1
|
Georgakis S, Ioannidou K, Mora BB, Orfanakis M, Brenna C, Muller YD, Del Rio Estrada PM, Sharma AA, Pantaleo G, de Leval L, Comte D, Gottardo R, Petrovas C. Cellular and molecular determinants mediating the dysregulated germinal center immune dynamics in systemic lupus erythematosus. Front Immunol 2025; 16:1530327. [PMID: 40070830 PMCID: PMC11894538 DOI: 10.3389/fimmu.2025.1530327] [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: 11/18/2024] [Accepted: 01/17/2025] [Indexed: 03/14/2025] Open
Abstract
Introduction Systemic lupus erythematosus (SLE) is characterized by dysregulated humoral immunity, leading to the generation of autoreactive B cells that can differentiate both within and outside of lymph node (LN) follicles. Methods Here, we employed spatial transcriptomics and multiplex imaging to investigate the follicular immune landscaping and the in situ transcriptomic profile in LNs from SLE individuals. Results Our spatial transcriptomic analysis revealed robust type I IFN and plasma cell signatures in SLE compared to reactive, control follicles. Cell deconvolution revealed that follicular T cell subsets are mainly affected by the type I IFN fingerprint of SLE follicles. Dysregulation of TFH differentiation was documented by i) the significant reduction of Bcl6hi TFH cells, ii) the reduced cell density of potential IL-4 producing TFH cell subsets associated with the impaired transcriptomic signature of follicular IL-4 signaling and iii) the loss of their correlation with GC-B cells. This profile was accompanied by a marked reduction of Bcl6hi B cells and an enrichment of extrafollicular CD19hiCD11chiTbethi, age-associated B cells (ABCs), known for their autoreactive potential. The increased prevalence of follicular IL-21hi cells further reveals a hyperactive microenvironment in SLE compared to control. Discussion Taken together, our findings highlight the altered immunological landscape of SLE follicles, likely fueled by potent inflammatory signals such as sustained type I IFN and/or IL-21 signaling. Our work provides novel insights into the spatial molecular and cellular signatures of SLE follicular B and TFH cell dynamics, and points to druggable targets to restore immune tolerance and enhance vaccine responses in SLE patients.
Collapse
Affiliation(s)
- Spiros Georgakis
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Kalliopi Ioannidou
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Bernat Bramon Mora
- Biomedical Data Science Center, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Michail Orfanakis
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Cloe Brenna
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Yannick D. Muller
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Perla M. Del Rio Estrada
- Pathology Advanced Translational Research Unit, Department of Pathology, Emory University School of Medicine, Atlanta, GA, United States
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Ashish A. Sharma
- Pathology Advanced Translational Research Unit, Department of Pathology, Emory University School of Medicine, Atlanta, GA, United States
| | - Giuseppe Pantaleo
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Laurence de Leval
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Denis Comte
- Service of Internal Medicine, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Raphael Gottardo
- Biomedical Data Science Center, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
- Swiss Institute for Bioinformatics, Lausanne, Switzerland
| | - Constantinos Petrovas
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| |
Collapse
|
2
|
Hu K, O’Neil TR, Baharlou H, Austin PJ, Karrasch JF, Sarkawt L, Li Y, Bertram KM, Cunningham AL, Patrick E, Harman AN. The spatial biology of HIV infection. PLoS Pathog 2025; 21:e1012888. [PMID: 39854613 PMCID: PMC11760614 DOI: 10.1371/journal.ppat.1012888] [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: 01/26/2025] Open
Abstract
HIV infection implicates a spectrum of tissues in the human body starting with viral transmission in the anogenital tract and subsequently persisting in lymphoid tissues and brain. Though studies using isolated cells have contributed significantly towards our understanding of HIV infection, the tissue microenvironment is characterised by a complex interplay of a range of factors, all of which can influence the course of infection but are otherwise missed in ex vivo studies. To address this knowledge gap, it is necessary to investigate the dynamics of infection and the host immune response in situ using imaging-based approaches. Over the last decade, emerging imaging techniques have continually redefined the limits of detection, both in terms of the scope and the scale of the targets. In doing so, this has opened up new questions that can be answered by in situ studies. This review discusses the high-dimensional imaging modalities that are now available and their application towards understanding the spatial biology of HIV infection.
