1
|
Dhenni R, Hoppé AC, Reynaldi A, Kyaw W, Handoko NT, Grootveld AK, Keith YH, Bhattacharyya ND, Ahel HI, Telfser AJ, McCorkindale AN, Yazar S, Bui CHT, Smith JT, Khoo WH, Boyd M, Obeid S, Milner B, Starr M, Brilot F, Milogiannakis V, Akerman A, Aggarwal A, Davenport MP, Deenick EK, Chaffer CL, Croucher PI, Brink R, Goldstein LD, Cromer D, Turville SG, Kelleher AD, Venturi V, Munier CML, Phan TG. Macrophages direct location-dependent recall of B cell memory to vaccination. Cell 2025:S0092-8674(25)00407-6. [PMID: 40300604 DOI: 10.1016/j.cell.2025.04.005] [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: 09/19/2023] [Revised: 08/31/2024] [Accepted: 04/02/2025] [Indexed: 05/01/2025]
Abstract
Vaccines generate long-lived plasma cells and memory B cells (Bmems) that may re-enter secondary germinal centers (GCs) to further mutate their B cell receptor upon boosting and re-exposure to antigen. We show in mouse models that lymph nodes draining the site of primary vaccination harbor a subset of Bmems that reside in the subcapsular niche, generate larger recall responses, and are more likely to re-enter GCs compared with circulating Bmems in non-draining lymph nodes. This location-dependent recall of Bmems into the GC in the draining lymph node was dependent on CD169+ subcapsular sinus macrophages (SSMs) in the subcapsular niche. In human participants, boosting of the BNT162b2 vaccine in the same arm generated more rapid secretion of broadly neutralizing antibodies, GC participation, and clonal expansion of SARS-CoV-2-specific B cells than boosting of the opposite arm. These data reveal an unappreciated role for primed draining lymph node SSMs in Bmem cell fate determination.
Collapse
Affiliation(s)
- Rama Dhenni
- Precision Immunology Program, Garvan Institute of Medical Research, Sydney, NSW, Australia; St. Vincent's Healthcare Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, Sydney, NSW, Australia
| | - Alexandra Carey Hoppé
- Immunovirology and Pathogenesis Program, Kirby Institute, UNSW Sydney, Sydney, NSW, Australia
| | - Arnold Reynaldi
- Infection Analytics Program, Kirby Institute, UNSW Sydney, Sydney, NSW, Australia
| | - Wunna Kyaw
- Precision Immunology Program, Garvan Institute of Medical Research, Sydney, NSW, Australia; St. Vincent's Healthcare Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, Sydney, NSW, Australia
| | - Nathalie Tricia Handoko
- Precision Immunology Program, Garvan Institute of Medical Research, Sydney, NSW, Australia; St. Vincent's Healthcare Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, Sydney, NSW, Australia
| | - Abigail K Grootveld
- Precision Immunology Program, Garvan Institute of Medical Research, Sydney, NSW, Australia; St. Vincent's Healthcare Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, Sydney, NSW, Australia
| | - Yuki Honda Keith
- Precision Immunology Program, Garvan Institute of Medical Research, Sydney, NSW, Australia; St. Vincent's Healthcare Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, Sydney, NSW, Australia
| | - Nayan Deger Bhattacharyya
- Precision Immunology Program, Garvan Institute of Medical Research, Sydney, NSW, Australia; St. Vincent's Healthcare Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, Sydney, NSW, Australia
| | - Holly I Ahel
- Precision Immunology Program, Garvan Institute of Medical Research, Sydney, NSW, Australia; St. Vincent's Healthcare Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, Sydney, NSW, Australia
| | - Aiden Josiah Telfser
- Precision Immunology Program, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Andrew N McCorkindale
- Data Science Platform, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Seyhan Yazar
- Precision Immunology Program, Garvan Institute of Medical Research, Sydney, NSW, Australia; St. Vincent's Healthcare Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, Sydney, NSW, Australia
| | - Christina H T Bui
- St. Vincent's Healthcare Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, Sydney, NSW, Australia; Cancer Plasticity and Dormancy Program, The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - James T Smith
- St. Vincent's Healthcare Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, Sydney, NSW, Australia; Cancer Plasticity and Dormancy Program, The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Weng Hua Khoo
- Cancer Plasticity and Dormancy Program, The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Mollie Boyd
- Immunovirology and Pathogenesis Program, Kirby Institute, UNSW Sydney, Sydney, NSW, Australia
| | - Solange Obeid
