated with reduced parasitaemia in malaria infection while others report increased parasitaemia. Schistosomes stimulate different immune components in early versus late infection. Using agent-based modelling we explore whether stage of infection could be a factor explaining the conflicting coinfection outcomes. Effects of schistosomes on blood stage malaria were modelled by adjusting the immune components within the model according to the response provoked by each schistosome stage. We find the dynamics of malaria infections are greatly influenced by the stage of schistosomes, with acute and chronic schistosome infections having opposite effects on both peak infected erythrocyte counts and duration. Our findings offer a possible explanation for the apparent contradictions between studies and highlight the importance of considering the stage of schistosome infection when exploring the relationship between these two parasites.
Collapse
Affiliation(s)
| | | | - Joanne Lello
- School of Biosciences, Cardiff University, Cardiff, UK
| |
Collapse
|
2
|
Sala E, Nelli M, Laura C, Di Lucia P, Beccaria CG, Bono EB, Mangione M, Marotta D, Sperto V, Grillo M, Giustini L, Tosi F, Nie J, Kim D, Furiato G, Malpighi C, Consolo E, Becher B, David E, Cohen M, Giladi A, Amit I, Bosselut R, Guidotti LG, Iannacone M, Kuka M. T-cell-derived IFN-γ suppresses T follicular helper cell differentiation and antibody responses. EMBO J 2025; 44:2400-2423. [PMID: 40169810 PMCID: PMC12048687 DOI: 10.1038/s44318-025-00414-3] [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: 09/17/2024] [Revised: 02/23/2025] [Accepted: 03/03/2025] [Indexed: 04/03/2025] Open
Abstract
CD4+ T cells play a critical role in antiviral humoral and cellular immune responses. We have previously reported that subcutaneous lymphocytic choriomeningitis virus (s.c. LCMV) infection is characterized by a stark compartmentalization of CD4+ T cells, leading to strong TH1 cell polarization but virtually absent T follicular helper (TFH) cells, key drivers of humoral immunity. Here, we investigate the mechanisms responsible for this impaired TFH differentiation. We show that T-bet+ cells induced by LCMV infection encompass a TH1 cell subset expressing granzyme B (GzmB), and a Tcf-1+ cell subset that retains the potential for TFH differentiation without expressing mature TFH markers. Notably, IFN-γ blockade enables full differentiation of Tcf-1+ cells into TFH cells, formation of germinal centers, and increased antibody production. Suppression of TFH cells by IFN-γ is not directly mediated by CD4+ T cells but rather involves another cell type, likely dendritic cells (DCs). Our study provides novel insights into the mechanisms underlying early CD4+ T-cell polarization and humoral responses to viruses, with the potential to facilitate the development of effective vaccine strategies.
Collapse
Affiliation(s)
- Eleonora Sala
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria Nelli
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Laura
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Pietro Di Lucia
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Cristian Gabriel Beccaria
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elisa B Bono
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marta Mangione
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Davide Marotta
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Valentina Sperto
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marta Grillo
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Leonardo Giustini
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Fabio Tosi
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Jia Nie
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Daehong Kim
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Giuliana Furiato
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Malpighi
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Eleonora Consolo
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Eyal David
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Merav Cohen
- Department of Clinical Microbiology and Immunology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Amir Giladi
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Remy Bosselut
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Luca G Guidotti
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Matteo Iannacone
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy.
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Experimental Imaging Centre, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Mirela Kuka
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy.
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
3
|
Osada Y, Shimizu S, Morita K. Parasitic helminths and protozoa: Treasure boxes of disease modifying anti-rheumatic drugs. Parasitol Int 2025; 105:103000. [PMID: 39592081 DOI: 10.1016/j.parint.2024.103000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 11/28/2024]
Abstract
Parasites generally survive in their hosts by employing various immunomodulation and immune evasion mechanisms. "helminth therapy" is one strategy that harnesses these parasite-specific beneficial properties for the therapeutic treatment of autoimmune and allergic diseases. Although numerous experimental reports have documented the anti-autoimmune activities of parasitic infections and parasite-derived products, the underlying mechanisms remain insufficiently elucidated due to the significant diversity among parasite species and autoimmune conditions. Rheumatoid arthritis (RA) is one of the most prevalent autoimmune disorders, presenting a substantial opportunity for the therapeutic use of parasites as novel disease-modifying antirheumatic drugs (DMARDs). In this paper, we summarize the immunomodulatory properties of parasites, focusing on their anti-arthritic mechanisms, and discuss the potential of parasite-derived products for the treatment of RA.
Collapse
Affiliation(s)
- Yoshio Osada
- Department of Immunology and Parasitology, University of Occupational and Environmental Health, Japan.
| | - Shoichi Shimizu
- Department of Immunology and Parasitology, University of Occupational and Environmental Health, Japan
| | - Kentaro Morita
- Department of Immunology and Parasitology, University of Occupational and Environmental Health, Japan
| |
Collapse
|
4
|
Du MRM, Wang C, Law CW, Amann-Zalcenstein D, Anttila CJA, Ling L, Hickey PF, Sargeant CJ, Chen Y, Ioannidis LJ, Rajasekhar P, Yip RKH, Rogers KL, Hansen DS, Bowden R, Ritchie ME. Benchmarking spatial transcriptomics technologies with the multi-sample SpatialBenchVisium dataset. Genome Biol 2025; 26:77. [PMID: 40156041 PMCID: PMC11954323 DOI: 10.1186/s13059-025-03543-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 03/13/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Spatial transcriptomics allows gene expression to be measured within complex tissue contexts. Among the array of spatial capture technologies available is 10x Genomics' Visium platform, a popular method which enables transcriptome-wide profiling of tissue sections. Visium offers a range of sample handling and library construction methods which introduces a need for benchmarking to compare data quality and assess how well the technology can recover expected tissue features and biological signatures. RESULTS Here we present SpatialBenchVisium, a unique reference dataset generated from spleen tissue of mice responding to malaria infection spanning several tissue preparation protocols (both fresh frozen and FFPE, with either manual or CytAssist tissue placement). We note better quality control metrics in reference samples prepared using probe-based capture methods, particularly those processed with CytAssist, validating the improvement in data quality produced with the platform. Our analysis of replicate samples extends to explore spatially variable gene detection, the outcomes of clustering and cell deconvolution using matched single-cell RNA-sequencing data and publicly available reference data to identify cell types and tissue regions expected in the spleen. Multi-sample differential expression analysis recovered known gene signatures related to biological sex or gene knockout.
Collapse
Affiliation(s)
- Mei R M Du
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Changqing Wang
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Charity W Law
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Daniela Amann-Zalcenstein
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Casey J A Anttila
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Ling Ling
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Peter F Hickey
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Callum J Sargeant
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Yunshun Chen
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Lisa J Ioannidis
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Pradeep Rajasekhar
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Raymond K H Yip
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Kelly L Rogers
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Diana S Hansen
- Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Rory Bowden
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Matthew E Ritchie
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
5
|
Fusco EM, Bower L, Polidoro R, Minns AM, Lindner SE, Schmidt NW. Microbiome-mediated modulation of immune memory to P. yoelii affects the resistance to secondary cerebral malaria challenge. Immunohorizons 2025; 9:vlaf009. [PMID: 40193560 PMCID: PMC12086675 DOI: 10.1093/immhor/vlaf009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 01/29/2025] [Indexed: 04/09/2025] Open
Abstract
Malaria is caused by protozoan parasites in the genus Plasmodium. Over time individuals slowly develop clinical immunity to malaria, but this process occurs at variable rates, and the mechanism of protection is not fully understood. We have recently demonstrated that in genetically identical C57BL/6N mice, gut microbiota composition dramatically impacts the quality of the humoral immune response to Plasmodium yoelii and subsequent protection against a lethal secondary challenge with Plasmodium berghei ANKA in C57BL/6N mice. Here, we utilize this genetically identical, gut microbiome-dependent model to investigate how the gut microbiota modulate immunological memory, hypothesizing that the gut microbiome impacts the formation and functionality of immune memory. In support of this hypothesis, P. yoelii hyperparasitemia-resistant C57BL/6N mice exhibit increased protection against P. berghei ANKA-induced experimental cerebral malaria (ECM) compared to P. yoelii hyperparasitemia-susceptible C57BL/6N mice. Despite differences in protection against ECM, P. yoelii-resistant and -susceptible mice accumulate similar numbers of memory B cells (MBCs) and memory T cells. Following challenge with P. berghei ANKA, P. yoelii-resistant mice generated more rapid germinal center reactions; however, P. yoelii-resistant and -susceptible mice had similar titers of P. yoelii- and P. berghei-specific antibodies. In contrast, P. yoelii-resistant mice had an increased number of regulatory T cells in response to secondary challenge with P. berghei ANKA, which may dampen the immune-mediated breakdown of the blood-brain barrier and susceptibility to P. berghei-induced ECM. These findings demonstrate the ability of the gut microbiome to shape immune memory and the potential to enhance resistance to severe malaria outcomes.
Collapse
Affiliation(s)
- Elizabeth M Fusco
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Layne Bower
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Rafael Polidoro
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Allen M Minns
- The Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA, United States
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, United States
- The Huck Center for Malaria Research, University Park, PA, United States
| | - Scott E Lindner
- The Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA, United States
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, United States
- The Huck Center for Malaria Research, University Park, PA, United States
| | - Nathan W Schmidt
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
6
|
Hildebrand JA, Daniels NR, Dehm EM, Fisher BD, Guter JK, Janse CJ, Lucas ED, Sangala JA, Tankersley TN, Hart GT, Hamilton SE. Severe malaria enforces short-lived effector cell differentiation but does not prevent effective secondary responses by memory CD8 T cells. PLoS Pathog 2025; 21:e1012993. [PMID: 40163479 PMCID: PMC11957282 DOI: 10.1371/journal.ppat.1012993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 02/18/2025] [Indexed: 04/02/2025] Open
Abstract
Parasitic infections are a major worldwide health burden, yet most studies of CD8 T cell differentiation focus on acute viral and bacterial infections. To understand effector and memory CD8 T cell responses during erythrocytic malaria infection in mice, we utilized transgenic OT-I T cells and compared CD8 T cell responses between infection with OVA-expressing strains of Listeria monocytogenes (Lm) and Plasmodium berghei ANKA (PbA). We find that CD8 T cells expand vigorously during both infections. However, in contrast to Lm infection, PbA infection induces T cells that are heavily biased toward an IL-7Ra-deficient and KLRG1+ short-lived effector cell (SLEC) phenotype at the expense of memory precursor effector cell (MPECs) formation. PbA-induced inflammation, including IFNγ, is partially responsible for this outcome. Following treatment with antimalarial drugs and T cell contraction, PbA-primed memory T cells are rarely found in the blood and peripheral tissues but do maintain a low presence in the spleen and bone marrow. Despite these poor numbers, PbA memory T cells robustly expand upon vaccination or viral infection, control pathogen burden, and form secondary memory pools. Thus, despite PbA enforced SLEC formation and limited memory, effective secondary responses can still proceed.
Collapse
Affiliation(s)
- Jacob A. Hildebrand
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Noah R. Daniels
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Emma M. Dehm
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Benjamin D. Fisher
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Joseph K. Guter
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Chris J. Janse
- Leiden Malaria Research Group, Department of Parasitology, Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Erin D. Lucas
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jules A. Sangala
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Division of Infectious Disease and Internal Medicine, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Trevor N. Tankersley
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Geoffrey T. Hart
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Division of Infectious Disease and Internal Medicine, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Sara E. Hamilton
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
7
|
Kalkal M, Das J. Differential B cell mediated immune response during Plasmodium yoelii infection in mice. Acta Trop 2025; 263:107533. [PMID: 39889895 DOI: 10.1016/j.actatropica.2025.107533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/15/2025] [Accepted: 01/22/2025] [Indexed: 02/03/2025]
Abstract
B lymphocytes are essential components of the humoral immune response and categorized into various subsets according to specific surface markers, functions, and developmental stages. Each subset of B cells plays a distinct role in the immune response, contributing to the overall effectiveness of the immune system. In this study, we investigated the modulation of different splenic subsets of B cells during Plasmodium yoelii infection. Balb/c mice infected with each Plasmodium yoelii XL and Plasmodium yoelii XNL parasite were used for phenotypic characterization of splenic B cell subsets through flow-cytometry. Our findings indicate that both lethal and non-lethal infections of Plasmodium yoelii result in significant alterations within the B cell compartment of the spleen in Balb/c mice during malaria infection. Notably, a differential expansion of immature B cell subsets T1 and T2 was noticed. A continuous reduction in frequency of both subsets (T1 and T2) during infection with lethal parasite while an increase in these subsets during the recovery from infection with non-lethal parasite was observed. Further, the frequencies of mature B cell subsets, follicular B cells and marginal zone B cells, were reduced during lethal infection which may be leading to susceptibility. Whereas non-lethal parasite infection resulted in increased frequency of follicular B cells in spleen which indicates towards establishment of germinal centre for generation of long-term immunity/resistance to infection. This differential expansion of splenic B cell subsets reflects the distinct characteristics of lethal and non-lethal parasite. Overall, these findings illustrate the potential role of B cells in resistance/susceptibility during malaria infection and further enhance our understanding of the B cell mediated immunological aspects of Plasmodium infection.