Collapse
Affiliation(s)
- Kevin Hu
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Thomas R. O’Neil
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Heeva Baharlou
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Paul J. Austin
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Brain and Mind Centre, School of Medical of Medical Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Jackson F. Karrasch
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Brain and Mind Centre, School of Medical of Medical Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Lara Sarkawt
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Yuchen Li
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Kirstie M. Bertram
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Anthony L. Cunningham
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Ellis Patrick
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Mathematics and Statistics, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Andrew N. Harman
- The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
3
|
Zaman F, Smith ML, Balagopal A, Durand CM, Redd AD, Tobian AAR. Spatial technologies to evaluate the HIV-1 reservoir and its microenvironment in the lymph node. mBio 2024; 15:e0190924. [PMID: 39058091 PMCID: PMC11324018 DOI: 10.1128/mbio.01909-24] [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] [Indexed: 07/28/2024] Open
Abstract
The presence of the HIV-1 reservoir, a group of immune cells that contain intact, integrated, and replication-competent proviruses, is a major challenge to cure HIV-1. HIV-1 reservoir cells are largely unaffected by the cytopathic effects of viruses, antiviral immune responses, or antiretroviral therapy (ART). The HIV-1 reservoir is seeded early during HIV-1 infection and augmented during active viral replication. CD4+ T cells are the primary target for HIV-1 infection, and recent studies suggest that memory T follicular helper cells within the lymph node, more precisely in the B cell follicle, harbor integrated provirus, which contribute to viral rebound upon ART discontinuation. The B cell follicle, more specifically the germinal center, possesses a unique environment because of its distinct property of being partly immune privileged, potentially allowing HIV-1-infected cells within the lymph nodes to be protected from CD8+ T cells. This modified immune response in the germinal center of the follicle is potentially explained by the exclusion of CD8+ T cells and the presence of T regulatory cells at the junction of the follicle and extrafollicular region. The proviral makeup of HIV-1-infected cells is similar in lymph nodes and blood, suggesting trafficking between these compartments. Little is known about the cell-to-cell interactions, microenvironment of HIV-1-infected cells in the follicle, and trafficking between the lymph node follicle and other body compartments. Applying a spatiotemporal approach that integrates genomics, transcriptomics, and proteomics to investigate the HIV-1 reservoir and its neighboring cells in the lymph node has promising potential for informing HIV-1 cure efforts.
Collapse
Affiliation(s)
- Fatima Zaman
- Department of
Pathology, Johns Hopkins University School of
Medicine, Baltimore,
Maryland, USA
| | - Melissa L. Smith
- Department of
Biochemistry and Molecular Genetics, University of Louisville School of
Medicine, Louisville,
Kentucky, USA
| | - Ashwin Balagopal
- Division of Infectious
Diseases, Department of Medicine, Johns Hopkins
University, Baltimore,
Maryland, USA
| | - Christine M. Durand
- Division of Infectious
Diseases, Department of Medicine, Johns Hopkins
University, Baltimore,
Maryland, USA
| | - Andrew D. Redd
- Division of Infectious
Diseases, Department of Medicine, Johns Hopkins
University, Baltimore,
Maryland, USA
- Laboratory of
Immunoregulation, National Institute of Allergy and Infectious Diseases,
National Institutes of Health,
Bethesda, Maryland, USA
- Institute of
Infectious Disease and Molecular Medicine, University of Cape
Town, Cape Town,
South Africa
| | - Aaron A. R. Tobian
- Department of
Pathology, Johns Hopkins University School of
Medicine, Baltimore,
Maryland, USA
- Division of Infectious
Diseases, Department of Medicine, Johns Hopkins
University, Baltimore,
Maryland, USA
| |
Collapse
|
4
|
Georgakis S, Orfanakis M, Brenna C, Burgermeister S, Del Rio Estrada PM, González-Navarro M, Torres-Ruiz F, Reyes-Terán G, Avila-Rios S, Luna-Villalobos YA, Chén OY, Pantaleo G, Koup RA, Petrovas C. Follicular Immune Landscaping Reveals a Distinct Profile of FOXP3 hiCD4 hi T Cells in Treated Compared to Untreated HIV. Vaccines (Basel) 2024; 12:912. [PMID: 39204036 PMCID: PMC11359267 DOI: 10.3390/vaccines12080912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 09/03/2024] Open
Abstract
Follicular helper CD4hi T cells (TFH) are a major cellular pool for the maintenance of the HIV reservoir. Therefore, the delineation of the follicular (F)/germinal center (GC) immune landscape will significantly advance our understanding of HIV pathogenesis. We have applied multiplex confocal imaging, in combination with the relevant computational tools, to investigate F/GC in situ immune dynamics in viremic (vir-HIV), antiretroviral-treated (cART HIV) People Living With HIV (PLWH) and compare them to reactive, non-infected controls. Lymph nodes (LNs) from viremic and cART PLWH could be further grouped based on their TFH cell densities in high-TFH and low-TFH subgroups. These subgroups were also characterized by different in situ distributions of PD1hi TFH cells. Furthermore, a significant accumulation of follicular FOXP3hiCD4hi T cells, which were characterized by a low scattering in situ distribution profile and strongly correlated with the cell density of CD8hi T cells, was found in the cART-HIV low-TFH group. An inverse correlation between plasma viral load and LN GrzBhiCD8hi T and CD16hiCD15lo cells was found. Our data reveal the complex GC immune landscaping in HIV infection and suggest that follicular FOXP3hiCD4hi T cells could be negative regulators of TFH cell prevalence in cART-HIV.