- St. Vincent's Hospital Sydney, Sydney, NSW, Australia
| | - Brad Milner
- St. Vincent's Hospital Sydney, Sydney, NSW, Australia
| | - Mitchell Starr
- St. Vincent's Centre for Applied Medical Research, Sydney, NSW, Australia
| | - Fabienne Brilot
- Brain Autoimmunity Group, Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, School of Medical Sciences, Sydney, NSW, Australia; School of Medical Science, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Vanessa Milogiannakis
- Immunovirology and Pathogenesis Program, Kirby Institute, UNSW Sydney, Sydney, NSW, Australia
| | - Anouschka Akerman
- Immunovirology and Pathogenesis Program, Kirby Institute, UNSW Sydney, Sydney, NSW, Australia
| | - Anupriya Aggarwal
- Immunovirology and Pathogenesis Program, Kirby Institute, UNSW Sydney, Sydney, NSW, Australia
| | - Miles P Davenport
- Infection Analytics Program, Kirby Institute, UNSW Sydney, Sydney, NSW, Australia
| | - Elissa K Deenick
- Precision Immunology Program, Garvan Institute of Medical Research, Sydney, NSW, Australia; Immunovirology and Pathogenesis Program, Kirby Institute, UNSW Sydney, Sydney, NSW, Australia
| | - Christine L Chaffer
- St. Vincent's Healthcare Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, Sydney, NSW, Australia; Cancer Plasticity and Dormancy Program, The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Peter I Croucher
- St. Vincent's Healthcare Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, Sydney, NSW, Australia; Cancer Plasticity and Dormancy Program, The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Robert Brink
- Precision Immunology Program, Garvan Institute of Medical Research, Sydney, NSW, Australia; Immune Biotherapies Program, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Leonard D Goldstein
- St. Vincent's Healthcare Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, Sydney, NSW, Australia; Data Science Platform, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Deborah Cromer
- Infection Analytics Program, Kirby Institute, UNSW Sydney, Sydney, NSW, Australia
| | - Stuart G Turville
- Immunovirology and Pathogenesis Program, Kirby Institute, UNSW Sydney, Sydney, NSW, Australia
| | - Anthony D Kelleher
- Immunovirology and Pathogenesis Program, Kirby Institute, UNSW Sydney, Sydney, NSW, Australia; St. Vincent's Hospital Sydney, Sydney, NSW, Australia.
| | - Vanessa Venturi
- Immunovirology and Pathogenesis Program, Kirby Institute, UNSW Sydney, Sydney, NSW, Australia.
| | - C Mee Ling Munier
- Immunovirology and Pathogenesis Program, Kirby Institute, UNSW Sydney, Sydney, NSW, Australia.
| | - Tri Giang Phan
- Precision Immunology Program, Garvan Institute of Medical Research, Sydney, NSW, Australia; St. Vincent's Healthcare Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, Sydney, NSW, Australia.
| |
Collapse
|
2
|
Kardava L, Lim J, Buckner CM, Lopes de Assis F, Zhang X, Wang W, Melnyk ML, El Merhebi O, Trihemasava K, Teng IT, Carroll R, Jethmalani Y, Castro M, Lin BC, Praiss LH, Seamon CA, Kwong PD, Koup RA, Serebryannyy L, Nickle DC, Chun TW, Moir S. Phenotypic heterogeneity defines B cell responses to repeated SARS-CoV-2 exposures through vaccination and infection. Cell Rep 2025; 44:115557. [PMID: 40222009 PMCID: PMC12080740 DOI: 10.1016/j.celrep.2025.115557] [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: 09/23/2024] [Revised: 01/29/2025] [Accepted: 03/21/2025] [Indexed: 04/15/2025] Open
Abstract
Long-lived humoral memory is key to durable immunity against pathogens yet remains challenging to define due to heterogeneity among antigen-reactive B cells. We addressed this gap through longitudinal sampling over the course of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mRNA vaccinations with or without breakthrough infection. High-dimensional phenotypic profiling performed on ∼72 million B cells showed that receptor-binding domain (RBD) reactivity was associated with five distinct immunoglobulin G (IgG) B cell populations. Two expressed the activation marker CD71, both correlated with neutralizing antibodies, yet the one lacking the memory marker CD27 was induced by vaccination and blunted by infection. Two were resting memory populations; one lacking CD73 arose early and contributed to cross-reactivity; the other, expressing CD73, arose later and correlated with neutralizing antibodies. The fifth, a rare germinal center-like population, contributed to recall responses and was highly cross reactive. Overall, robust and distinct responses to booster vaccination overcame the superiority of hybrid immunity provided by breakthrough infection.