Collapse
Affiliation(s)
- Meenu Kalkal
- Immunology Division, ICMR-National Institute of Malaria Research, Dwarka, New Delhi 110077, India
| | - Jyoti Das
- Immunology Division, ICMR-National Institute of Malaria Research, Dwarka, New Delhi 110077, India.
| |
Collapse
|
8
|
Zhou Y, Hubscher CH. Biomarker expression level changes within rectal gut-associated lymphoid tissues in spinal cord-injured rats. Immunohorizons 2025; 9:vlaf002. [PMID: 40048710 PMCID: PMC11884801 DOI: 10.1093/immhor/vlaf002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 01/31/2025] [Indexed: 03/09/2025] Open
Abstract
Neurogenic bowel dysfunction (NBD) is common after spinal cord injury (SCI). Gut-associated lymphoid tissue (GALT), an organized structure within the mucosal immune system, is important for the maintenance of gut homeostasis and body health and serves as the first line barrier/defense against diet antigens, commensal microbiota, pathogens, and toxins in mucosal areas. The current study examined gene expression levels along six segments of anorectal tissue using real-time polymerase chain reaction (RT-PCR) in uninjured rats (28-day sham surgical controls) and at both 28- and 42-days post-T9 contusion injury. Consistent with our previous report of functional regional differences in the ano-rectum, we demonstrate the existence of GALTs located primarily within the segment at 3-4.5 cm from the rectal dentate line (termed rectal GALTs-rGALTs) in shams with upregulated gene expression levels of multiple biomarkers, including B cell and T cell-related genes, major histocompatibility complex (MHC) class II molecules, and germinal center (GC)-related genes, which was further confirmed by histologic examination. In the same rectal tissue segment following T9 SCI, inflammation-related genes were upregulated at 28 days post-injury (DPI) indicating that microbial infection and inflammation of rGALTs modified structure and function of rGALTs, while at 42 DPI rGALTs exhibited resolution of inflammation and impaired structure/function for extrafollicular B cell responses. Taken together, our data suggest that rGALTs exists in rat rectum for homeostasis of gut microbiota/barrier. SCI induces microbial infection and inflammation in rectal tissues containing rGALTs, which could contribute to development of SCI-related gut microbiome dysbiosis, NBD, and systemic diseases.
Collapse
Affiliation(s)
- Yun Zhou
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, United States
- Kentucky Spinal Cord Injury Research Center, Louisville, KY, United States
| | - Charles H Hubscher
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, United States
- Kentucky Spinal Cord Injury Research Center, Louisville, KY, United States
| |
Collapse
|
9
|
Dookie RS, Villegas-Mendez A, Cheeseman A, Jones AP, Barroso R, Barrett JR, Draper SJ, Janse CJ, Grogan JL, MacDonald AS, Couper KN. Synergistic blockade of TIGIT and PD-L1 increases type-1 inflammation and improves parasite control during murine blood-stage Plasmodium yoelii non-lethal infection. Infect Immun 2024; 92:e0034524. [PMID: 39324794 PMCID: PMC11556036 DOI: 10.1128/iai.00345-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 09/03/2024] [Indexed: 09/27/2024] Open
Abstract
Pro-inflammatory immune responses are rapidly suppressed during blood-stage malaria but the molecular mechanisms driving this regulation are still incompletely understood. In this study, we show that the co-inhibitory receptors TIGIT and PD-1 are upregulated and co-expressed by antigen-specific CD4+ T cells (ovalbumin-specific OT-II cells) during non-lethal Plasmodium yoelii expressing ovalbumin (PyNL-OVA) blood-stage infection. Synergistic blockade of TIGIT and PD-L1, but not individual blockade of each receptor, during the early stages of infection significantly improved parasite control during the peak stages (days 10-15) of infection. Mechanistically, this protection was correlated with significantly increased plasma levels of IFN-γ, TNF, and IL-2, and an increase in the frequencies of IFN-γ-producing antigen-specific T-bet+ CD4+ T cells (OT-II cells), but not antigen-specific CD8+ T cells (OT-I cells), along with expansion of the splenic red pulp and monocyte-derived macrophage populations. Collectively, our study identifies a novel role for TIGIT in combination with the PD1-PD-L1 axis in regulating specific components of the pro-inflammatory immune response and restricting parasite control during the acute stages of blood-stage PyNL infection.
Collapse
Affiliation(s)
- Rebecca S. Dookie
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Ana Villegas-Mendez
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Antonn Cheeseman
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Adam P. Jones
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Ruben Barroso
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | | | - Simon J. Draper
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Chris J. Janse
- Leiden Malaria Group, Center of Infectious Diseases, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Jane L. Grogan
- Department of Cancer Immunology, Genentech, South San Francisco, California, USA
| | - Andrew S. MacDonald
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Kevin N. Couper
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
10
|
Vaishalli PM, Das R, Cheema HS, Ghosh S, Chandana M, Anand A, Murmu KC, Padmanaban G, Ravindran B, Nagaraj VA. Plasmodium berghei HMGB1 controls the host immune responses and splenic clearance by regulating the expression of pir genes. J Biol Chem 2024; 300:107829. [PMID: 39341498 PMCID: PMC11541847 DOI: 10.1016/j.jbc.2024.107829] [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: 08/14/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 10/01/2024] Open
Abstract
High mobility group box (HMGB) proteins belong to the high mobility group (HMG) superfamily of non-histone nuclear proteins that are involved in chromatin remodeling, regulation of gene expression, and DNA repair. When extracellular, HMGBs serve as alarmins inducing inflammation, and this is attributed to the proinflammatory activity of box B. Here, we show that Plasmodium HMGB1 has key amino acid changes in box B resulting in the loss of TNF-α stimulatory activity. Site-directed mutagenesis of the critical amino acids in box B with respect to mouse HMGB1 renders recombinant Plasmodium berghei (Pb) HMGB1 capable of inducing TNF-α release. Targeted deletion of PbHMGB1 and a detailed in vivo phenotyping show that PbHMGB1 knockout (KO) parasites can undergo asexual stage development. Interestingly, Balb/c mice-infected with PbHMGB1KO parasites display a protective phenotype with subsequent clearance of blood parasitemia and develop long-lasting protective immunity against the challenges performed with Pb wildtype parasites. The characterization of splenic responses shows prominent germinal centers leading to effective humoral responses and enhanced T follicular helper cells. There is also complete protection from experimental cerebral malaria in CBA/CaJ mice susceptible to cerebral pathogenesis with subsequent parasite clearance. Transcriptomic studies suggest the involvement of PbHMGB1 in pir expression. Our findings highlight the gene regulatory function of parasite HMGB1 and its in vivo significance in modulating the host immune responses. Further, clearance of asexual stages in PbHMGB1KO-infected mice underscores the important role of parasite HMGB1 in host immune evasion. These findings have implications in developing attenuated blood-stage vaccines for malaria.
Collapse
Affiliation(s)
- Pradeep Mini Vaishalli
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, India; Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Rahul Das
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, India; Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Harveer Singh Cheema
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, India; Department of Botany, Meerut College, Meerut, Uttar Pradesh, India
| | - Sourav Ghosh
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, India; Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Manjunatha Chandana
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, India
| | - Aditya Anand
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, India; Regional Centre for Biotechnology, Faridabad, Haryana, India
| | | | | | | | | |
Collapse
|
11
|
Hu Y, Yang A, Li H, Zhao R, Bao C, Yu Y, Wang Y, Wang Z, Zhuo L, Han Q, Zhang Z, Zhang J, Zhao H. Lymph node-targeted STING agonist nanovaccine against chronic HBV infection. Cell Mol Life Sci 2024; 81:372. [PMID: 39196331 PMCID: PMC11358573 DOI: 10.1007/s00018-024-05404-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/03/2024] [Accepted: 08/07/2024] [Indexed: 08/29/2024]
Abstract
Chronic hepatitis B virus (HBV) infection is a global health problem that substantially increases the risk of developing liver disease. The development of a novel strategy to induce anti-HB seroconversion and achieve a long-lasting immune response against chronic HBV infection remains challenging. Here, we found that chronic HBV infection affected the signaling pathway involved in STING-mediated induction of host immune responses in dendritic cells (DCs) and then generated a lymph node-targeted nanovaccine that co-delivered hepatitis B surface antigen (HBsAg) and cyclic diguanylate monophosphate (c-di-GMP) (named the PP-SG nanovaccine). The feasibility and efficiency of the PP-SG nanovaccine for CHB treatment were evaluated in HBV-carrier mice. Serum samples were analyzed for HBsAg, anti-HBs, HBV DNA, and alanine aminotransferase levels, and liver samples were evaluated for HBV DNA and RNA and HBcAg, accompanied by an analysis of HBV-specific cellular and humoral immune responses during PP-SG nanovaccine treatment. The PP-SG nanovaccine increased antigen phagocytosis and DC maturation, efficiently and safely eliminated HBV, achieved a long-lasting immune response against HBV reinjection, and disrupted chronic HBV infection-induced immune tolerance, as characterized by the generation and multifunctionality of HBV-specific CD8+ T and CD4+ T cells and the downregulation of immune checkpoint molecules. HBV-carrier mice immunized with the PP-SG nanovaccine achieved partial anti-HBs seroconversion. The PP-SG nanovaccine can induce sufficient and persistent viral suppression and achieve anti-HBs seroconversion, rendering it a promising vaccine candidate for clinical chronic hepatitis B therapy.
Collapse
Affiliation(s)
- Yifei Hu
- Institute of Immunopharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Ailu Yang
- Institute of Immunopharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Hui Li
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Rongrong Zhao
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong Key Laboratory of Brain Function Remodeling, Shandong University, Jinan, Shandong, China
| | - Cuiping Bao
- Institute of Immunopharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yating Yu
- Institute of Immunopharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yucan Wang
- Institute of Immunopharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Zixuan Wang
- Institute of Immunopharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Li Zhuo
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, Shandong University, Qingdao, Shandong, China
| | - Qiuju Han
- Institute of Immunopharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Zhiyue Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jian Zhang
- Institute of Immunopharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| | - Huajun Zhao
- Institute of Immunopharmaceutical Sciences, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
12
|
Araujo S, Mabille D, Garcia AB, Caljon G. A breath of fresh air: impact of insect-borne protozoan parasites on the respiratory system. Trends Parasitol 2024; 40:717-730. [PMID: 39013660 DOI: 10.1016/j.pt.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/17/2024] [Accepted: 06/19/2024] [Indexed: 07/18/2024]
Abstract
The protozoan parasites Plasmodium, Leishmania, and Trypanosoma are transmitted by hematophagous insects and cause severe diseases in humans. These infections pose a global threat, particularly in low-resource settings, and are increasingly extending beyond the current endemic regions. Tropism of parasites is crucial for their development, and recent studies have revealed colonization of noncanonical tissues, aiding their survival and immune evasion. Despite receiving limited attention, cumulative evidence discloses the respiratory system as a significant interface for host-pathogen interactions, influencing the course of (co)infection and disease onset. Due to its pathophysiological and clinical implications, we emphasize that further research is needed to better understand the involvement of the respiratory system and its potential to improve prevention, diagnosis, treatment, and interruption of the chain of transmission.
Collapse
Affiliation(s)
- Sergio Araujo
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium
| | - Dorien Mabille
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium
| | - Alvaro Baeza Garcia
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium.
| |
Collapse
|
13
|
Ng TW, Furuyama W, Wirchnianski AS, Saavedra-Ávila NA, Johndrow CT, Chandran K, Jacobs WR, Marzi A, Porcelli SA. A viral vaccine design harnessing prior BCG immunization confers protection against Ebola virus. Front Immunol 2024; 15:1429909. [PMID: 39081315 PMCID: PMC11286471 DOI: 10.3389/fimmu.2024.1429909] [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: 05/08/2024] [Accepted: 06/25/2024] [Indexed: 08/02/2024] Open
Abstract
Previous studies have demonstrated the efficacy and feasibility of an anti-viral vaccine strategy that takes advantage of pre-existing CD4+ helper T (Th) cells induced by Mycobacterium bovis bacille Calmette-Guérin (BCG) vaccination. This strategy uses immunization with recombinant fusion proteins comprised of a cell surface expressed viral antigen, such as a viral envelope glycoprotein, engineered to contain well-defined BCG Th cell epitopes, thus rapidly recruiting Th cells induced by prior BCG vaccination to provide intrastructural help to virus-specific B cells. In the current study, we show that Th cells induced by BCG were localized predominantly outside of germinal centers and promoted antibody class switching to isotypes characterized by strong Fc receptor interactions and effector functions. Furthermore, BCG vaccination also upregulated FcγR expression to potentially maximize antibody-dependent effector activities. Using a mouse model of Ebola virus (EBOV) infection, this vaccine strategy provided sustained antibody levels with strong IgG2c bias and protection against lethal challenge. This general approach can be easily adapted to other viruses, and may be a rapid and effective method of immunization against emerging pandemics in populations that routinely receive BCG vaccination.