Collapse
Affiliation(s)
- Spiros Georgakis
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital, University of Lausanne, Rue du Bugnon 25, CH-1011 Lausanne, Switzerland (M.O.)
| | - Michail Orfanakis
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital, University of Lausanne, Rue du Bugnon 25, CH-1011 Lausanne, Switzerland (M.O.)
| | - Cloe Brenna
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital, University of Lausanne, Rue du Bugnon 25, CH-1011 Lausanne, Switzerland (M.O.)
| | - Simon Burgermeister
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital, University of Lausanne, Rue du Bugnon 25, CH-1011 Lausanne, Switzerland (M.O.)
| | - Perla M. Del Rio Estrada
- Centro de Investigacion en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City 14080, Mexico (M.G.-N.)
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Mauricio González-Navarro
- Centro de Investigacion en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City 14080, Mexico (M.G.-N.)
| | - Fernanda Torres-Ruiz
- Centro de Investigacion en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City 14080, Mexico (M.G.-N.)
| | - Gustavo Reyes-Terán
- Institutos Nacionales de Salud y Hospitales de Alta Especialidad, Secretaría de Salud de México, Mexico City 14610, Mexico
| | - Santiago Avila-Rios
- Centro de Investigacion en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City 14080, Mexico (M.G.-N.)
| | - Yara Andrea Luna-Villalobos
- Centro de Investigacion en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City 14080, Mexico (M.G.-N.)
| | - Oliver Y. Chén
- Department of Laboratory Medicine and Pathology, Faculty of Biology and Medicine, Lausanne University Hospital, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Giuseppe Pantaleo
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, CH-1011 Lausanne, Switzerland
| | - Richard A. Koup
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Constantinos Petrovas
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital, University of Lausanne, Rue du Bugnon 25, CH-1011 Lausanne, Switzerland (M.O.)
| |
Collapse
|
5
|
Moysi E, Sharma AA, O’Dell S, Georgakis S, Del Rio Estrada PM, Torres-Ruiz F, Navarro MG, Villalobos YAL, Rios SA, Reyes-Teran G, Beddall MH, Ko SH, Belinky F, Orfanakis M, de Leval L, Enriquez AB, Buckner CM, Moir S, Doria-Rose N, Boritz E, Mascola JR, Sekaly RP, Koup RA, Petrovas C. Neutralization activity in chronic HIV infection is characterized by a distinct programming of follicular helper CD4 T cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.31.605954. [PMID: 39131331 PMCID: PMC11312598 DOI: 10.1101/2024.07.31.605954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
A subset of people living with HIV (PLWH) can produce broadly neutralizing antibodies (bNAbs) against HIV, but the lymph node (LN) dynamics that promote the generation of these antibodies are poorly understood. Here, we explored LN-associated histological, immunological, and virological mechanisms of bNAb generation in a cohort of anti-retroviral therapy (ART)-naïve PLWH. We found that participants who produce bNAbs, termed neutralizers, have a superior LN-associated B cell follicle architecture compared with PLWH who do not. The latter was associated with a significantly higher in situ prevalence of Bcl-6hi follicular helper CD4 T cells (TFH), expressing a molecular program that favors their differentiation and stemness, and significantly reduced IL-10 follicular suppressor CD4 T cells. Furthermore, our data reveal possible molecular targets mediating TFH- B cell interactions in neutralizers. Together, we identify cellular and molecular mechanisms that contribute to the development of bNAbs in PLWH.