Collapse
Affiliation(s)
- Lela Kardava
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - James Lim
- Monoceros Biosystems, San Diego, CA 29130, USA
| | - Clarisa M Buckner
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Felipe Lopes de Assis
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Xiaozhen Zhang
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Wei Wang
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Mattie L Melnyk
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Omar El Merhebi
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Krittin Trihemasava
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - I-Ting Teng
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Robin Carroll
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | | | - Mike Castro
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Bob C Lin
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Lauren H Praiss
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Catherine A Seamon
- Critical Care Medicine Department, Clinical Center, NIH, Bethesda, MD 20892, USA
| | - Peter D Kwong
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA; Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Richard A Koup
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | | | - David C Nickle
- Monoceros Biosystems, San Diego, CA 29130, USA; Department of Global Health, University of Washington, Seattle, WA 98105, USA
| | - Tae-Wook Chun
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Susan Moir
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| |
Collapse
|
3
|
White TLA, Henry DJ, Roberts SDA, Jin Y, Gong YN, Morel PA. HnRNP L is essential for peripheral T cell proliferation and survival. Front Immunol 2025; 16:1543145. [PMID: 40276511 PMCID: PMC12018431 DOI: 10.3389/fimmu.2025.1543145] [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: 12/10/2024] [Accepted: 03/14/2025] [Indexed: 04/26/2025] Open
Abstract
Introduction During T cell development, heterogeneous nuclear ribonucleoprotein (hnRNP) L is known to regulate CD4 T helper subset differentiation, the proliferation and migration of thymocytes, as loss of hnRNP L in early T cell development results in a failure of T cells to reach the periphery. Methods To better understand the role of hnRNP L in modulating peripheral T cell trafficking and function, we analyzed T survival and activation in newly generated CD4Cre x hnRNP LFl/Fl (KO) mice. In vitro and in vivo analyses of CD4 T cell differentiation, T cell proliferation and death post activation were performed. Results Our initial study of the steady state profile of the KO mice showed normal migration of T cells from the thymus, but peripheral T cell numbers were reduced. Analysis of TCR-mediated signaling pathways revealed normal early T cell activation. However, T cells lacking hnRNP L had marked defects in their ability to differentiate into T helper cell subsets due to reduced proliferation and increased death. In vivo, using immunization studies, KO CD4 T cells failed to fully differentiate into T follicular helper (Tfh) cells and were unable to support the formation of germinal center B cells. Death of activated hnRNP L KO cells could be reversed by treating the cells with zVAD, a pan-caspase inhibitor. In addition, hnRNP L KO cells failed to upregulate the anti-apoptotic protein Bcl-XL following activation. Discussion These studies suggest that hnRNP L plays an important role in T cell activation and survival. Our studies suggest that hnRNP L plays a critical pro-survival role in activated T cells and that alternative splicing of factors that prevent apoptosis may be an important mechanism by which this is achieved.