Collapse
Affiliation(s)
- Tony W. Ng
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Wakako Furuyama
- Laboratory of Virology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institute of Health, Hamilton, MT, United States
| | - Ariel S. Wirchnianski
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Noemí A. Saavedra-Ávila
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Christopher T. Johndrow
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - William R. Jacobs
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Andrea Marzi
- Laboratory of Virology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institute of Health, Hamilton, MT, United States
| | - Steven A. Porcelli
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
14
|
Cheng Q, Yang X, Zou T, Sun L, Zhang X, Deng L, Wu M, Gai W, Jiang H, Guo T, Lu Y, Dong J, Niu C, Pan W, Zhang J. RACK1 enhances STAT3 stability and promotes T follicular helper cell development and function during blood-stage Plasmodium infection in mice. PLoS Pathog 2024; 20:e1012352. [PMID: 39024388 PMCID: PMC11288429 DOI: 10.1371/journal.ppat.1012352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 07/30/2024] [Accepted: 06/19/2024] [Indexed: 07/20/2024] Open
Abstract
CD4+ T cells are central mediators of protective immunity to blood-stage malaria, particularly for their capacity in orchestrating germinal center reaction and generating parasite-specific high-affinity antibodies. T follicular helper (Tfh) cells are predominant CD4+ effector T cell subset implicated in these processes, yet the factors and detailed mechanisms that assist Tfh cell development and function during Plasmodium infection are largely undefined. Here we provide evidence that receptor for activated C kinase 1 (RACK1), an adaptor protein of various intracellular signals, is not only important for CD4+ T cell expansion as previously implied but also plays a prominent role in Tfh cell differentiation and function during blood-stage Plasmodium yoelii 17XNL infection. Consequently, RACK1 in CD4+ T cells contributes significantly to germinal center formation, parasite-specific IgG production, and host resistance to the infection. Mechanistic exploration detects specific interaction of RACK1 with STAT3 in P. yoelii 17XNL-responsive CD4+ T cells, ablation of RACK1 leads to defective STAT3 phosphorylation, accompanied by substantially lower amount of STAT3 protein in CD4+ T cells, whereas retroviral overexpression of RACK1 or STAT3 in RACK1-deficient CD4+ T cells greatly restores STAT3 activity and Bcl-6 expression under the Tfh polarization condition. Further analyses suggest RACK1 positively regulates STAT3 stability by inhibiting the ubiquitin-proteasomal degradation process, thus promoting optimal STAT3 activity and Bcl-6 induction during Tfh cell differentiation. These findings uncover a novel mechanism by which RACK1 participates in posttranslational regulation of STAT3, Tfh cell differentiation, and subsequent development of anti-Plasmodium humoral immunity.
Collapse
Affiliation(s)
- Qianqian Cheng
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Xiqin Yang
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Tao Zou
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Lin Sun
- Shanghai Immune Therapy Institute, Shanghai Jiao Tong University School of Medicine-Affiliated Renji Hospital, Shanghai, China
| | - Xueting Zhang
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Lijiao Deng
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Mengyao Wu
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Wenbin Gai
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Hui Jiang
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Tingting Guo
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Yuchen Lu
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Jie Dong
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Chunxiao Niu
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Weiqing Pan
- Department of Tropical Diseases, Navy Medical University, Shanghai, China
| | - Jiyan Zhang
- Beijing Institute of Basic Medical Sciences, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
| |
Collapse
|
15
|
Lu F, Xu J, Liu Y, Ren Z, Chen J, Gong W, Yin Y, Li Y, Qian L, He X, Han X, Lin Z, Lu J, Zhang W, Liu J, Menard D, Han ET, Cao J. Plasmodium vivax serological exposure markers: PvMSP1-42-induced humoral and memory B-cell response generates long-lived antibodies. PLoS Pathog 2024; 20:e1012334. [PMID: 38941356 PMCID: PMC11239109 DOI: 10.1371/journal.ppat.1012334] [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: 01/23/2024] [Revised: 07/11/2024] [Accepted: 06/10/2024] [Indexed: 06/30/2024] Open
Abstract
Plasmodium vivax serological exposure markers (SEMs) have emerged as promising tools for the actionable surveillance and implementation of targeted interventions to accelerate malaria elimination. To determine the dynamic profiles of SEMs in current and past P. vivax infections, we screened and selected 11 P. vivax proteins from 210 putative proteins using protein arrays, with a set of serum samples obtained from patients with acute P. vivax and documented past P. vivax infections. Then we used a murine protein immune model to initially investigate the humoral and memory B cell response involved in the generation of long-lived antibodies. We show that of the 11 proteins, especially C-terminal 42-kDa region of P. vivax merozoite surface protein 1 (PvMSP1-42) induced longer-lasting long-lived antibodies, as these antibodies were detected in individuals infected with P. vivax in the 1960-1970s who were not re-infected until 2012. In addition, we provide a potential mechanism for the maintenance of long-lived antibodies after the induction of PvMSP1-42. The results indicate that PvMSP1-42 induces more CD73+CD80+ memory B cells (MBCs) compared to P. vivax GPI-anchored micronemal antigen (PvGAMA), allowing IgG anti-PvMSP1-42 antibodies to be maintained for a long time.
Collapse
Affiliation(s)
- Feng Lu
- Department of Pathogenic Biology and Immunology, School of Medicine, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Jiahui Xu
- Department of Pathogenic Biology and Immunology, School of Medicine, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 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, China
| | - Zhenyu Ren
- Department of Pathogenic Biology and Immunology, School of Medicine, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Junhu Chen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China
| | - Weijuan Gong
- Department of Pathogenic Biology and Immunology, School of Medicine, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Yi Yin
- Department of Pathogenic Biology and Immunology, School of Medicine, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Yinyue Li
- Department of Pathogenic Biology and Immunology, School of Medicine, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Li Qian
- Department of Pathogenic Biology and Immunology, School of Medicine, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Xinlong He
- Department of Pathogenic Biology and Immunology, School of Medicine, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Xiu Han
- Department of Pathogenic Biology and Immunology, School of Medicine, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Zhijie Lin
- Department of Pathogenic Biology and Immunology, School of Medicine, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Jingyuan Lu
- Department of Pathogenic Biology and Immunology, School of Medicine, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Wenwen Zhang
- Department of Pathogenic Biology and Immunology, School of Medicine, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Jiali Liu
- Department of Pathogenic Biology and Immunology, School of Medicine, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Didier Menard
- Institut Pasteur, Université Paris Cité, Malaria Parasite Biology and Vaccines Unit, Paris, France
- Université de Strasbourg, UR 3073—Pathogens Host Arthropods Vectors Interactions Unit, Malaria Genetics and Resistance Team (MEGATEAM), Strasbourg, France
- CHU Strasbourg, Laboratory of Parasitology and Medical Mycology, Strasbourg, France
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, 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, China
| |
Collapse
|
16
|
Osada Y, Shimizu S, Morita K, Gaballah EM, Wu Z, Maekawa Y. Helminth-induced impairment of humoral immunity differently contribute to their anti-arthritic effects in mice: Comparison of Schistosoma mansoni and Trichinella spiralis. Exp Parasitol 2024; 261:108752. [PMID: 38604301 DOI: 10.1016/j.exppara.2024.108752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024]
Abstract
AIMS We have previously reported reduction of anti-type II collagen (IIC) IgG levels in collagen-induced arthritis (CIA) by Schistosoma mansoni (Sm) and Trichinella spiralis (Ts). To clarify the contribution of the impairment of humoral immunity to their anti-arthritic activities, we herein investigated the relationship between anti-IIC IgG levels and arthritic swelling in Sm- or Ts-infected mice. METHODS AND RESULTS Male DBA/1J mice were infected with Sm cercariae or Ts muscle larvae prior to the IIC immunization. In the Sm-infected mice, paw swelling and anti-IIC IgG levels were continuously lower than those of non-infected control group. In contrast, arthritic swelling in the Ts-infected mice only decreased in the early phase of CIA progression, despite the continued impairment of anti-IIC IgG production throughout the experimental period. Correlation coefficients between residual paw swelling and anti-IIC IgG titers were similar or higher in the Sm group than in the control group, but were similar or lower in the Ts group than in the control group. CONCLUSION The down-modulations of anti-IIC IgG levels by the two parasitic infections and the correlation analyses suggest that the anti-arthritic activity of Sm was primarily attributed to the modulation of IgG-independent arthritogenic mechanisms and secondarily to the impairment of anti-IIC IgG production. In contrast, Ts could alleviate CIA mainly via the impairment of antibody production.
Collapse
Affiliation(s)
- Yoshio Osada
- Department of Immunology and Parasitology, University of Occupational and Environmental Health, Japan, 1-1, Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan.
| | - Shoichi Shimizu
- Department of Immunology and Parasitology, University of Occupational and Environmental Health, Japan, 1-1, Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Kentaro Morita
- Department of Immunology and Parasitology, University of Occupational and Environmental Health, Japan, 1-1, Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan
| | - Eman M Gaballah
- Department of Immunology and Parasitology, University of Occupational and Environmental Health, Japan, 1-1, Iseigaoka, Yahatanishi-ku, Kitakyushu, 807-8555, Japan; Department of Medical Parasitology, Faculty of Medicine, Damietta University, Damietta, Egypt
| | - Zhiliang Wu
- Department of Parasitology and Infectious Diseases, Gifu University Graduate School of Medicine, 1-1, Yanagito, Gifu, 501-1194, Japan
| | - Yoichi Maekawa
- Department of Parasitology and Infectious Diseases, Gifu University Graduate School of Medicine, 1-1, Yanagito, Gifu, 501-1194, Japan; Division of Preemptive Food Research, Preemptive Food Research Center (PFRC), Gifu University Institute for Advanced Study (GUiAS), 1-1 Yanagido, Gifu, 501-1193, Japan; Division of Animal Medical Science, Center for One Medicine Innovative Translational Research (COMIT), Gifu University Institute for Advanced Study (GUiAS), 1-1 Yanagido, Gifu, 501-1193, Japan
| |
Collapse
|
17
|
Ng TW, Furuyama W, Wirchnianski AS, Saavedra-Ávila NA, Johndrow CT, Chandran K, Jacobs WR, Marzi A, Porcelli SA. A viral vaccine design harnessing prior BCG immunization confers protection against Ebola virus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.28.595735. [PMID: 38853867 PMCID: PMC11160617 DOI: 10.1101/2024.05.28.595735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Previous studies have demonstrated the efficacy and feasibility of an anti-viral vaccine strategy that takes advantage of pre-existing CD4 + helper T (Th) cells induced by Mycobacterium bovis bacille Calmette-Guérin (BCG) vaccination. This strategy uses immunization with recombinant fusion proteins comprised of a cell surface expressed viral antigen, such as a viral envelope glycoprotein, engineered to contain well-defined BCG Th cell epitopes, thus rapidly recruiting Th cells induced by prior BCG vaccination to provide intrastructural help to virus-specific B cells. In the current study, we show that Th cells induced by BCG were localized predominantly outside of germinal centers and promoted antibody class switching to isotypes characterized by strong Fc receptor interactions and effector functions. Furthermore, BCG vaccination also upregulated FcγR expression to potentially maximize antibody-dependent effector activities. Using a mouse model of Ebola virus (EBOV) infection, this vaccine strategy provided sustained antibody levels with strong IgG2c bias and protection against lethal challenge. This general approach can be easily adapted to other viruses, and may be a rapid and effective method of immunization against emerging pandemics in populations that routinely receive BCG vaccination.
Collapse
|
18
|
Edner NM, Houghton LP, Ntavli E, Rees-Spear C, Petersone L, Wang C, Fabri A, Elfaki Y, Rueda Gonzalez A, Brown R, Kisand K, Peterson P, McCoy LE, Walker LSK. TIGIT +Tfh show poor B-helper function and negatively correlate with SARS-CoV-2 antibody titre. Front Immunol 2024; 15:1395684. [PMID: 38868776 PMCID: PMC11167088 DOI: 10.3389/fimmu.2024.1395684] [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: 03/04/2024] [Accepted: 05/15/2024] [Indexed: 06/14/2024] Open
Abstract
Circulating follicular helper T cells (cTfh) can show phenotypic alterations in disease settings, including in the context of tissue-damaging autoimmune or anti-viral responses. Using severe COVID-19 as a paradigm of immune dysregulation, we have explored how cTfh phenotype relates to the titre and quality of antibody responses. Severe disease was associated with higher titres of neutralising S1 IgG and evidence of increased T cell activation. ICOS, CD38 and HLA-DR expressing cTfh correlated with serum S1 IgG titres and neutralising strength, and interestingly expression of TIGIT by cTfh showed a negative correlation. TIGIT+cTfh expressed increased IFNγ and decreased IL-17 compared to their TIGIT-cTfh counterparts, and showed reduced capacity to help B cells in vitro. Additionally, TIGIT+cTfh expressed lower levels of CD40L than TIGIT-cTfh, providing a potential explanation for their poor B-helper function. These data identify phenotypic changes in polyclonal cTfh that correlate with specific antibody responses and reveal TIGIT as a marker of cTfh with altered function.