Collapse
Affiliation(s)
- Eirini Moysi
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Ashish A. Sharma
- Pathology Advanced Translational Research Unit, Department of Pathology, Emory University School of Medicine, Atlanta, GA, USA
| | - Sijy O’Dell
- Virology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Spiros Georgakis
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Perla Mariana Del Rio Estrada
- Pathology Advanced Translational Research Unit, Department of Pathology, Emory University School of Medicine, Atlanta, GA, USA
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Fernanda Torres-Ruiz
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Mauricio González Navarro
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City, Mexico, Subdireccion de Otorrinolaringologia, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarra Ibarra”
| | - Yara Andrea Luna Villalobos
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Santiago Avila Rios
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Gustavo Reyes-Teran
- Institutos Nacionales de Salud y Hospitales de Alta Especialidad, Secretaría de Salud de México, Mexico City, Mexico
| | - Margaret H. Beddall
- ImmunoTechnology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Sung-Hee Ko
- Virus Persistence and Dynamics Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Frida Belinky
- Virus Persistence and Dynamics Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Michail Orfanakis
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Laurence de Leval
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Ana B. Enriquez
- Pathology Advanced Translational Research Unit, Department of Pathology, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Susan Moir
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, MD, USA
| | - Nicole Doria-Rose
- Virology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Eli Boritz
- Virus Persistence and Dynamics Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - John R. Mascola
- Virology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
- ModeX Therapeutics, Weston, MA, USA
| | - Rafick-Pierre Sekaly
- Pathology Advanced Translational Research Unit, Department of Pathology, Emory University School of Medicine, Atlanta, GA, USA
| | - Richard A. Koup
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Constantinos Petrovas
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
6
|
Orfanakis M, Molyvdas A, Petrovas C. In Situ Characterization of Human Follicular Helper CD4 T Cells. Methods Mol Biol 2024; 2813:281-293. [PMID: 38888784 DOI: 10.1007/978-1-0716-3890-3_18] [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] [Indexed: 06/20/2024]
Abstract
The development of an effective humoral response to pathogens and immunogens is a multiphase biological process, which is mediated by the coordinated function of specialized immune cell types in secondary lymphoid organs and particularly in T cell and follicular areas. More specifically, within the follicular/germinal center area, the orchestrated interplay between B cells, follicular helper CD4 T cells (Tfh), and stromal cells triggers a cascade of immune reactions leading to the development of memory B cells and plasma cells able to generate effective, antigen-specific antibodies. The role of Tfh cells in this process is critical. Given the need for vaccines capable to induce antibodies of high affinity, neutralizing activity, and durability, understanding the cellular and molecular mechanisms regulating Tfh cell development is of great importance. Here, we describe novel approaches for the comprehensive understanding of these cells and possible implications for future studies in vaccine development and the understanding of the pathogenesis of relevant diseases.
Collapse
Affiliation(s)
- Michail Orfanakis
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Adam Molyvdas
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Constantinos Petrovas
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland.
| |
Collapse
|
7
|
Hegewisch-Solloa E, Nalin AP, Freud AG, Mace EM. Deciphering the localization and trajectory of human natural killer cell development. J Leukoc Biol 2023; 114:487-506. [PMID: 36869821 DOI: 10.1093/jleuko/qiad027] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/07/2023] [Accepted: 02/12/2023] [Indexed: 03/05/2023] Open
Abstract
Innate immune cells represent the first line of cellular immunity, comprised of both circulating and tissue-resident natural killer cells and innate lymphoid cells. These innate lymphocytes arise from a common CD34+ progenitor that differentiates into mature natural killer cells and innate lymphoid cells. The successive stages in natural killer cell maturation are characterized by increased lineage restriction and changes to phenotype and function. Mechanisms of human natural killer cell development have not been fully elucidated, especially the role of signals that drive the spatial localization and maturation of natural killer cells. Cytokines, extracellular matrix components, and chemokines provide maturation signals and influence the trafficking of natural killer cell progenitors to peripheral sites of differentiation. Here we present the latest advances in our understanding of natural killer and innate lymphoid cell development in peripheral sites, including secondary lymphoid tissues (i.e. tonsil). Recent work in the field has provided a model for the spatial distribution of natural killer cell and innate lymphoid cell developmental intermediates in tissue and generated further insights into the developmental niche. In support of this model, future studies using multifaceted approaches seek to fully map the developmental trajectory of human natural killer cells and innate lymphoid cells in secondary lymphoid tissues.