Collapse
Affiliation(s)
| | | | | | | | | | - Penelope A. Morel
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
4
|
Neu SD, Gurski CJ, Meinhardt NJ, Jennings KC, Dittel BN. Gut IgA-antibody secreting cells segregate into four Blimp1+ subsets based on differential expression of IgA and Ki-67 and are retained following prolonged αCD20 B cell depletion in mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:780-794. [PMID: 40073093 PMCID: PMC12041773 DOI: 10.1093/jimmun/vkae046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 12/02/2024] [Indexed: 03/14/2025]
Abstract
B cell depletion is an efficacious therapy for multiple sclerosis, but its long-term safety profile in the gastrointestinal tract has not been specifically studied. This is of importance because the gut is the largest reservoir of IgA in the body, which maintains gut homeostasis in part by regulating the composition of the gut microbiota. This was addressed by development of a prolonged B cell depletion model using human CD20 transgenic mice and B cell depletion with the anti-human CD20 antibodies rituximab, a humanized mouse monoclonal, and 2H7, the mouse precursor to ocrelizumab. Both antibodies depleted B cells in the spleen, mesenteric lymph nodes, small intestine, and large intestine, with 2H7 being more efficient. Because gut IgA+ antibody secreting cells (ASC) are poorly defined a flow cytometry strategy was developed using differential expression of IgA and Ki-67 by Blimp1+ cells that identified four IgA-ASC subsets across a developmental spectrum. Neither antibody was efficacious in depleting of any IgA-ASC subset in the intestines. Consequently, fecal IgA levels and percentage of IgA-bound fecal microbes were unaltered. Cumulatively, these studies demonstrate that prolonged B cell-depletion did not substantially impact IgA levels nor overall gut health, providing important insight into the safety profile of B cell depletion drugs.
Collapse
Affiliation(s)
- Savannah D Neu
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
- Versiti Blood Research Institute, Milwaukee, WI, United States
| | - Cody J Gurski
- Versiti Blood Research Institute, Milwaukee, WI, United States
| | - Nathan J Meinhardt
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
- Versiti Blood Research Institute, Milwaukee, WI, United States
| | - Kevin C Jennings
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Bonnie N Dittel
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
- Versiti Blood Research Institute, Milwaukee, WI, United States
| |
Collapse
|
5
|
Castañeda J, Poblete L, Rosemblatt MV, Sauma D, Rosemblatt M, Bono MR, Nuñez S. Early-life homeostatic differentiation of thymus-resident B cells into memory B cells. Front Immunol 2025; 16:1567788. [PMID: 40226628 PMCID: PMC11985456 DOI: 10.3389/fimmu.2025.1567788] [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: 01/28/2025] [Accepted: 03/12/2025] [Indexed: 04/15/2025] Open
Abstract
The thymus contains various antigen-presenting cells, including B cells, which remain activated even under steady-state conditions, suggesting ongoing local stimulation. In this study, we identify class-switched memory B cells in the thymus. Some of these cells switch their immunoglobulin to IgG2b and IgA, and express typical memory markers CD73 and PD-L2. Memory B cell differentiation in the thymus begins in neonatal mice, preceding the appearance of class-switched B cells in other peripheral lymphoid organs. Notably, exposure to environmental antigens does not influence their differentiation. Additionally, cognate interaction with CD4+ positive thymocytes is crucial for the development of memory B cells in the thymus. Our findings demonstrate that the thymus supports the local differentiation of memory B cells through a steady-state process, independent of foreign antigen stimulation and driven by interactions with developing T cells.
Collapse
Affiliation(s)
- Justine Castañeda
- Escuela de Postgrado, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Lilian Poblete
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Mariana V. Rosemblatt
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Los Leones, Santiago, Chile
| | - Daniela Sauma
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
- Centro Ciencia & Vida, Santiago, Chile
| | - Mario Rosemblatt
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Los Leones, Santiago, Chile
- Centro Ciencia & Vida, Santiago, Chile
| | - María Rosa Bono
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
- Centro Ciencia & Vida, Santiago, Chile
| | - Sarah Nuñez
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Sede Los Leones, Santiago, Chile
- Centro Ciencia & Vida, Santiago, Chile
| |
Collapse
|
6
|
Knox JJ, Karolyi K, Monslow J, Cromley D, Rader DJ, Puré E, Cancro MP. T-bet-expressing B cells promote atherosclerosis in apolipoprotein E-deficient mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:vkae027. [PMID: 40073097 PMCID: PMC11952879 DOI: 10.1093/jimmun/vkae027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 11/18/2024] [Indexed: 03/14/2025]
Abstract
The humoral immune system influences the development of atherosclerosis, but the contributions of specific memory B cell subsets and IgG isotypes are poorly understood. We assessed the relationship between atherosclerosis and age-associated B cells (ABCs), a T-bet-expressing memory B cell subset that is enriched for IgG2c production and implicated in humoral autoimmunity. We found increased numbers of splenic CD11c+ ABCs in 6-mo-old, chow-fed Apoe-/- mice versus C57BL/6 control mice, which were exacerbated by high-fat diet. Deletion of T-bet in the B lineage in high-fat diet-fed Apoe-/- mice reduced aortic lesion area, and this correlated with decreased splenic CD11c+ B cells and reduced serum oxidized low-density lipoprotein-specific IgG2c. Our findings suggest that T-bet-expressing B cells are atherogenic agents in the Apoe-/- model and indicate that interventions to inhibit a T-bet-driven humoral response may improve atherosclerotic disease.