Collapse
Affiliation(s)
- Natalie M. Edner
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Luke P. Houghton
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Elisavet Ntavli
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Chloe Rees-Spear
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Lina Petersone
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Chunjing Wang
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Astrid Fabri
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Yassin Elfaki
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Andrea Rueda Gonzalez
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Rachel Brown
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
- Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Kai Kisand
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Pärt Peterson
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Laura E. McCoy
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Lucy S. K. Walker
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| |
Collapse
|
19
|
Qi R, Fu R, Lei X, He J, Jiang Y, Zhang L, Wu Y, Wang S, Guo X, Chen F, Nie M, Yang M, Chen Y, Zeng J, Xu J, Xiong H, Fang M, Que Y, Yao Y, Wang Y, Cao J, Ye H, Zhang Y, Zheng Z, Cheng T, Zhang J, Lin X, Yuan Q, Zhang T, Xia N. Therapeutic vaccine-induced plasma cell differentiation is defective in the presence of persistently high HBsAg levels. J Hepatol 2024; 80:714-729. [PMID: 38336348 DOI: 10.1016/j.jhep.2023.12.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 12/15/2023] [Accepted: 12/29/2023] [Indexed: 02/12/2024]
Abstract
BACKGROUND & AIMS Mechanisms behind the impaired response of antigen-specific B cells to therapeutic vaccination in chronic hepatitis B virus (HBV) infection remain unclear. The development of vaccines or strategies to overcome this obstacle is vital for advancing the management of chronic hepatitis B. METHODS A mouse model, denominated as E6F6-B, was engineered to feature a knock-in of a B-cell receptor (BCR) that specifically recognizes HBsAg. This model served as a valuable tool for investigating the temporal and spatial dynamics of humoral responses following therapeutic vaccination under continuous antigen exposure. Using a suite of immunological techniques, we elucidated the differentiation trajectory of HBsAg-specific B cells post-therapeutic vaccination in HBV carrier mice. RESULTS Utilizing the E6F6-B transfer model, we observed a marked decline in antibody-secreting cells 2 weeks after vaccination. A dysfunctional and atypical pre-plasma cell population (BLIMP-1+ IRF4+ CD40- CD138- BCMA-) emerged, manifested by sustained BCR signaling. By deploying an antibody to purge persistent HBsAg, we effectively prompted the therapeutic vaccine to provoke conventional plasma cell differentiation. This resulted in an enhanced anti-HBs antibody response and facilitated HBsAg clearance. CONCLUSIONS Sustained high levels of HBsAg limit the ability of therapeutic hepatitis B vaccines to induce the canonical plasma cell differentiation necessary for anti-HBs antibody production. Employing a strategy combining antibodies with vaccines can surmount this altered humoral response associated with atypical pre-plasma cells, leading to improved therapeutic efficacy in HBV carrier mice. IMPACT AND IMPLICATIONS Therapeutic vaccines aimed at combatting HBV encounter suboptimal humoral responses in clinical settings, and the mechanisms impeding their effectiveness have remained obscure. Our research, utilizing the innovative E6F6-B mouse transfer model, reveals that the persistence of HBsAg can lead to the emergence of an atypical pre-plasma cell population, which proves to be relevant to the potency of therapeutic HBV vaccines. Targeting the aberrant differentiation process of these atypical pre-plasma cells stands out as a critical strategy to amplify the humoral response elicited by HBV therapeutic vaccines in carrier mouse models. This discovery suggests a compelling avenue for further study in the context of human chronic hepatitis B. Encouragingly, our findings indicate that synergistic therapy combining HBV-specific antibodies with vaccines offers a promising approach that could significantly advance the pursuit of a functional cure for HBV.
Collapse
Affiliation(s)
- Ruoyao Qi
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Rao Fu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Xing Lei
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Jinhang He
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Yao Jiang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Liang Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Yangtao Wu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Siling Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Xueran Guo
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Feng Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Meifeng Nie
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Man Yang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Yiyi Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Jing Zeng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China; Department of clinical laboratory, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Jingjing Xu
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Hualong Xiong
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Mujin Fang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Yuqiong Que
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Youliang Yao
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Yingbin Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Jiali Cao
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China; Department of clinical laboratory, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Huiming Ye
- Department of clinical laboratory, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Yali Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Zizheng Zheng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Tong Cheng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Jun Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China
| | - Xu Lin
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China.
| | - Quan Yuan
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China.
| | - Tianying Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China.
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health and School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, NMPA Key Laboratory for Research and Evaluation of Infectious Disease Diagnostic Technology, Xiamen University, Xiamen 361102, Fujian, China.
| |
Collapse
|
20
|
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
|
21
|
DiToro D, Murakami N, Pillai S. T-B Collaboration in Autoimmunity, Infection, and Transplantation. Transplantation 2024; 108:386-398. [PMID: 37314442 PMCID: PMC11345790 DOI: 10.1097/tp.0000000000004671] [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] [Indexed: 06/15/2023]
Abstract
We have attempted here to provide an up-to-date review of the collaboration between helper T cells and B cells in response to protein and glycoprotein antigens. This collaboration is essential as it not only protects from many pathogens but also contributes to a litany of autoimmune and immune-mediated diseases.
Collapse
Affiliation(s)
- Daniel DiToro
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA
| | - Naoka Murakami
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Shiv Pillai
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, MA
| |
Collapse
|
22
|
Kimenyi KM, Akinyi MY, Mwikali K, Gilmore T, Mwangi S, Omer E, Gichuki B, Wambua J, Njunge J, Obiero G, Bejon P, Langhorne J, Abdi A, Ochola-Oyier LI. Distinct transcriptomic signatures define febrile malaria depending on initial infective states, asymptomatic or uninfected. BMC Infect Dis 2024; 24:140. [PMID: 38287287 PMCID: PMC10823747 DOI: 10.1186/s12879-024-08973-2] [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: 09/21/2023] [Accepted: 01/01/2024] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND Cumulative malaria parasite exposure in endemic regions often results in the acquisition of partial immunity and asymptomatic infections. There is limited information on how host-parasite interactions mediate the maintenance of chronic symptomless infections that sustain malaria transmission. METHODS Here, we determined the gene expression profiles of the parasite population and the corresponding host peripheral blood mononuclear cells (PBMCs) from 21 children (< 15 years). We compared children who were defined as uninfected, asymptomatic and those with febrile malaria. RESULTS Children with asymptomatic infections had a parasite transcriptional profile characterized by a bias toward trophozoite stage (~ 12 h-post invasion) parasites and low parasite levels, while early ring stage parasites were characteristic of febrile malaria. The host response of asymptomatic children was characterized by downregulated transcription of genes associated with inflammatory responses, compared with children with febrile malaria,. Interestingly, the host responses during febrile infections that followed an asymptomatic infection featured stronger inflammatory responses, whereas the febrile host responses from previously uninfected children featured increased humoral immune responses. CONCLUSIONS The priming effect of prior asymptomatic infection may explain the blunted acquisition of antibody responses seen to malaria antigens following natural exposure or vaccination in malaria endemic areas.
Collapse
Affiliation(s)
- Kelvin M Kimenyi
- KEMRI‑Wellcome Trust Research Programme, Kilifi, Kenya
- Department of Biochemistry, University of Nairobi, Nairobi, Kenya
| | | | - Kioko Mwikali
- KEMRI‑Wellcome Trust Research Programme, Kilifi, Kenya
| | | | - Shaban Mwangi
- KEMRI‑Wellcome Trust Research Programme, Kilifi, Kenya
| | - Elisha Omer
- KEMRI‑Wellcome Trust Research Programme, Kilifi, Kenya
| | | | | | - James Njunge
- KEMRI‑Wellcome Trust Research Programme, Kilifi, Kenya
| | - George Obiero
- Department of Biochemistry, University of Nairobi, Nairobi, Kenya
| | - Philip Bejon
- KEMRI‑Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | | | | |
Collapse
|
23
|
Sakai Y, Mura S, Kuwabara Y, Kagimoto S, Sakurai M, Morimoto M, Park ES, Shimojima M, Nagata N, Ami Y, Yoshikawa T, Iwata-Yoshikawa N, Fukushi S, Watanabe S, Kurosu T, Okutani A, Kimura M, Imaoka K, Saijo M, Morikawa S, Suzuki T, Maeda K. Lethal severe fever with thrombocytopenia syndrome virus infection causes systemic germinal centre failure and massive T cell apoptosis in cats. Front Microbiol 2024; 14:1333946. [PMID: 38249467 PMCID: PMC10796997 DOI: 10.3389/fmicb.2023.1333946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 12/19/2023] [Indexed: 01/23/2024] Open
Abstract
Introduction Severe fever with thrombocytopenia syndrome (SFTS) is a fatal viral disease characterized by high fever, thrombocytopenia, leukopenia, and multi-organ haemorrhage. Disruption of the humoral immune response and decreased lymphocyte numbers are thought to contribute to the disease severity. These findings have been obtained through the analysis of peripheral blood leukocytes in human patients, whereas analysis of lymph nodes has been limited. Thus, in this study, we characterized the germinal centre response and apoptosis in the lymph nodes of cats with fatal SFTS, because SFTS in cats well mimics the pathology of human SFTS. Methods Lymph node tissue sections collected during necropsy from seven fatal SFTS patients and five non-SFTS cases were used for histopathological analysis. Additionally, lymph node tissue sections collected from cats with experimental infection of SFTS virus (SFTSV) were also analysed. Results In the lymphoid follicles of cats with SFTS, a drastic decrease in Bcl6- and Ki67-positive germinal centre B cells was observed. Together, the number of T cells in the follicles was also decreased in SFTS cases. In the paracortex, a marked increase in cleaved-caspase3 positivity was observed in T cells. These changes were independent of the number of local SFTS virus-positive cell. Furthermore, the analysis of cats with experimental SFTSV infection revealed that the intrafollicular Bcl6- and CD3-positive cell numbers in cats with low anti-SFTSV antibody production were significantly lower than those in cats with high anti-SFTSV antibody production. Discussion These results suggest that dysfunction of the humoral response in severe SFTS was caused by the loss of germinal centre formation and massive apoptosis of T cells in the lymph nodes due to systemically circulating viruses.
Collapse
Affiliation(s)
- Yusuke Sakai
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Serina Mura
- Laboratory of Veterinary Pathology, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Yuko Kuwabara
- Laboratory of Veterinary Pathology, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Saya Kagimoto
- Laboratory of Veterinary Pathology, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Masashi Sakurai
- Laboratory of Veterinary Pathology, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Masahiro Morimoto
- Laboratory of Veterinary Pathology, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Eun-sil Park
- Department of Veterinary Science, National Institute of Infectious Diseases, Tokyo, Japan
| | - Masayuki Shimojima
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Noriyo Nagata
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yasushi Ami
- Management Department of Biosafety and Laboratory Animal, National Institute of Infectious Diseases, Tokyo, Japan
| | - Tomoki Yoshikawa
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | | | - Shuetsu Fukushi
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Shumpei Watanabe
- Faculty of Veterinary Medicine, Okayama University of Science, Ehime, Japan
| | - Takeshi Kurosu
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Akiko Okutani
- Department of Veterinary Science, National Institute of Infectious Diseases, Tokyo, Japan
| | - Masanobu Kimura
- Department of Veterinary Science, National Institute of Infectious Diseases, Tokyo, Japan
| | - Koichi Imaoka
- Department of Veterinary Science, National Institute of Infectious Diseases, Tokyo, Japan
| | - Masayuki Saijo
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Shigeru Morikawa
- Faculty of Veterinary Medicine, Okayama University of Science, Ehime, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Ken Maeda
- Department of Veterinary Science, National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|
24
|
Furtado R, Paul M, Zhang J, Sung J, Karell P, Kim RS, Caillat-Zucman S, Liang L, Felgner P, Bauleni A, Gama S, Buchwald A, Taylor T, Seydel K, Laufer M, Delahaye F, Daily JP, Lauvau G. Cytolytic circumsporozoite-specific memory CD4 + T cell clones are expanded during Plasmodium falciparum infection. Nat Commun 2023; 14:7726. [PMID: 38001069 PMCID: PMC10673885 DOI: 10.1038/s41467-023-43376-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Clinical immunity against Plasmodium falciparum infection develops in residents of malaria endemic regions, manifesting in reduced clinical symptoms during infection and in protection against severe disease but the mechanisms are not fully understood. Here, we compare the cellular and humoral immune response of clinically immune (0-1 episode over 18 months) and susceptible (at least 3 episodes) during a mild episode of Pf malaria infection in a malaria endemic region of Malawi, by analysing peripheral blood samples using high dimensional mass cytometry (CyTOF), spectral flow cytometry and single-cell transcriptomic analyses. In the clinically immune, we find increased proportions of circulating follicular helper T cells and classical monocytes, while the humoral immune response shows characteristic age-related differences in the protected. Presence of memory CD4+ T cell clones with a strong cytolytic ZEB2+ T helper 1 effector signature, sharing identical T cell receptor clonotypes and recognizing the Pf-derived circumsporozoite protein (CSP) antigen are found in the blood of the Pf-infected participants gaining protection. Moreover, in clinically protected participants, ZEB2+ memory CD4+ T cells express lower level of inhibitory and chemotactic receptors. We thus propose that clonally expanded ZEB2+ CSP-specific cytolytic memory CD4+ Th1 cells may contribute to clinical immunity against the sporozoite and liver-stage Pf malaria.
Collapse
Affiliation(s)
- Raquel Furtado
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
- RF: BioNTech US, 40 Erie Street, Cambridge, MA, 02139, USA
| | - Mahinder Paul
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Jinghang Zhang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Joowhan Sung
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Paul Karell
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Ryung S Kim
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
| | - Sophie Caillat-Zucman
- Université de Paris, AP-HP, Hôpital Saint-Louis, Laboratoire d'Immunologie et Histocompatiblité, INSERM UMR976, 75010, Paris, France
| | - Li Liang
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Philip Felgner
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Andy Bauleni
- Malaria Alert Centre, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Syze Gama
- Blantyre Malaria Project, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Andrea Buchwald
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Terrie Taylor
- Blantyre Malaria Project, Kamuzu University of Health Sciences, Blantyre, Malawi
- Department of Osteopathic Medical Specialties, Michigan State University, East Lansing, MI, 48824, USA
| | - Karl Seydel
- Blantyre Malaria Project, Kamuzu University of Health Sciences, Blantyre, Malawi
- Department of Osteopathic Medical Specialties, Michigan State University, East Lansing, MI, 48824, USA
| | - Miriam Laufer
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Fabien Delahaye
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, 10461, USA
- FD: Precision Oncology, Sanofi, Vitry sur Seine, France
| | - Johanna P Daily
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, 10461, USA.