Collapse
Affiliation(s)
- Everardo Hegewisch-Solloa
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, 630 W 168th St. New York, NY 10032, USA
| | - Ansel P Nalin
- Biomedical Sciences Graduate Program, Medical Scientist Training Program, Comprehensive Cancer Center and The James Cancer Hospital and Solove Research Institute, The Ohio State University, 460 W 10th Ave. Columbus, OH 43210, USA
| | - Aharon G Freud
- Department of Pathology, Comprehensive Cancer Center and The James Cancer Hospital and Solove Research Institute, The Ohio State University, 460 W 12th Ave. Columbus, OH 43210, USA
| | - Emily M Mace
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, 630 W 168th St. New York, NY 10032, USA
| |
Collapse
|
8
|
Hegewisch-Solloa E, Melsen JE, Ravichandran H, Rendeiro AF, Freud AG, Mundy-Bosse B, Melms JC, Eisman SE, Izar B, Grunstein E, Connors TJ, Elemento O, Horowitz A, Mace EM. Mapping human natural killer cell development in pediatric tonsil by imaging mass cytometry and high-resolution microscopy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.05.556371. [PMID: 37732282 PMCID: PMC10508773 DOI: 10.1101/2023.09.05.556371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Natural killer (NK) cells develop from CD34+ progenitors in a stage-specific manner defined by changes in cell surface receptor expression and function. Secondary lymphoid tissues, including tonsil, are sites of human NK cell development. Here we present new insights into human NK cell development in pediatric tonsil using cyclic immunofluorescence and imaging mass cytometry. We show that NK cell subset localization and interactions are dependent on NK cell developmental stage and tissue residency. NK cell progenitors are found in the interfollicular domain in proximity to cytokine-expressing stromal cells that promote proliferation and maturation. Mature NK cells are primarily found in the T-cell rich parafollicular domain engaging in cell-cell interactions that differ depending on their stage and tissue residency. The presence of local inflammation results in changes in NK cell interactions, abundance, and localization. This study provides the first comprehensive atlas of human NK cell development in secondary lymphoid tissue.
Collapse
Affiliation(s)
- Everardo Hegewisch-Solloa
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York NY 10032
| | - Janine E Melsen
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
- Laboratory for Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Hiranmayi Ravichandran
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, 10065
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - André F Rendeiro
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, 10065
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14 AKH BT 25.3, 1090, Vienna, Austria
| | - Aharon G Freud
- Department of Pathology, The Ohio State University, Columbus, OH 43210, USA; Comprehensive Cancer Center and The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH 43210
| | - Bethany Mundy-Bosse
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA; Comprehensive Cancer Center and The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH 43210
| | - Johannes C Melms
- Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center, New York, NY, 10032
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY, 10032
| | - Shira E Eisman
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York NY 10032
| | - Benjamin Izar
- Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center, New York, NY, 10032
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY, 10032
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, 10032
- Program for Mathematical Genomics, Columbia University, New York, NY, 10032
| | - Eli Grunstein
- Department of Otolaryngology - Head and Neck Surgery, Columbia University Medical Center, New York, New York 10032
| | - Thomas J Connors
- Department of Pediatrics, Division of Pediatric Critical Care and Hospital Medicine, Columbia University Irving Medical Center, New York, NY 10024
| | - Olivier Elemento
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10065
| | - Amir Horowitz
- Department of Oncological Sciences, Precision Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029
| | - Emily M Mace
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York NY 10032
| |
Collapse
|
9
|
Oyler BL, Valencia-Dávila JA, Moysi E, Molyvdas A, Ioannidou K, March K, Ambrozak D, De Leval L, Fabozzi G, Woods AS, Koup RA, Petrovas C. Multilevel human secondary lymphoid immune system compartmentalization revealed by complementary imaging approaches. iScience 2023; 26:107261. [PMID: 37520703 PMCID: PMC10371825 DOI: 10.1016/j.isci.2023.107261] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 04/12/2023] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
Secondary human lymphoid tissue immune reactions take place in a highly coordinated environment with compartmentalization representing a fundamental feature of this organization. In situ profiling methodologies are indispensable for the understanding of this compartmentalization. Here, we propose a complementary experimental approach aiming to reveal different aspects of this process. The analysis of human tonsils, using a combination of single cell phenotypic analysis based on flow cytometry and multiplex imaging and mass spectrometry-based methodologies, revealed a compartmentalized organization at the cellular and molecular levels. More specifically, the skewed distribution of highly specialized immune cell subsets and relevant soluble mediators was accompanied by a compartmentalized localization of several lipids across different anatomical areas of the tonsillar tissue. The performance of such combinatorial experimental approaches could lead to the identification of novel in situ interactions and molecular targets for the in vivo manipulation of lymphoid organ, particularly the germinal center, immune reactions.