Collapse
Affiliation(s)
- James J Knox
- Department of Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Katalin Karolyi
- Department of Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - James Monslow
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Debra Cromley
- Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Daniel J Rader
- Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ellen Puré
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Michael P Cancro
- Department of Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
7
|
Gurski CJ, Hajiyeva Z, Veltri AJ, Fenton K, O'Dell S, Obeidat AZ, Dittel BN. Elevated frequencies of activated memory B cells in multiple sclerosis are reset to healthy control levels after B cell depletion with Ocrelizumab. J Neuroimmunol 2025; 399:578502. [PMID: 39643951 PMCID: PMC12076485 DOI: 10.1016/j.jneuroim.2024.578502] [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: 09/25/2024] [Revised: 11/22/2024] [Accepted: 11/29/2024] [Indexed: 12/09/2024]
Abstract
In multiple sclerosis (MS) the B cell depleting drug ocrelizumab has shown high efficacy in reducing inflammatory activity. Its mechanism of action is unclear due to B cell subset complexity and unknown roles in pathogenesis. Here, we comprehensively phenotyped and quantitated peripheral blood B cell subsets before and after ocrelizumab infusion to gain insight into the fate of B cell subsets with pathogenic potential. Peripheral blood B cells were collected from treatment naïve patients at baseline and months one, three, and six following the first course of ocrelizumab treatment; at 6 months following the second treatment cycle; ∼14 months following their last infusion; and from healthy controls. Flow cytometry combined with cluster analysis was used to track depletion and repletion of naïve, memory, and antibody secreting cells. By month one, naïve B cells were depleted, but a small subset of memory B cells were retained with no depletion of antibody secreting cells. Uniform manifold approximation and projection for dimension reduction analysis of flow cytometry data revealed two non-class switched naïve clusters and two class switched memory clusters. One class switched cluster was activated in MS patients but largely absent in healthy controls. Both memory B cell subsets underwent depletion after a single six-month course of ocrelizumab treatment after which their proportions were reset to heathy control levels. These observations suggest that activated class-switched memory B cells could serve as a biomarker of recent or ongoing MS disease activity to guide redosing.
Collapse
Affiliation(s)
- Cody J Gurski
- Versiti Blood Research Institute, Milwaukee, WI, USA
| | | | | | - Kaylan Fenton
- Department of Neurology, Medical College of Wisconsin, USA
| | | | | | - Bonnie N Dittel
- Versiti Blood Research Institute, Milwaukee, WI, USA; Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
8
|
Zhang W, Cui H, Xu J, Shi M, Bian L, Cui L, Jiang C, Zhang Y. Biodistribution and mechanisms of action of MF59 and MF59-like adjuvants. J Control Release 2025; 378:573-587. [PMID: 39719213 DOI: 10.1016/j.jconrel.2024.12.044] [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: 08/24/2024] [Revised: 11/28/2024] [Accepted: 12/18/2024] [Indexed: 12/26/2024]
Abstract
Recently, adjuvants have received increasing attention as essential vaccine components. Nearly 100 years have passed since Gaston Roman introduced the concept of adjuvants in 1925, during which numerous preclinical and clinical studies related to vaccine adjuvants have been conducted. However, to date, only a few adjuvants have been successfully used in marketed vaccines. This low clinical translational efficiency is mainly owing to the lack of comprehensive and in-depth understanding of the mechanisms of action of adjuvants in complex biological systems. In fact, MF59 is the first non‑aluminum adjuvant applied in approved human vaccines and is still used today; however, many unknowns regarding its mechanism of action remain. Therefore, in this review, the current status of the mechanism of action of MF59 and MF59-like adjuvants is discussed based on evidence-based temporal and spatial processes of immune-regulatory events. Moreover, the key factors of MF59 and MF59-like adjuvants that regulate humoral and cellular immune responses have been summarized. In addition, studies on the distribution and elimination of both antigens and adjuvants were analyzed based on published studies to gain mechanistic insights into the safety of MF59-adjuvanted vaccines. This review will not only benefit future prospects for the development of novel squalene-based adjuvants and their use in clinical applications but will also be valuable for future mechanistic investigations on other vaccine adjuvants.