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, 10461, USA.
| | - Grégoire Lauvau
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, 10461, USA.
| |
Collapse
|
25
|
Tao Q, Du JX, Zhang S, Lin W, Luo Y, Liu Y, Zeng J, Chen XL. Longitudinal multi-functional analysis identified responses of T cells, B cells, and monocytes as hallmarks of immunotherapy tolerance in patients with merkel cell carcinoma. PLoS One 2023; 18:e0293922. [PMID: 37983224 PMCID: PMC10659156 DOI: 10.1371/journal.pone.0293922] [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: 04/13/2023] [Accepted: 10/21/2023] [Indexed: 11/22/2023] Open
Abstract
PURPOSE Merkel cell carcinoma (MCC) is a neuroendocrine carcinoma originating in the skin. Studies are needed to determine the mechanisms of immune escape in patients with MCC, and malignant cell conditions that promote immune evasion. METHODS We used Single-cell RNA sequencing (scRNA-seq) to determine cellular features associated with MCC disease trajectory. A longitudinal multi-omics study was performed using scRNA-seq data of peripheral blood harvested from four-time points. Six major cell types and fifteen cell subgroups were identified and confirmed their presence by expression of characteristic markers. The expression patterns and specific changes of different cells at different time points were investigated. Subsequently, bulk RNA data was used to validate key findings. RESULTS The dynamic characteristics of the cells were identified during the critical period between benign improvement and acquisition of resistance. Combined with the results of the validation cohort, the resistance program expressed in the relapse stage is mainly associated with T cell exhaustion and immune cell crosstalk disorder. Coinciding with immune escape, we also identified a decrease non-classical monocytes and an expansion of classical monocytes with features of high inflammation and immune deficiency. CONCLUSION Changes in cellular status, such as depletion of T cells and dysregulation of B cell proliferation and differentiation, may lead to drug resistance in MCC patients. Meanwhile, the widespread decreased antigen presentation ability and immune disorders caused by deletion of MHC class II gene expression should not be ignored.
Collapse
Affiliation(s)
- Quyuan Tao
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jia-xin Du
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shijing Zhang
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Wenjia Lin
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yongxin Luo
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Ying Liu
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jingyan Zeng
- Shenzhen Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xin-lin Chen
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
26
|
O'Neal KA, Zeltner SL, Foscue CL, Stumhofer JS. Bhlhe40 limits early IL-10 production from CD4 + T cells during Plasmodium yoelii 17X infection. Infect Immun 2023; 91:e0036723. [PMID: 37843306 PMCID: PMC10652903 DOI: 10.1128/iai.00367-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
The cytokine IL-10 suppresses T-cell-mediated immunity, which is required to control infection with Plasmodium yoelii. Consequently, IL-10 can delay the time needed to resolve this infection, leading to a higher parasite burden. While the pathways that lead to IL-10 production by CD4+ T cells are well defined, much less is known about the mediators that suppress the expression of this potent anti-inflammatory cytokine. Here, we show that the transcription factor basic helix-loop-helix family member e40 (Bhlhe40) contributes to controlling parasite burden in response to P. yoelii infection in mice. Loss of Bhlhe40 expression in mice results in higher Il10 expression, higher peak parasitemia, and a delay in parasite clearance. The observed phenotype was not due to defects in T-cell activation and proliferation or the humoral response. Nor was it due to changes in regulatory T-cell numbers. However, blocking IL-10 signaling reversed the outcome in Bhlhe40-/ - mice, suggesting that excess IL-10 production limits their ability to control the infection properly. In addition to suppressing Il10 expression in CD4+ T cells, Bhlhe40 can promote Ifng expression. Indeed, IFN-γ production by CD4+ T cells isolated from the liver was significantly affected by the loss of Bhlhe40. Lastly, Bhlhe40 deletion in T cells resulted in a phenotype similar to that observed in the Bhlhe40-/ - mice, indicating that Bhlhe40 expression in T cells contributes to the ability of mice to control infection with P. yoelii.
Collapse
Affiliation(s)
- Kara A. O'Neal
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Sheldon L. Zeltner
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Camille L. Foscue
- 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
|
27
|
Garner-Spitzer E, Wagner A, Gudipati V, Schoetta AM, Orola-Taus M, Kundi M, Kunert R, Mayrhofer P, Huppa JB, Stockinger H, Carsetti R, Gattinger P, Valenta R, Kratzer B, Sehgal ANA, Pickl WF, Reinisch W, Novacek G, Wiedermann U. Lower magnitude and faster waning of antibody responses to SARS-CoV-2 vaccination in anti-TNF-α-treated IBD patients are linked to lack of activation and expansion of cTfh1 cells and impaired B memory cell formation. EBioMedicine 2023; 96:104788. [PMID: 37672867 PMCID: PMC10485594 DOI: 10.1016/j.ebiom.2023.104788] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 08/08/2023] [Accepted: 08/22/2023] [Indexed: 09/08/2023] Open
Abstract
BACKGROUND Patients with inflammatory bowel disease (IBD) and healthy controls received primary SARS-CoV-2-mRNA vaccination and a booster after six months. Anti-TNF-α-treated patients showed significantly lower antibody (Ab) levels and faster waning than α4β7-integrin-antagonist recipients and controls. This prospective cohort study aimed to elucidate the underlying mechanisms on the basis of circulating T-follicular helper cells (cTfh) and B memory cells. METHODS We measured SARS-CoV-2- Wuhan and Omicron specific Abs, B- and T-cell subsets at baseline and kinetics of Spike (S)-specific B memory cells along with distributions of activated cTfh subsets before and after primary and booster vaccination. FINDINGS Lower and faster waning of Ab levels in anti-TNF-α treated IBD patients was associated with low numbers of total and naïve B cells vs. expanded plasmablasts prior to vaccination. Along with their low Ab levels against Wuhan and Omicron VOCs, reduced S-specific B memory cells were identified after the 2nd dose which declined to non-detectable after 6 months. In contrast, IBD patients with α4β7-integrin-antagonists and controls mounted and retained high Ab levels after the 2nd dose, which was associated with a pronounced increase in S-specific B memory cells that were maintained or expanded up to 6 months. Booster vaccination led to a strong increase of Abs with neutralizing capacity and S-specific B memory cells in these groups, which was not the case in anti-TNF-α treated IBD patients. Of note, Ab levels and S-specific B memory cells in particular post-booster correlated with the activation of cTfh1 cells after primary vaccination. INTERPRETATIONS The reduced magnitude, persistence and neutralization capacity of SARS-CoV-2 specific Abs after vaccination in anti-TNF-α-treated IBD patients were associated with impaired formation and maintenance of S-specific B memory cells, likely due to absent cTfh1 activation leading to extra-follicular immune responses and diminished B memory cell diversification. These observations have implications for patient-tailored vaccination schedules/vaccines in anti-TNF-α-treated patients, irrespective of their underlying disease. FUNDING The study was funded by third party funding of the Institute of Specific Prophylaxis and Tropical Medicine at the Medical University Vienna. The funders had no role in study design, data collection, data analyses, interpretation, or writing of report.
Collapse
Affiliation(s)
- Erika Garner-Spitzer
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Specific Prophylaxis and Tropical Medicine, Vienna, Austria.
| | - Angelika Wagner
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Specific Prophylaxis and Tropical Medicine, Vienna, Austria
| | - Venugopal Gudipati
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Anna-Margarita Schoetta
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Maria Orola-Taus
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Specific Prophylaxis and Tropical Medicine, Vienna, Austria
| | - Michael Kundi
- Medical University of Vienna, Center for Public Health, Vienna, Austria
| | - Renate Kunert
- Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Vienna, Austria
| | - Patrick Mayrhofer
- Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Vienna, Austria
| | - Johannes B Huppa
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Hannes Stockinger
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Rita Carsetti
- B Cell Research Unit, Immunology Research Area, Bambino Gesu Children's Hospital, IRCCS; Rome, Italy
| | - Pia Gattinger
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Department of Pathophysiology and Allergy Research, Division of Immunopathology, Vienna, Austria
| | - Rudolf Valenta
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Department of Pathophysiology and Allergy Research, Division of Immunopathology, Vienna, Austria; Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Bernhard Kratzer
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Vienna, Austria
| | - Al Nasar Ahmed Sehgal
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Vienna, Austria
| | - Winfried F Pickl
- Karl Landsteiner University of Health Sciences, Krems, Austria; Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Vienna, Austria
| | - Walter Reinisch
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Gottfried Novacek
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Ursula Wiedermann
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Specific Prophylaxis and Tropical Medicine, Vienna, Austria.
| |
Collapse
|
28
|
Bollimpelli VS, Reddy PBJ, Gangadhara S, Charles TP, Burton SL, Tharp GK, Styles TM, Labranche CC, Smith JC, Upadhyay AA, Sahoo A, Legere T, Shiferaw A, Velu V, Yu T, Tomai M, Vasilakos J, Kasturi SP, Shaw GM, Montefiori D, Bosinger SE, Kozlowski PA, Pulendran B, Derdeyn CA, Hunter E, Amara RR. Intradermal but not intramuscular modified vaccinia Ankara immunizations protect against intravaginal tier2 simian-human immunodeficiency virus challenges in female macaques. Nat Commun 2023; 14:4789. [PMID: 37553348 PMCID: PMC10409804 DOI: 10.1038/s41467-023-40430-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/28/2023] [Indexed: 08/10/2023] Open
Abstract
Route of immunization can markedly influence the quality of immune response. Here, we show that intradermal (ID) but not intramuscular (IM) modified vaccinia Ankara (MVA) vaccinations provide protection from acquisition of intravaginal tier2 simian-human immunodeficiency virus (SHIV) challenges in female macaques. Both routes of vaccination induce comparable levels of serum IgG with neutralizing and non-neutralizing activities. The protection in MVA-ID group correlates positively with serum neutralizing and antibody-dependent phagocytic activities, and envelope-specific vaginal IgA; while the limited protection in MVA-IM group correlates only with serum neutralizing activity. MVA-ID immunizations induce greater germinal center Tfh and B cell responses, reduced the ratio of Th1 to Tfh cells in blood and showed lower activation of intermediate monocytes and inflammasome compared to MVA-IM immunizations. This lower innate activation correlates negatively with induction of Tfh responses. These data demonstrate that the MVA-ID vaccinations protect against intravaginal SHIV challenges by modulating the innate and T helper responses.
Collapse
Affiliation(s)
- Venkata S Bollimpelli
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Pradeep B J Reddy
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Sailaja Gangadhara
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Tysheena P Charles
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Samantha L Burton
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Gregory K Tharp
- NHP Genomics Core Laboratory, Emory National Primate Research Center, Atlanta, GA, 30329, USA
| | - Tiffany M Styles
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Celia C Labranche
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Justin C Smith
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Amit A Upadhyay
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Anusmita Sahoo
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Traci Legere
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Ayalnesh Shiferaw
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Vijayakumar Velu
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory National Primate Research Center, Atlanta, GA, USA
| | - Tianwei Yu
- Rollins School of Public Health, Emory University, Atlanta, GA, 30322, USA
| | - Mark Tomai
- 3M Corporate Research and Materials Lab, Saint Paul, MN, USA
| | | | - Sudhir P Kasturi
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory National Primate Research Center, Atlanta, GA, USA
| | - George M Shaw
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David Montefiori
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Steven E Bosinger
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory National Primate Research Center, Atlanta, GA, USA
| | - Pamela A Kozlowski
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Bali Pulendran
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Cynthia A Derdeyn
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory National Primate Research Center, Atlanta, GA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Eric Hunter
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory National Primate Research Center, Atlanta, GA, USA
| | - Rama R Amara
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA.
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
29
|
Matz HC, McIntire KM, Ellebedy AH. 'Persistent germinal center responses: slow-growing trees bear the best fruits'. Curr Opin Immunol 2023; 83:102332. [PMID: 37150126 PMCID: PMC10829534 DOI: 10.1016/j.coi.2023.102332] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/06/2023] [Accepted: 04/09/2023] [Indexed: 05/09/2023]
Abstract
Germinal centers (GCs) are key microanatomical sites in lymphoid organs where responding B cells mature and undergo affinity-based selection. The duration of the GC reaction has long been assumed to be relatively brief, but recent studies in humans, nonhuman primates, and mice indicate that GCs can last for weeks to months after initial antigen exposure. This review examines recent studies investigating the factors that influence GC duration, including antigen persistence, T-follicular helper cells, and mode of immunization. Potential mechanisms for how persistent GCs influence the B-cell repertoire are considered. Overall, these studies provide a blueprint for how to design better vaccines that elicit persistent GC responses.
Collapse
Affiliation(s)
- Hanover C Matz
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Katherine M McIntire
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Ali H Ellebedy
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA; Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO, USA; The Andrew M. and Jane M. Bursky Center for Human Immunology & Immunotherapy Programs, USA.
| |
Collapse
|
30
|
Gonzales C, Liang Y, Fisher J, Card G, Sun J, Soong L. Alterations in germinal center formation and B cell activation during severe Orientia tsutsugamushi infection in mice. PLoS Negl Trop Dis 2023; 17:e0011090. [PMID: 37146079 PMCID: PMC10191367 DOI: 10.1371/journal.pntd.0011090] [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: 01/11/2023] [Revised: 05/17/2023] [Accepted: 04/17/2023] [Indexed: 05/07/2023] Open
Abstract
Scrub typhus is a poorly studied but life-threatening disease caused by the intracellular bacterium Orientia tsutsugamushi (Ot). Cellular and humoral immunity in Ot-infected patients is not long-lasting, waning as early as one-year post-infection; however, its underlying mechanisms remain unclear. To date, no studies have examined germinal center (GC) or B cell responses in Ot-infected humans or experimental animals. This study was aimed at evaluating humoral immune responses at acute stages of severe Ot infection and possible mechanisms underlying B cell dysfunction. Following inoculation with Ot Karp, a clinically dominant strain known to cause lethal infection in C57BL/6 mice, we measured antigen-specific antibody titers, revealing IgG2c as the dominant isotype induced by infection. Splenic GC responses were evaluated by immunohistology, co-staining for B cells (B220), T cells (CD3), and GCs (GL-7). Organized GCs were evident at day 4 post-infection (D4), but they were nearly absent at D8, accompanied by scattered T cells throughout splenic tissues. Flow cytometry revealed comparable numbers of GC B cells and T follicular helper (Tfh) cells at D4 and D8, indicating that GC collapse was not due to excessive death of these cell subtypes at D8. B cell RNAseq analysis revealed significant differences in expression of genes associated with B cell adhesion and co-stimulation at D8 versus D4. The significant downregulation of S1PR2 (a GC-specific adhesion gene) was most evident at D8, correlating with disrupted GC formation. Signaling pathway analysis uncovered downregulation of 71% of B cell activation genes at D8, suggesting attenuation of B cell activation during severe infection. This is the first study showing the disruption of B/T cell microenvironment and dysregulation of B cell responses during Ot infection, which may help understand the transient immunity associated with scrub typhus.