Collapse
Affiliation(s)
- Benjamin L. Oyler
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | | | - Eirini Moysi
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Adam Molyvdas
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Kalliopi Ioannidou
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Kylie March
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - David Ambrozak
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Laurence De Leval
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Giulia Fabozzi
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Amina S. Woods
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Richard A. Koup
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Constantinos Petrovas
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| |
Collapse
|
10
|
Ozulumba T, Montalbine AN, Ortiz-Cárdenas JE, Pompano RR. New tools for immunologists: models of lymph node function from cells to tissues. Front Immunol 2023; 14:1183286. [PMID: 37234163 PMCID: PMC10206051 DOI: 10.3389/fimmu.2023.1183286] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/20/2023] [Indexed: 05/27/2023] Open
Abstract
The lymph node is a highly structured organ that mediates the body's adaptive immune response to antigens and other foreign particles. Central to its function is the distinct spatial assortment of lymphocytes and stromal cells, as well as chemokines that drive the signaling cascades which underpin immune responses. Investigations of lymph node biology were historically explored in vivo in animal models, using technologies that were breakthroughs in their time such as immunofluorescence with monoclonal antibodies, genetic reporters, in vivo two-photon imaging, and, more recently spatial biology techniques. However, new approaches are needed to enable tests of cell behavior and spatiotemporal dynamics under well controlled experimental perturbation, particularly for human immunity. This review presents a suite of technologies, comprising in vitro, ex vivo and in silico models, developed to study the lymph node or its components. We discuss the use of these tools to model cell behaviors in increasing order of complexity, from cell motility, to cell-cell interactions, to organ-level functions such as vaccination. Next, we identify current challenges regarding cell sourcing and culture, real time measurements of lymph node behavior in vivo and tool development for analysis and control of engineered cultures. Finally, we propose new research directions and offer our perspective on the future of this rapidly growing field. We anticipate that this review will be especially beneficial to immunologists looking to expand their toolkit for probing lymph node structure and function.
Collapse
Affiliation(s)
- Tochukwu Ozulumba
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
| | - Alyssa N. Montalbine
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, United States
| | - Jennifer E. Ortiz-Cárdenas
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Department of Bioengineering, Stanford University, Stanford, CA, United States
| | - Rebecca R. Pompano
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
- Carter Immunology Center and University of Virginia (UVA) Cancer Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
11
|
Le Coz C, Oldridge DA, Herati RS, De Luna N, Garifallou J, Cruz Cabrera E, Belman JP, Pueschl D, Silva LV, Knox AVC, Reid W, Yoon S, Zur KB, Handler SD, Hakonarson H, Wherry EJ, Gonzalez M, Romberg N. Human T follicular helper clones seed the germinal center-resident regulatory pool. Sci Immunol 2023; 8:eade8162. [PMID: 37027481 PMCID: PMC10329285 DOI: 10.1126/sciimmunol.ade8162] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 03/16/2023] [Indexed: 04/09/2023]
Abstract
The mechanisms by which FOXP3+ T follicular regulatory (Tfr) cells simultaneously steer antibody formation toward microbe or vaccine recognition and away from self-reactivity remain incompletely understood. To explore underappreciated heterogeneity in human Tfr cell development, function, and localization, we used paired TCRVA/TCRVB sequencing to distinguish tonsillar Tfr cells that are clonally related to natural regulatory T cells (nTfr) from those likely induced from T follicular helper (Tfh) cells (iTfr). The proteins iTfr and nTfr cells differentially expressed were used to pinpoint their in situ locations via multiplex microscopy and establish their divergent functional roles. In silico analyses and in vitro tonsil organoid tracking models corroborated the existence of separate Treg-to-nTfr and Tfh-to-iTfr developmental trajectories. Our results identify human iTfr cells as a distinct CD38+, germinal center-resident, Tfh-descended subset that gains suppressive function while retaining the capacity to help B cells, whereas CD38- nTfr cells are elite suppressors primarily localized in follicular mantles. Interventions differentially targeting specific Tfr cell subsets may provide therapeutic opportunities to boost immunity or more precisely treat autoimmune diseases.