Collapse
Affiliation(s)
- Wen Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Haicheng Cui
- R&D Center, Changchun BCHT Biotechnology Co., Changchun 130012, China
| | - Jixuan Xu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Mingze Shi
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Lijun Bian
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Lili Cui
- Beijing Research Center, CSPC Pharmaceutical Group Co., Ltd., Beijing 102629, China.
| | - Chunlai Jiang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China; Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China; NMPA Key Laboratory of Humanized Animal Models for Evaluation of Vaccines and Cell Therapy Products, Jilin University, Changchun 130012, China
| | - Yong Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China; Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China; NMPA Key Laboratory of Humanized Animal Models for Evaluation of Vaccines and Cell Therapy Products, Jilin University, Changchun 130012, China.
| |
Collapse
|
9
|
Mima A, Kimura A, Ito R, Hatano Y, Tsujimoto H, Mae SI, Yamane J, Fujibuchi W, Uza N, Toyoda T, Seno H, Osafune K. Mechanistic elucidation of human pancreatic acinar development using single-cell transcriptome analysis on a human iPSC differentiation model. Sci Rep 2025; 15:4668. [PMID: 39920294 PMCID: PMC11806057 DOI: 10.1038/s41598-025-88690-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 01/30/2025] [Indexed: 02/09/2025] Open
Abstract
Few effective treatments have been developed for intractable pancreatic exocrine disorders due to the lack of suitable disease models using human cells. Pancreatic acinar cells differentiated from human induced pluripotent stem cells (hiPSCs) have the potential to solve this issue. In this study, we aimed to elucidate the developmental mechanisms of pancreatic exocrine acinar lineages to establish a directed differentiation method for pancreatic acinar cells from hiPSCs. hiPSC-derived pancreatic endoderm cells were spontaneously differentiated into both pancreatic exocrine and endocrine tissues by implantation into the renal subcapsular space of NOD/SCID mice. Single-cell RNA-seq analysis of the retrieved grafts confirmed the differentiation of pancreatic acinar lineage cells and identified REG4 as a candidate marker for pancreatic acinar progenitor cells. Furthermore, differential gene expression analysis revealed upregulated pathways, including cAMP-related signals, involved in the differentiation of hiPSC-derived pancreatic acinar lineage cells in vivo, and we found that a cAMP activator, forskolin, facilitates the differentiation from hiPSC-derived pancreatic endoderm into pancreatic acinar progenitor cells in our in vitro differentiation culture. Therefore, this platform contributes to our understanding of the developmental mechanisms of pancreatic acinar lineage cells and the establishment of differentiation methods for acinar cells from hiPSCs.