Collapse
Affiliation(s)
- Casey Gonzales
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Yuejin Liang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - James Fisher
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Galen Card
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Jiaren Sun
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Lynn Soong
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, Texas, United States of America
| |
Collapse
|
31
|
Marcial-Juárez E, Pérez-Toledo M, Nayar S, Pipi E, Alshayea A, Persaud R, Jossi SE, Lamerton R, Barone F, Henderson IR, Cunningham AF. Salmonella infection induces the reorganization of follicular dendritic cell networks concomitant with the failure to generate germinal centers. iScience 2023; 26:106310. [PMID: 36950118 PMCID: PMC10025972 DOI: 10.1016/j.isci.2023.106310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/07/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
Germinal centers (GCs) are sites where plasma and memory B cells form to generate high-affinity, Ig class-switched antibodies. Specialized stromal cells called follicular dendritic cells (FDCs) are essential for GC formation. During systemic Salmonella Typhimurium (STm) infection GCs are absent, whereas extensive extrafollicular and switched antibody responses are maintained. The mechanisms that underpin the absence of GC formation are incompletely understood. Here, we demonstrate that STm induces a reversible disruption of niches within the splenic microenvironment, including the T and B cell compartments and the marginal zone. Alongside these effects after infection, mature FDC networks are strikingly absent, whereas immature FDC precursors, including marginal sinus pre-FDCs (MadCAM-1+) and perivascular pre-FDCs (PDGFRβ+) are enriched. As normal FDC networks re-establish, extensive GCs become detectable throughout the spleen. Therefore, the reorganization of FDC networks and the loss of GC responses are key, parallel features of systemic STm infections.
Collapse
Affiliation(s)
- Edith Marcial-Juárez
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
| | - Marisol Pérez-Toledo
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
| | - Saba Nayar
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
| | - Elena Pipi
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
| | - Areej Alshayea
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
| | - Ruby Persaud
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
| | - Sian E. Jossi
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
| | - Rachel Lamerton
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
| | - Francesca Barone
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, UK and Sandwell and West Birmingham Trust, Birmingham, West Midlands, B15 2TH, United Kingdom
| | - Ian R. Henderson
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD4072, Australia
| | - Adam F. Cunningham
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
| |
Collapse
|
32
|
Deng S, Graham ML, Chen XM. The Complexity of Interferon Signaling in Host Defense against Protozoan Parasite Infection. Pathogens 2023; 12:319. [PMID: 36839591 PMCID: PMC9962834 DOI: 10.3390/pathogens12020319] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/11/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Protozoan parasites, such as Plasmodium, Leishmania, Toxoplasma, Cryptosporidium, and Trypanosoma, are causative agents of health-threatening diseases in both humans and animals, leading to significant health risks and socioeconomic losses globally. The development of effective therapeutic and prevention strategies for protozoan-caused diseases requires a full understanding of the pathogenesis and protective events occurring in infected hosts. Interferons (IFNs) are a family of cytokines with diverse biological effects in host antimicrobial defense and disease pathogenesis, including protozoan parasite infection. Type II IFN (IFN-γ) has been widely recognized as the essential defense cytokine in intracellular protozoan parasite infection, whereas recent studies also revealed the production and distinct function of type I and III IFNs in host defense against these parasites. Decoding the complex network of the IFN family in host-parasite interaction is critical for exploring potential new therapeutic strategies against intracellular protozoan parasite infection. Here, we review the complex effects of IFNs on the host defense against intracellular protozoan parasites and the crosstalk between distinct types of IFN signaling during infections.
Collapse
Affiliation(s)
- Silu Deng
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Marion L. Graham
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612, USA
| | - Xian-Ming Chen
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
33
|
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: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
34
|
Kalkal M, Das J. Current understanding of the immune potential of B-cell subsets in malarial pathogenesis. Front Microbiol 2023; 14:1046002. [PMID: 36778886 PMCID: PMC9909418 DOI: 10.3389/fmicb.2023.1046002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 01/02/2023] [Indexed: 01/28/2023] Open
Abstract
In the past several decades, our understanding of how B cells are generated and what function they perform has continued to advance. It is widely accepted that B-cell subsets play a critical role in mediating immune response. Surprisingly, human and murine malarial infections cause major alterations in the composition of B-cell subsets in both the spleen and periphery. Multiple B-cell subsets are well characterized in murine models following primary and secondary infection, although in human malarial infection, these subsets are not well defined. Furthermore, a rare known function of B cells includes the potential role of regulating the activities of other cells in the body as regulatory cells. Plasmodium infection strongly alters the frequency of these regulatory B cells indicating the immunoregulatory function of B cells in malarial. It is important to note that these subsets, taken together, form the cellular basis of humoral immune responses, allowing protection against a wide array of Plasmodium antigens to be achieved. However, it remains a challenge and an important area of investigation to understand how these B-cell subsets work together to provide protection against Plasmodium infection.
Collapse
|
35
|
Starshinova A, Malkova A, Zinchenko Y, Kudryavtsev I, Serebriakova M, Akisheva T, Lapin S, Mazing A, Kudlay D, Glushkova A, Yablonskiy P, Shoenfeld Y. Identification of autoimmune markers in pulmonary tuberculosis. Front Immunol 2023; 13:1059714. [PMID: 36761174 PMCID: PMC9905676 DOI: 10.3389/fimmu.2022.1059714] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 12/14/2022] [Indexed: 01/26/2023] Open
Abstract
Introduction Pathogenesis of many autoimmune diseases is mainly promoted by poorly regulated and/or wrong targeted immune response to pathogens including M. tuberculosis. Autoimmunity is one of the processes with are characteristics of tuberculosis (Tbc). The aim was to determine the autoimmune clinical and immunological features in patients with pulmonary Tbc. Materials and methods A prospective comparative study was performed in 2017 - 2019 with the inclusion of 46 patients with Tbc. The trigger factors and clinical manifestations, autoantibodies, peripheral blood B cell subsets were stained with fluorochrome-conjugated monoclonal antibodies. 40 healthy volunteers in the control group, were matched for age with no chronic diseases, contacts with TB patients and changes in their laboratory parameters. A statistical analysis was done with GraphPad Prism 6, Statistica 10 (Statsoft) and MedCalc - version 18.2.1 values. Results There were no significant ASIA triggers in Tbc patients and control group. 21.1% of Tbc patients had a high level of a rheumatoid factor and in 47.4% complement system factor C3 was high; anti-MCV was detected in 60.7% of Tbc patients. Relative and absolute frequencies of "naïve" Bm1 cells and eBm5 were significantly decreased and activated pre-germinal-center Bm2' cells were significantly increased in Tbc patients. The CD24++CD38++ B cells were increased in Tbc vs control group (10.25% vs 5.42%), p < 0.001, and 19 cell/1μL (10; 290 vs 11 cell/1μL (6; 20), p = 0.029, respectively). The frequency of CXCR3+CCR4- Tfh1 cells was significantly lower in Tbc vs control one (26.52% vs. 31.00%, p = 0.004), while CXCR3-CCR4+ Tfh2 cells were increased in Tbc (20.31% vs. controls (16.56%, p = 0.030). The absolute numbers of Tfh1 cells were decreased in the Tbc vs. control (24 cell/1μL vs. 37 cell/1μL p = 0.005). Conclusion The results of our study showed that the detection of a rheumatoid factor, the components of complement system and anti-MCV in complex with alterations in B cells and follicular Th cell subsets may indicate a presence of autoimmunity in the pathogenesis of tuberculosis, but they are not specific. The indicators of autoimmune-related provide new opportunities in the Tbc treatment.
Collapse
Affiliation(s)
- Anna Starshinova
- St. Petersburg State University, St. Petersburg, Russia,*Correspondence: Anna Starshinova,
| | - Anna Malkova
- St. Petersburg State University, St. Petersburg, Russia
| | - Yulia Zinchenko
- St. Petersburg State University, St. Petersburg, Russia,St. Petersburg Research Institute of Phthisiopulmonology, St. Petersburg, Russia
| | - Igor Kudryavtsev
- Department of Immunology, Institution of Experimental Medicine, St. Petersburg, Russia
| | - Maria Serebriakova
- Department of Immunology, Institution of Experimental Medicine, St. Petersburg, Russia
| | - Tatiana Akisheva
- Department of Immunology, Institution of Experimental Medicine, St. Petersburg, Russia
| | - Sergey Lapin
- St. Petersburg State Medical University, St. Petersburg, Russia
| | | | - Dmitry Kudlay
- Institute of Immunology, Moscow, Russia,Bekhterev Psychoneurological Institute, St. Petersburg, Russia
| | - Anzhela Glushkova
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel Hashomer, Israel
| | - Piotr Yablonskiy
- St. Petersburg State University, St. Petersburg, Russia,St. Petersburg Research Institute of Phthisiopulmonology, St. Petersburg, Russia
| | - Yehuda Shoenfeld
- St. Petersburg State University, St. Petersburg, Russia,Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel Hashomer, Israel
| |
Collapse
|
36
|
Zhu Q, Xu Y, Wang T, Xie F. Innate and adaptive immune response in SARS-CoV-2 infection-Current perspectives. Front Immunol 2022; 13:1053437. [PMID: 36505489 PMCID: PMC9727711 DOI: 10.3389/fimmu.2022.1053437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 11/09/2022] [Indexed: 11/24/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) has been a global pandemic, caused by a novel coronavirus strain with strong infectivity, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). With the in-depth research, the close relationship between COVID-19 and immune system has been dug out. During the infection, macrophages, dendritic cells, natural killer cells, CD8+ T cells, Th1, Th17, Tfh cells and effector B cells are all involved in the anti-SARS-CoV-2 responses, however, the dysfunctional immune responses will ultimately lead to the excessive inflammation, acute lung injury, even other organ failure. Thus, a detailed understanding of pertinent immune response during COVID-19 will provide insights in predicting disease outcomes and developing appropriate therapeutic approaches. In this review, we mainly clarify the role of immune cells in COVID-19 and the target-vaccine development and treatment.
Collapse
Affiliation(s)
- Qiugang Zhu
- Department of Laboratory Medicine, Shangyu People’s Hospital of Shaoxing, Shaoxing, China
| | - Yan Xu
- Department of Respiratory Medicine, Shangyu People’s Hospital of Shaoxing, Shaoxing, China
| | - Ting Wang
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Feiting Xie
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China,*Correspondence: Feiting Xie,
| |
Collapse
|
37
|
Kim MJ, Chu KB, Lee SH, Kang HJ, Yoon KW, Ahmed MA, Quan FS. Recombinant Vaccinia Virus Expressing Plasmodium berghei Apical Membrane Antigen 1 or Microneme Protein Enhances Protection against P. berghei Infection in Mice. Trop Med Infect Dis 2022; 7:350. [PMID: 36355892 PMCID: PMC9698705 DOI: 10.3390/tropicalmed7110350] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 07/27/2023] Open
Abstract
Recombinant vaccinia viruses (rVV) are effective antigen delivery vectors and are researched widely as vaccine platforms against numerous diseases. Apical membrane antigen 1 (AMA1) is one of the candidate antigens for malaria vaccines but rising concerns regarding its genetic diversity and polymorphism have necessitated the need to search for an alternative antigen. Here, we compare the efficacies of the rVV vaccines expressing either AMA1 or microneme protein (MIC) of Plasmodium berghei in mice. Mice (BALB/c) were immunized with either rVV-AMA1 or rVV-MIC and subsequently challenge-infected with P. berghei. Compared to the control group, both antigens elicited elevated levels of parasite-specific antibody responses. Immunization with either one of the two vaccines induced high levels of T cells and germinal center B cell responses. Interestingly, rVV-MIC immunization elicited higher levels of cellular immune response compared to rVV-AMA1 immunization, and significantly reduced pro-inflammatory cytokine productions were observed from the former vaccine. While differences in parasitemia and bodyweight changes were negligible between rVV-AMA1 and rVV-MIC immunization groups, prolonged survival was observed for the latter of the two. Based on these results, our findings suggest that the rVV expressing the P. berghei MIC could be a vaccine-candidate antigen.