Collapse
Affiliation(s)
- Carole Le Coz
- Division of Immunology and Allergy, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Derek A. Oldridge
- Center for Computational and Genomic Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, Philadelphia, PA
| | - Ramin S. Herati
- Department of Medicine, NYU Grossman School of Medicine, New York, NY
| | - Nina De Luna
- Division of Immunology and Allergy, Children’s Hospital of Philadelphia, Philadelphia, PA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - James Garifallou
- Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Emylette Cruz Cabrera
- Division of Immunology and Allergy, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Jonathan P Belman
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, Philadelphia, PA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Dana Pueschl
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Luisa V. Silva
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Ainsley V. C. Knox
- Division of Immunology and Allergy, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Whitney Reid
- Division of Immunology and Allergy, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Samuel Yoon
- Division of Immunology and Allergy, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Karen B. Zur
- Pediatric Otolaryngology, Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Otolaryngology: Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Steven D. Handler
- Pediatric Otolaryngology, Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Otolaryngology: Head and Neck Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Hakon Hakonarson
- Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics, Perelman School of Medicine, Philadelphia, PA
| | - E. John Wherry
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Michael Gonzalez
- Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA
- Center for Cytokine Storm Treatment & Laboratory, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Neil Romberg
- Division of Immunology and Allergy, Children’s Hospital of Philadelphia, Philadelphia, PA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Pediatrics, Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
12
|
Noto A, Suffiotti M, Joo V, Mancarella A, Procopio FA, Cavet G, Leung Y, Corpataux JM, Cavassini M, Riva A, Stamatatos L, Gottardo R, McDermott AB, Koup RA, Fenwick C, Perreau M, Pantaleo G. The deficiency in Th2-like Tfh cells affects the maturation and quality of HIV-specific B cell response in viremic infection. Front Immunol 2022; 13:960120. [PMID: 36091040 PMCID: PMC9450063 DOI: 10.3389/fimmu.2022.960120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Optimal T follicular helper (Tfh) cells function is important to promote the development of germinal centers and maturation of high affinity antigen-specific B cells. We have found that the expression of CXCR3 defines distinct Tfh subsets: CXCR3+ Th1-like Tfh cells mainly producing single IFN-γ and dual IL-21/IFN-γ and CXCR3- Th2-like Tfh cells mainly producing single IL-4 and dual IL-21/IL-4 cytokines. CXCR3- Th2-like Tfhs are significantly reduced during ongoing HIV replication. While the percentage of Th2-like Tfh cells correlates with that of total and cycling HIV-specific B cells, the percentage of CXCR3+ Th1-like Tfhs correlates with HIV-specific B cells expressing T-bet and CXCR3. Of note, only IL-4 and IL-21 cytokines boosted efficient maturation of HIV-specific B cells while IFN-γ induced expression of T-bet and CXCR3 in B cells. Interestingly, total and HIV-specific CXCR3+ B cells showed lower rate of somatic hypermutation, as compared to CXCR3- B cells. Therefore, the imbalance in Th2/Th1-like Tfhs affects B cell responses in viremic HIV infection.
Collapse
Affiliation(s)
- Alessandra Noto
- Service Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Madeleine Suffiotti
- Service Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Victor Joo
- Service Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Antonio Mancarella
- Service Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Francesco A. Procopio
- Service Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Guy Cavet
- Atreca, Redwood City, CA, United States
| | | | - Jean-Marc Corpataux
- Service of Vascular Surgery, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Matthias Cavassini
- Service of Infectious Diseases, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Agostino Riva
- Division of Infectious Diseases, Luigi Sacco Hospital, University of Milan, Milan, Italy
| | - Leonidas Stamatatos
- Department of Global Health, Seattle University of Washington, Seattle, WA, United States
| | - Raphael Gottardo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Adrian B. McDermott
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Richard A. Koup
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Craig Fenwick
- Service Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Matthieu Perreau
- Service Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Giuseppe Pantaleo
- Service Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland,Swiss Vaccine Research Institute, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland,*Correspondence: Giuseppe Pantaleo,
| |
Collapse
|
13
|
Peluso MJ, Donatelli J, Henrich TJ. Long-term immunologic effects of SARS-CoV-2 infection: leveraging translational research methodology to address emerging questions. Transl Res 2022; 241:1-12. [PMID: 34780969 PMCID: PMC8588584 DOI: 10.1016/j.trsl.2021.11.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/02/2021] [Accepted: 11/05/2021] [Indexed: 11/01/2022]
Abstract
The current era of COVID-19 is characterized by emerging variants of concern, waning vaccine- and natural infection-induced immunity, debate over the timing and necessity of vaccine boosting, and the emergence of post-acute sequelae of SARS-CoV-2 infection. As a result, there is an ongoing need for research to promote understanding of the immunology of both natural infection and prevention, especially as SARS-CoV-2 immunology is a rapidly changing field, with new questions arising as the pandemic continues to grow in complexity. The next phase of COVID-19 immunology research will need focus on clearer characterization of the immune processes defining acute illness, development of a better understanding of the immunologic processes driving protracted symptoms and prolonged recovery (ie, post-acute sequelae of SARS-CoV-2 infection), and a growing focus on the impact of therapeutic and prophylactic interventions on the long-term consequences of SARS-CoV-2 infection. In this review, we address what is known about the long-term immune consequences of SARS-CoV-2 infection and propose how experience studying the translational immunology of other infections might inform the approach to some of the key questions that remain.