Collapse
Affiliation(s)
- Atsushi Mima
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
- Department of Gastroenterology and Hepatology, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Azuma Kimura
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
- Rege Nephro Co., Ltd., Med-Pharm Collaboration Building, Kyoto University, 46-29 Yoshidashimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Ryo Ito
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yu Hatano
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hiraku Tsujimoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
- Rege Nephro Co., Ltd., Med-Pharm Collaboration Building, Kyoto University, 46-29 Yoshidashimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Shin-Ichi Mae
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Junko Yamane
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Wataru Fujibuchi
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Norimitsu Uza
- Department of Gastroenterology and Hepatology, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Taro Toyoda
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kenji Osafune
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
10
|
Zheng H, Xu S, Yang R, Jiao WE, Qiao YL, Liu JY, Fan HM, Zhou YT, Ni HF, Chen J, Deng YQ, Chen SM. Changes in and Potential Mechanisms of Circulating IgA+CD27-Class-Switched Memory B Cells in Patients With Allergic Rhinitis. J Asthma Allergy 2025; 18:69-83. [PMID: 39867643 PMCID: PMC11766316 DOI: 10.2147/jaa.s501775] [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: 10/19/2024] [Accepted: 01/08/2025] [Indexed: 01/28/2025] Open
Abstract
Background The role of memory B cells and their subgroups in allergic rhinitis (AR) and allergen immunotherapy (AIT) remains unclear. This study aimed to investigate the characteristics of memory B cells in the circulation of patients with AR and those undergoing AIT, as well as their clinical significance. Methods This study involved a cohort comprising 32 healthy control subjects, 39 individuals diagnosed with AR, and 31 AR patients who had received AIT for over one year. Visual analog scale (VAS) scores were used for symptom assessment, and the serum concentrations of immunoglobulins and cytokines were quantified. This study evaluated alterations in the proportions of peripheral blood memory B cells and their subpopulations, plasma cells, and various T-cell subsets across the three participant groups. Results The proportion of IgA+CD27- class-switched memory B cells in the AR group significantly decreased compared to the control group, but significantly increased following AIT (P < 0.05). In AR patients, circulating IgA+CD27- class-switched memory B cells were significantly positively correlated with Treg cells, IL-10, and IL-4 and significantly negatively correlated with IFN-γ, total IgE, sIgE, and VAS scores (P < 0.05). After AIT, the number of circulating IgA+CD27- class-switched memory B cells in AR patients was significantly positively correlated with the number of Treg cells and IL-10 and significantly negatively correlated with the VAS score (P < 0.05). Conclusion The IgA+CD27- class-switched memory cell subset in human peripheral blood may serve as a potential biomarker for evaluating AR symptoms and treatment efficacy. Its mechanism may be associated with interactions between T and B cells.
Collapse
Affiliation(s)
- Han Zheng
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
| | - Shan Xu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
| | - Rui Yang
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
| | - Wo-Er Jiao
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
| | - Yue-Long Qiao
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Jia-Yu Liu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
| | - Hui-Ming Fan
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
| | - Yan-Ting Zhou
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
| | - Hai-Feng Ni
- Department of Otolaryngology-Head and Neck Surgery, Zhejiang Hospital, Hangzhou, Zhejiang, People’s Republic of China
| | - Jin Chen
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
| | - Yu-Qin Deng
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
| | - Shi-Ming Chen
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People’s Republic of China
| |
Collapse
|
11
|
Athavale A, Gaur A, Ahmed N, Subramaniam A, Dandotiya J, Raj S, Upadhyay SK, Samal S, Pandey AK, Rai RC, Awasthi A. Receptor Binding Domain-Specific B Cell Memory Responses Among Individuals Vaccinated Against SARS-CoV-2. Vaccines (Basel) 2024; 12:1396. [PMID: 39772064 PMCID: PMC11680197 DOI: 10.3390/vaccines12121396] [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: 09/19/2024] [Revised: 10/14/2024] [Accepted: 10/22/2024] [Indexed: 01/11/2025] Open
Abstract
Background: The COVID-19 pandemic prompted unprecedented vaccine development efforts against SARS-CoV-2. India, which was one of the countries most impacted by COVID-19, developed its indigenous vaccine in addition to utilizing the ones developed by other countries. While antibody levels and neutralizing antibody titres are considered initial correlates of immune protection, long-term protection from the pathogen relies on memory B and T cells and their recall responses. In this regard, global research has primarily focused on mRNA-based vaccines. The studies on immune memory response, particularly B cell memory response induced by the vaccines given to Indians, remain relatively obscure. Methods: We assessed Receptor Binding Domain-specific memory B cells in the peripheral circulation and their ability to secrete antigen-specific antibodies among Indians vaccinated with Covaxin (BBV152), Covishield (AZD1222), Corbevax (BECOV2D), and Sputnik Light, as well as unvaccinated individuals. Results: Corbevax and Sputnik Light conferred better antibody-secreting cell (ASC) responses over time compared to other groups. Conclusions: These findings contribute to our understanding of vaccine-induced immune memory in the Indian population; providing insights that could inform future vaccine strategies.