Collapse
Affiliation(s)
- Min-Ju Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Ki-Back Chu
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Su-Hwa Lee
- Department of Medical Zoology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Hae-Ji Kang
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Keon-Woong Yoon
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Md Atique Ahmed
- ICMR-Regional Medical Research Centre, NE Region, Dibrugarh 786010, Assam, India
| | - Fu-Shi Quan
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
- Department of Medical Zoology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| |
Collapse
|
38
|
Nishio A, Hasan S, Park H, Park N, Salas JH, Salinas E, Kardava L, Juneau P, Frumento N, Massaccesi G, Moir S, Bailey JR, Grakoui A, Ghany MG, Rehermann B. Serum neutralization activity declines but memory B cells persist after cure of chronic hepatitis C. Nat Commun 2022; 13:5446. [PMID: 36114169 PMCID: PMC9481596 DOI: 10.1038/s41467-022-33035-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/30/2022] [Indexed: 11/09/2022] Open
Abstract
The increasing incidence of hepatitis C virus (HCV) infections underscores the need for an effective vaccine. Successful vaccines to other viruses generally depend on a long-lasting humoral response. However, data on the half-life of HCV-specific responses are lacking. Here we study archived sera and mononuclear cells that were prospectively collected up to 18 years after cure of chronic HCV infection to determine the role of HCV antigen in maintaining neutralizing antibody and B cell responses. We show that HCV-neutralizing activity decreases rapidly in potency and breadth after curative treatment. In contrast, HCV-specific memory B cells persist, and display a restored resting phenotype, normalized chemokine receptor expression and preserved ability to differentiate into antibody-secreting cells. The short half-life of HCV-neutralizing activity is consistent with a lack of long-lived plasma cells. The persistence of HCV-specific memory B cells and the reduced inflammation after cure provide an opportunity for vaccination to induce protective immunity against re-infection.
Collapse
Affiliation(s)
- Akira Nishio
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD, 20892, USA
| | - Sharika Hasan
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD, 20892, USA
| | - Heiyoung Park
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD, 20892, USA
| | - Nana Park
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD, 20892, USA
| | - Jordan H Salas
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Eduardo Salinas
- Division of Infectious Diseases, Emory Vaccine Center, Division of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Emory National Primate Research Center, Emory Vaccine Center, Atlanta, GA, 30329, USA
| | - Lela Kardava
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, DHHS, Bethesda, MD, 20892, USA
| | - Paul Juneau
- Division of Data Services, NIH Library, Office of Research Services, National Institutes of Health, Bethesda, MD, USA
- Contractor- Zimmerman Associates, Inc, Fairfax, VA, USA
| | - Nicole Frumento
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Guido Massaccesi
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Susan Moir
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, DHHS, Bethesda, MD, 20892, USA
| | - Justin R Bailey
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Arash Grakoui
- Division of Infectious Diseases, Emory Vaccine Center, Division of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Emory National Primate Research Center, Emory Vaccine Center, Atlanta, GA, 30329, USA
| | - Marc G Ghany
- Clinical Research Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD, 20892, USA
| | - Barbara Rehermann
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD, 20892, USA.
| |
Collapse
|
39
|
Olatunde AC, Cornwall DH, Roedel M, Lamb TJ. Mouse Models for Unravelling Immunology of Blood Stage Malaria. Vaccines (Basel) 2022; 10:1525. [PMID: 36146602 PMCID: PMC9501382 DOI: 10.3390/vaccines10091525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
Malaria comprises a spectrum of disease syndromes and the immune system is a major participant in malarial disease. This is particularly true in relation to the immune responses elicited against blood stages of Plasmodium-parasites that are responsible for the pathogenesis of infection. Mouse models of malaria are commonly used to dissect the immune mechanisms underlying disease. While no single mouse model of Plasmodium infection completely recapitulates all the features of malaria in humans, collectively the existing models are invaluable for defining the events that lead to the immunopathogenesis of malaria. Here we review the different mouse models of Plasmodium infection that are available, and highlight some of the main contributions these models have made with regards to identifying immune mechanisms of parasite control and the immunopathogenesis of malaria.
Collapse
Affiliation(s)
| | | | | | - Tracey J. Lamb
- Department of Pathology, University of Utah, Emma Eccles Jones Medical Research Building, 15 N Medical Drive E, Room 1420A, Salt Lake City, UT 84112, USA
| |
Collapse
|
40
|
Nunes-Cabaço H, Moita D, Prudêncio M. Five decades of clinical assessment of whole-sporozoite malaria vaccines. Front Immunol 2022; 13:977472. [PMID: 36159849 PMCID: PMC9493004 DOI: 10.3389/fimmu.2022.977472] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
In 1967, pioneering work by Ruth Nussenzweig demonstrated for the first time that irradiated sporozoites of the rodent malaria parasite Plasmodium berghei protected mice against a challenge with infectious parasites of the same species. This remarkable finding opened up entirely new prospects of effective vaccination against malaria using attenuated sporozoites as immunization agents. The potential for whole-sporozoite-based immunization in humans was established in a clinical study in 1973, when a volunteer exposed to X-irradiated P. falciparum sporozoites was found to be protected against malaria following challenge with a homologous strain of this parasite. Nearly five decades later, much has been achieved in the field of whole-sporozoite malaria vaccination, and multiple reports on the clinical evaluation of such candidates have emerged. However, this process has known different paces before and after the turn of the century. While only a few clinical studies were published in the 1970’s, 1980’s and 1990’s, remarkable progress was made in the 2000’s and beyond. This article reviews the history of the clinical assessment of whole-sporozoite malaria vaccines over the last forty-nine years, highlighting the impressive achievements made over the last few years, and discussing some of the challenges ahead.
Collapse
|
41
|
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
|
42
|
Masiá M, Fernández-González M, García JA, Padilla S, García-Abellán J, Botella Á, Mascarell P, Agulló V, Gutiérrez F. Robust long-term immunity to SARS-CoV-2 in patients recovered from severe COVID-19 after interleukin-6 blockade. EBioMedicine 2022; 82:104153. [PMID: 35816896 PMCID: PMC9265168 DOI: 10.1016/j.ebiom.2022.104153] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/30/2022] [Accepted: 06/24/2022] [Indexed: 11/16/2022] Open
Abstract
Background Whether interleukin-6 (IL-6) blockade in patients with COVID-19 will affect the protective immunity against SARS-CoV-2 has become an important concern for anti-IL-6 therapy. We aimed to investigate the effects of IL-6 blockade on long-term immunity to SARS-CoV-2. Methods Prospective, longitudinal cohort study conducted in patients hospitalized for severe or critical COVID-19 with laboratory confirmed SARS-CoV-2 infection. We assessed humoral (anti-S1 domain of the spike [S], anti-nucleocapsid [N], anti-trimeric spike [TrimericS] IgG, and neutralizing antibodies [Nab]) and T-cell (interferon-γ release assay [IGRA]) responses and evaluated the incidence of reinfections over one year after infection in patients undergoing IL-6 blockade with tocilizumab and compared them with untreated subjects. Findings From 150 adults admitted with confirmed SARS-CoV-2 infection, 78 were 1:1 propensity score-matched. Patients receiving anti-IL6 therapy showed a shorter time to S-IgG seropositivity and stronger S-IgG and N-IgG antibody responses. Among unvaccinated subjects one year after infection, median (Q1-Q3) levels of TrimericS-IgG (295 vs 121 BAU/mL; p = 0.011) and Nab (74.7 vs 41.0 %IH; p = 0.012) were higher in those undergoing anti-IL6 therapy, and a greater proportion of them had Nab (80.6% vs 57.7%; p = 0.028). T-cell immunity was also better in those treated with anti-IL6, with higher median (Q1-Q3) interferon-γ responses (1760 [702–3992] vs 542 [35–1716] mIU/mL; p = 0.013) and more patients showing positive T-cell responses in the IGRA one year after infection. Patients treated with anti-IL6 had fewer reinfections during follow-up and responded to vaccination with robust increase in both antibody and T-cell immunity. Interpretation IL-6 blockade in patients with severe COVID-19 does not have deleterious effects on long-term immunity to SARS-CoV-2. The magnitude of both antibody and T-cell responses was stronger than the observed in non-anti-cytokine-treated patients with no increase in the risk of reinfections. Funding Instituto de Salud Carlos-III (Spain).
Collapse
|
43
|
Chan JA, Loughland JR, de la Parte L, Okano S, Ssewanyana I, Nalubega M, Nankya F, Musinguzi K, Rek J, Arinaitwe E, Tipping P, Bourke P, Andrew D, Dooley N, SheelaNair A, Wines BD, Hogarth PM, Beeson JG, Greenhouse B, Dorsey G, Kamya M, Hartel G, Minigo G, Feeney M, Jagannathan P, Boyle MJ. Age-dependent changes in circulating Tfh cells influence development of functional malaria antibodies in children. Nat Commun 2022; 13:4159. [PMID: 35851033 PMCID: PMC9293980 DOI: 10.1038/s41467-022-31880-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 07/08/2022] [Indexed: 01/29/2023] Open
Abstract
T-follicular helper (Tfh) cells are key drivers of antibodies that protect from malaria. However, little is known regarding the host and parasite factors that influence Tfh and functional antibody development. Here, we use samples from a large cross-sectional study of children residing in an area of high malaria transmission in Uganda to characterize Tfh cells and functional antibodies to multiple parasites stages. We identify a dramatic re-distribution of the Tfh cell compartment with age that is independent of malaria exposure, with Th2-Tfh cells predominating in early childhood, while Th1-Tfh cell gradually increase to adult levels over the first decade of life. Functional antibody acquisition is age-dependent and hierarchical acquired based on parasite stage, with merozoite responses followed by sporozoite and gametocyte antibodies. Antibodies are boosted in children with current infection, and are higher in females. The children with the very highest antibody levels have increased Tfh cell activation and proliferation, consistent with a key role of Tfh cells in antibody development. Together, these data reveal a complex relationship between the circulating Tfh compartment, antibody development and protection from malaria.
Collapse
Affiliation(s)
- Jo-Anne Chan
- Burnet Institute, Melbourne, VIC, Australia
- Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
| | - Jessica R Loughland
- QIMR-Berghofer Medical Research Institute, Herston, QLD, Australia
- Global and Tropical Health Division, Menzies School of Health Research, Tiwi, Australia
| | | | - Satomi Okano
- QIMR-Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Isaac Ssewanyana
- Infectious Diseases Research Collaboration, Kampala, Uganda
- London School of Hygiene and Tropical Medicine, London, UK
| | - Mayimuna Nalubega
- QIMR-Berghofer Medical Research Institute, Herston, QLD, Australia
- Infectious Diseases Research Collaboration, Kampala, Uganda
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | | | | | - John Rek
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | - Peta Tipping
- Global and Tropical Health Division, Menzies School of Health Research, Tiwi, Australia
| | - Peter Bourke
- Division of Medicine, Cairns Hospital, Manunda, QLD, Australia
| | - Dean Andrew
- QIMR-Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Nicholas Dooley
- QIMR-Berghofer Medical Research Institute, Herston, QLD, Australia
- Griffith University, Brisbane, QLD, Australia
| | - Arya SheelaNair
- QIMR-Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Bruce D Wines
- Burnet Institute, Melbourne, VIC, Australia
- Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, Australia
| | - P Mark Hogarth
- Burnet Institute, Melbourne, VIC, Australia
- Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, Australia
| | - James G Beeson
- Burnet Institute, Melbourne, VIC, Australia
- Department of Medicine, The University of Melbourne, Parkville, VIC, Australia
- Department of Microbiology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | | | - Grant Dorsey
- University of California San Francisco, San Francisco, CA, USA
| | - Moses Kamya
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Gunter Hartel
- QIMR-Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Gabriela Minigo
- Global and Tropical Health Division, Menzies School of Health Research, Tiwi, Australia
- College of Health and Human Sciences, Charles Darwin University, Darwin, NT, Australia
| | - Margaret Feeney
- University of California San Francisco, San Francisco, CA, USA
| | | | - Michelle J Boyle
- Burnet Institute, Melbourne, VIC, Australia.
- QIMR-Berghofer Medical Research Institute, Herston, QLD, Australia.
- Global and Tropical Health Division, Menzies School of Health Research, Tiwi, Australia.
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.
- Griffith University, Brisbane, QLD, Australia.
| |
Collapse
|
44
|
Yu D, Walker LSK, Liu Z, Linterman MA, Li Z. Targeting T FH cells in human diseases and vaccination: rationale and practice. Nat Immunol 2022; 23:1157-1168. [PMID: 35817844 DOI: 10.1038/s41590-022-01253-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 05/24/2022] [Indexed: 12/13/2022]
Abstract
The identification of CD4+ T cells localizing to B cell follicles has revolutionized the knowledge of how humoral immunity is generated. Follicular helper T (TFH) cells support germinal center (GC) formation and regulate clonal selection and differentiation of memory and antibody-secreting B cells, thus controlling antibody affinity maturation and memory. TFH cells are essential in sustaining protective antibody responses necessary for pathogen clearance in infection and vaccine-mediated protection. Conversely, aberrant and excessive TFH cell responses mediate and sustain pathogenic antibodies to autoantigens, alloantigens, and allergens, facilitate lymphomagenesis, and even harbor viral reservoirs. TFH cell generation and function are determined by T cell antigen receptor (TCR), costimulation, and cytokine signals, together with specific metabolic and survival mechanisms. Such regulation is crucial to understanding disease pathogenesis and informing the development of emerging therapies for disease or novel approaches to boost vaccine efficacy.