Collapse
Affiliation(s)
- Michael J Peluso
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, California
| | - Joanna Donatelli
- Division Experimental Medicine, University of California, San Francisco, California
| | - Timothy J Henrich
- Division Experimental Medicine, University of California, San Francisco, California.
| |
Collapse
|
14
|
Moysi E, Paris RM, Le Grand R, Koup RA, Petrovas C. Human lymph node immune dynamics as driver of vaccine efficacy: an understudied aspect of immune responses. Expert Rev Vaccines 2022; 21:633-644. [PMID: 35193447 DOI: 10.1080/14760584.2022.2045198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION During the last century, changes in hygiene, sanitation, and the advent of childhood vaccination have resulted in profound reductions in mortality from infectious diseases. Despite this success, infectious diseases remain an enigmatic public health threat, where effective vaccines for influenza, human immunodeficiency virus (HIV), tuberculosis, and malaria, among others remain elusive. AREA COVERED In addition to the immune evasion tactics employed by complex pathogens, our understanding of immunopathogenesis and the development of effective vaccines is also complexified by the inherent variability of human immune responses. Lymph nodes (LNs) are the anatomical sites where B cell responses develop. An important, but understudied component of immune response complexity is variation in LN immune dynamics and in particular variation in germinal center follicular helper T cells (Tfh) and B cells which can be impacted by genetic variation, aging, the microbiome and chronic infection. EXPERT OPINION This review describes the contribution of genetic variation, aging, microbiome and chronic infection on LN immune dynamics and associated Tfh responses and offers perspective on how inclusion of LN immune subset and cytoarchitecture analyses, along with peripheral blood biomarkers can supplement systems vaccinology or immunology approaches for the development of vaccines or other interventions to prevent infectious diseases.
Collapse
Affiliation(s)
- Eirini Moysi
- Tissue Analysis Core, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | | | - Roger Le Grand
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Richard A Koup
- Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Constantinos Petrovas
- Tissue Analysis Core, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA.,Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
15
|
Bošnjak B, Do KTH, Förster R, Hammerschmidt SI. Imaging dendritic cell functions. Immunol Rev 2021; 306:137-163. [PMID: 34859450 DOI: 10.1111/imr.13050] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/16/2021] [Accepted: 11/18/2021] [Indexed: 12/14/2022]
Abstract
Dendritic cells (DCs) are crucial for the appropriate initiation of adaptive immune responses. During inflammation, DCs capture antigens, mature, and migrate to lymphoid tissues to present foreign material to naïve T cells. These cells get activated and differentiate either into pathogen-specific cytotoxic CD8+ T cells that destroy infected cells or into CD4+ T helper cells that, among other effector functions, orchestrate antibody production by B cells. DC-mediated antigen presentation is equally important in non-inflammatory conditions. Here, DCs mediate induction of tolerance by presenting self-antigens or harmless environmental antigens and induce differentiation of regulatory T cells or inactivation of self-reactive immune cells. Detailed insights into the biology of DCs are, therefore, crucial for the development of novel vaccines as well as the prevention of autoimmune diseases. As in many other life science areas, our understanding of DC biology would be extremely restricted without bioimaging, a compilation of methods that visualize biological processes. Spatiotemporal tracking of DCs relies on various imaging tools, which not only enable insights into their positioning and migration within tissues or entire organs but also allow visualization of subcellular and molecular processes. This review aims to provide an overview of the imaging toolbox and to provide examples of diverse imaging techniques used to obtain fundamental insights into DC biology.
Collapse
Affiliation(s)
- Berislav Bošnjak
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Kim Thi Hoang Do
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Hannover, Germany.,Cluster of Excellence RESIST (EXC 2155) Hannover Medical School, Hannover, Germany.,German Centre for Infection Research (DZIF), Hannover, Germany
| | | |
Collapse
|