Collapse
Affiliation(s)
- Di Yu
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Australia. .,Ian Frazer Centre for Children's Immunotherapy Research, Child Health Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Australia.
| | - Lucy S K Walker
- Institute of Immunity & Transplantation, Division of Infection & Immunity, University College London, Royal Free Campus, London, UK
| | - Zheng Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | - Zhanguo Li
- Department of Rheumatology & Immunology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
45
|
Bhattacharya D. Instructing durable humoral immunity for COVID-19 and other vaccinable diseases. Immunity 2022; 55:945-964. [PMID: 35637104 PMCID: PMC9085459 DOI: 10.1016/j.immuni.2022.05.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/02/2022] [Accepted: 05/05/2022] [Indexed: 11/29/2022]
Abstract
Many aspects of SARS-CoV-2 have fully conformed with the principles established by decades of viral immunology research, ultimately leading to the crowning achievement of highly effective COVID-19 vaccines. Nonetheless, the pandemic has also exposed areas where our fundamental knowledge is thinner. Some key unknowns are the duration of humoral immunity post-primary infection or vaccination and how long booster shots confer protection. As a corollary, if protection does not last as long as desired, what are some ways it can be improved? Here, I discuss lessons from other infections and vaccines that point to several key features that influence durable antibody production and the perseverance of immunity. These include (1) the specific innate sensors that are initially triggered, (2) the kinetics of antigen delivery and persistence, (3) the starting B cell receptor (BCR) avidity and antigen valency, and (4) the memory B cell subsets that are recalled by boosters. I further highlight the fundamental B cell-intrinsic and B cell-extrinsic pathways that, if understood better, would provide a rational framework for vaccines to reliably provide durable immunity.
Collapse
Affiliation(s)
- Deepta Bhattacharya
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ 85724, USA.
| |
Collapse
|
46
|
Feng H, Zhao X, Xie J, Bai X, Fu W, Chen H, Tang H, Wang X, Dong C. Pathogen-associated T follicular helper cell plasticity is critical in anti-viral immunity. SCIENCE CHINA LIFE SCIENCES 2022; 65:1075-1090. [PMID: 35258743 DOI: 10.1007/s11427-021-2055-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/26/2021] [Indexed: 01/12/2023]
|
47
|
Ishola D, The EBOVAC-Salone Malaria Infection (MALI) Sub-Study Team
BahOsman MohamedBangalieFoday SumaBanguraAgnesDavidIfeoluDeenGibrilla FadluFombahAugustinJallohAbdulai BerberKamaraAbu BakarrKamaraIbrahim FranklynKamaraMichaelLeighBailahMoroviaFodayRogersBaimbaSamaiMohamedSerry-BanguraAlimamyShekuMahmudSwarayIbrahimAnumendemDicksonGaddahAugusteBockstalVikiKeshinroBabajideRobinsonCynthiaAfolabiMuhammedAkooPaulineAyiekoPhilipBaidenFrankGallagherKatherineGreenwoodBrianIsholaDavidKohnBrianKowuorDickensLawalBolarindeLoweBrettMannoDanielaOdenyLazarusOtienoTudaOwusu-KyeiKwabenaSmoutElizabethTindanbilDanielWatson-JonesDeborah. Asymptomatic Malaria Infection and the Immune Response to the 2-Dose Ad26.ZEBOV, MVA-BN-Filo Ebola Vaccine Regimen in Adults and Children. Clin Infect Dis 2022; 75:1585-1593. [PMID: 35640636 PMCID: PMC9617582 DOI: 10.1093/cid/ciac209] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Indexed: 11/14/2022] Open
Abstract
Background Malaria infection affects the immune response to some vaccines. As Ebola virus (EBOV) outbreaks have occurred mainly in malaria-endemic countries, we have assessed whether asymptomatic malaria affects immune responses to the 2-dose Ad26.ZEBOV, MVA-BN-Filo Ebola vaccine regimen. Methods In this sub-study of the EBOVAC-Salone Ebola vaccine trial in Sierra Leone, malaria microscopy was performed at the time of Ebola vaccination. Participants with symptomatic malaria were treated before vaccination. Ebola vaccine responses were assessed post-dose 1 (day 57) and post-dose 2 (day 78) by the EBOV glycoprotein FANG enzyme-linked immunosorbent assay (ELISA), and responses expressed as geometric mean concentrations (GMCs). Geometric mean ratios (GMRs) of the GMCs in malaria-positive versus malaria-negative participants were derived with 95% confidence intervals (CIs). Results A total of 587 participants were studied, comprising 188 adults (≥18 years) and 399 children (in age groups of 12–17, 4–11, and 1–3 years). Asymptomatic malaria was observed in 47.5% of adults and 51.5% of children on day 1. Post-dose 1, GMCs were lower in 1–3-year-old malaria-positive compared with malaria-negative children (age group–specific GMR, .56; 95% CI, .39–.81) but not in older age groups. Post-dose 2, there was no consistent effect of malaria infection across the different age groups but there was a trend toward a lower response (GMR, .82; 95% CI, .67–1.02). Conclusions The Ad26.ZEBOV, MVA-BN-Filo Ebola vaccine regimen is immunogenic in participants with asymptomatic malaria. Therefore, it is not necessary to screen for asymptomatic malaria infection prior to vaccination with this regimen.
Collapse
Affiliation(s)
- D Ishola
- Correspondence: D. Ishola, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK ()
| | | |
Collapse
|
48
|
Kudryavtsev I, Rubinstein A, Golovkin A, Kalinina O, Vasilyev K, Rudenko L, Isakova-Sivak I. Dysregulated Immune Responses in SARS-CoV-2-Infected Patients: A Comprehensive Overview. Viruses 2022; 14:1082. [PMID: 35632823 PMCID: PMC9147674 DOI: 10.3390/v14051082] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/13/2022] [Accepted: 05/16/2022] [Indexed: 12/20/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was first detected in humans more than two years ago and caused an unprecedented socio-economic burden on all countries around the world. Since then, numerous studies have attempted to identify various mechanisms involved in the alterations of innate and adaptive immunity in COVID-19 patients, with the ultimate goal of finding ways to correct pathological changes and improve disease outcomes. State-of-the-art research methods made it possible to establish precise molecular mechanisms which the new virus uses to trigger multisystem inflammatory syndrome and evade host antiviral immune responses. In this review, we present a comprehensive analysis of published data that provide insight into pathological changes in T and B cell subsets and their phenotypes, accompanying the acute phase of the SARS-CoV-2 infection. This knowledge might help reveal new biomarkers that can be utilized to recognize case severity early as well as to provide additional objective information on the effective formation of SARS-CoV-2-specific immunity and predict long-term complications of COVID-19, including a large variety of symptoms termed the 'post-COVID-19 syndrome'.
Collapse
Affiliation(s)
- Igor Kudryavtsev
- Institute of Experimental Medicine, 197022 Saint Petersburg, Russia; (I.K.); (A.R.); (K.V.); (L.R.)
| | - Artem Rubinstein
- Institute of Experimental Medicine, 197022 Saint Petersburg, Russia; (I.K.); (A.R.); (K.V.); (L.R.)
| | - Alexey Golovkin
- Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia; (A.G.); (O.K.)
| | - Olga Kalinina
- Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia; (A.G.); (O.K.)
| | - Kirill Vasilyev
- Institute of Experimental Medicine, 197022 Saint Petersburg, Russia; (I.K.); (A.R.); (K.V.); (L.R.)
| | - Larisa Rudenko
- Institute of Experimental Medicine, 197022 Saint Petersburg, Russia; (I.K.); (A.R.); (K.V.); (L.R.)
| | - Irina Isakova-Sivak
- Institute of Experimental Medicine, 197022 Saint Petersburg, Russia; (I.K.); (A.R.); (K.V.); (L.R.)
| |
Collapse
|
49
|
He C, Malone MJ, Wendel BS, Ma KY, Del Alcazar D, Weiner DB, De Jager PL, Del Río-Estrada PM, Ablanedo-Terrazas Y, Reyes-Terán G, Su LF, Jiang N. Transcriptome and TCR Repertoire Measurements of CXCR3 + T Follicular Helper Cells Within HIV-Infected Human Lymph Nodes. Front Immunol 2022; 13:859070. [PMID: 35619703 PMCID: PMC9128546 DOI: 10.3389/fimmu.2022.859070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/06/2022] [Indexed: 12/15/2022] Open
Abstract
Follicular-helper T cells (TFH) are an essential arm of the adaptive immune system. Although TFH were first discovered through their ability to contribute to antibody affinity maturation through co-stimulatory interactions with B cells, new light has been shed on their ability to remain a complex and functionally plastic cell type. Due to a lack sample availability, however, many studies have been limited to characterizing TFH in mice or non-canonical tissue types, such as peripheral blood. Such constraints have resulted in a limited, and sometimes contradictory, understanding of this fundamental cell type. One subset of TFH receiving attention in chronic infection are CXCR3-expressing TFH cells (CXCR3+TFH) due to their abnormal accumulation in secondary lymphoid tissues. Their function and clonal relationship with other TFH subsets in lymphoid tissues during infection, however, remains largely unclear. We thus systematically investigated this and other subsets of TFH within untreated HIV-infected human lymph nodes using Mass CyTOF and a combination of RNA and TCR repertoire sequencing. We show an inflation of the CXCR3+TFH compartment during HIV infection that correlates with a lower HIV burden. Deeper analysis into this population revealed a functional shift of CXCR3+TFH away from germinal center TFH (GC-TFH), including the altered expression of several important transcription factors and cytokines. CXCR3+TFH also upregulated cell migration transcriptional programs and were clonally related to peripheral TFH populations. In combination, these data suggest that CXCR3+TFH have a greater tendency to enter circulation than their CXCR3- counterparts, potentially functioning through distinct modalities that may lead to enhanced defense.
Collapse
Affiliation(s)
- Chenfeng He
- Department of Biomedical Engineering, Cockrell School of Engineering, University of Texas at Austin, Austin, TX, United States
| | - Michael J. Malone
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States,Interdisciplinary Life Sciences Graduate Program, University of Texas at Austin, Austin, TX, United States
| | - Ben S. Wendel
- Department of Biomedical Engineering, Cockrell School of Engineering, University of Texas at Austin, Austin, TX, United States,McKetta Department of Chemical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Ke-Yue Ma
- Interdisciplinary Life Sciences Graduate Program, University of Texas at Austin, Austin, TX, United States
| | - Daniel Del Alcazar
- Department of Medicine, Division of Rheumatology, Perelman School of Medicine, Institute for Immunology, University of Pennsylvania, Philadelphia, PA, United States,Corporal Michael J Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - David B. Weiner
- Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA, United States
| | - Philip L. De Jager
- Columbia University Medical Center, Center for Translational and Computational Neuroimmunology, New York, NY, United States
| | - Perla M. Del Río-Estrada
- Departamento de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias, Ciudad de México, Mexico
| | - Yuria Ablanedo-Terrazas
- Departamento de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias, Ciudad de México, Mexico
| | - Gustavo Reyes-Terán
- Comisión Coordinadora de Institutos Nacional de Salud y Hospitales de Alta Especialidad, Secretaría de Salud, Ciudad de México, Mexico
| | - Laura F. Su
- Department of Medicine, Division of Rheumatology, Perelman School of Medicine, Institute for Immunology, University of Pennsylvania, Philadelphia, PA, United States,Corporal Michael J Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States,*Correspondence: Ning Jiang, ; Laura F. Su,
| | - Ning Jiang
- Department of Biomedical Engineering, Cockrell School of Engineering, University of Texas at Austin, Austin, TX, United States,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States,Interdisciplinary Life Sciences Graduate Program, University of Texas at Austin, Austin, TX, United States,Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States,*Correspondence: Ning Jiang, ; Laura F. Su,
| |
Collapse
|
50
|
Dalit L, Alvarado C, Küijper L, Kueh AJ, Weir A, D’Amico A, Herold MJ, Vince JE, Nutt SL, Groom JR. CXCL11 expressing C57BL/6 mice have intact adaptive immune responses to viral infection. Immunol Cell Biol 2022; 100:312-322. [PMID: 35233830 PMCID: PMC9542850 DOI: 10.1111/imcb.12541] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 02/24/2022] [Accepted: 02/26/2022] [Indexed: 12/13/2022]
Abstract
The chemokine receptor CXCR3 is expressed on immune cells to co‐ordinate lymphocyte activation and migration. CXCR3 binds three chemokine ligands, CXCL9, CXCL10 and CXCL11. These ligands display distinct expression patterns and ligand signaling biases; however, how each ligand functions individually and collaboratively is incompletely understood. CXCL9 and CXCL10 are considered pro‐inflammatory chemokines during viral infection, while CXCL11 may induce a tolerizing state. The investigation of the individual role of CXCL11 in vivo has been hampered as C57BL/6 mice carry several mutations that result in a null allele. Here, CRISPR/Cas9 was used to correct these mutations on a C57BL/6 background. It was validated that CXCL11KI mice expressed CXCL11 protein in dendritic cells, spleen and lung. CXCL11KI mice were largely phenotypically indistinguishable from C57BL/6 mice, both at steady‐state and during two models of viral infection. While CXCL11 expression did not modify acute antiviral responses, this study provides a new tool to understand the role of CXCL11 in other experimental settings.
Collapse
Affiliation(s)
- Lennard Dalit
- Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia
- Department of Medical Biology University of Melbourne Parkville VIC Australia
| | - Carolina Alvarado
- Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia
| | - Lisan Küijper
- Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia
| | - Andrew J Kueh
- Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia
- Department of Medical Biology University of Melbourne Parkville VIC Australia
| | - Ashley Weir
- Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia
- Department of Medical Biology University of Melbourne Parkville VIC Australia
| | - Angela D’Amico
- Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia
| | - Marco J Herold
- Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia
- Department of Medical Biology University of Melbourne Parkville VIC Australia
| | - James E Vince
- Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia
- Department of Medical Biology University of Melbourne Parkville VIC Australia
| | - Stephen L Nutt
- Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia
- Department of Medical Biology University of Melbourne Parkville VIC Australia
| | - Joanna R Groom
- Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia
- Department of Medical Biology University of Melbourne Parkville VIC Australia
| |
Collapse
|