Collapse
Affiliation(s)
- Atharv Athavale
- BRIC-Translational Health Science and Technology Institute, Faridabad 21001, India; (A.A.); (A.G.); (N.A.); (A.S.); (J.D.); (S.R.); (S.S.)
| | - Anmol Gaur
- BRIC-Translational Health Science and Technology Institute, Faridabad 21001, India; (A.A.); (A.G.); (N.A.); (A.S.); (J.D.); (S.R.); (S.S.)
| | - Nafees Ahmed
- BRIC-Translational Health Science and Technology Institute, Faridabad 21001, India; (A.A.); (A.G.); (N.A.); (A.S.); (J.D.); (S.R.); (S.S.)
| | - Adarsh Subramaniam
- BRIC-Translational Health Science and Technology Institute, Faridabad 21001, India; (A.A.); (A.G.); (N.A.); (A.S.); (J.D.); (S.R.); (S.S.)
| | - Jyotsna Dandotiya
- BRIC-Translational Health Science and Technology Institute, Faridabad 21001, India; (A.A.); (A.G.); (N.A.); (A.S.); (J.D.); (S.R.); (S.S.)
| | - Sneha Raj
- BRIC-Translational Health Science and Technology Institute, Faridabad 21001, India; (A.A.); (A.G.); (N.A.); (A.S.); (J.D.); (S.R.); (S.S.)
| | | | - Sweety Samal
- BRIC-Translational Health Science and Technology Institute, Faridabad 21001, India; (A.A.); (A.G.); (N.A.); (A.S.); (J.D.); (S.R.); (S.S.)
| | | | - Ramesh Chandra Rai
- BRIC-Translational Health Science and Technology Institute, Faridabad 21001, India; (A.A.); (A.G.); (N.A.); (A.S.); (J.D.); (S.R.); (S.S.)
| | - Amit Awasthi
- BRIC-Translational Health Science and Technology Institute, Faridabad 21001, India; (A.A.); (A.G.); (N.A.); (A.S.); (J.D.); (S.R.); (S.S.)
| |
Collapse
|
12
|
Sen Chaudhuri A, Sun J. Lung-resident lymphocytes and their roles in respiratory infections and chronic respiratory diseases. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2024; 2:214-223. [PMID: 39834580 PMCID: PMC11742555 DOI: 10.1016/j.pccm.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Indexed: 01/22/2025]
Abstract
Recent scientific breakthroughs have blurred traditional boundaries between innate and adaptive immunity, revealing a sophisticated network of tissue-resident cells that deliver immediate, localized immune responses. These lymphocytes not only provide rapid frontline defense but also present a paradoxical role in the pathogenesis of respiratory diseases such as asthma, chronic obstructive pulmonary disease, pulmonary fibrosis, and the long-term tissue consequences of viral infections including severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). This review traverses the intricate landscape of lung-resident lymphocytes, delving into their origins, diverse functions, and their dualistic impact on pulmonary health. We dissect their interactions with the microenvironment and the regulatory mechanisms guiding their activity, with an emphasis on their contribution to both immune protection and immunopathology. This review aims to elucidate the complex narrative of these cells, enhancing our understanding of the development of precise therapeutic strategies to combat acute and chronic pulmonary diseases. Through this exploration, the review aspires to shed light on the potential of harnessing lung-resident lymphocytes for the treatment of respiratory conditions.
Collapse
Affiliation(s)
- Arka Sen Chaudhuri
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Jie Sun
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
13
|
Petriv N, Suo H, Hochnadel I, Timrott K, Bondarenko N, Neubert L, Reinhard E, Jedicke N, Kaufhold P, Guzmán CA, Lichtinghagen R, Manns MP, Bantel H, Yevsa T. Essential roles of B cell subsets in the progression of MASLD and HCC. JHEP Rep 2024; 6:101189. [PMID: 39611128 PMCID: PMC11602976 DOI: 10.1016/j.jhepr.2024.101189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 08/05/2024] [Accepted: 08/10/2024] [Indexed: 11/30/2024] Open
Abstract
Background & Aims Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related death. Metabolic dysfunction-associated steatotic liver disease (MASLD) is a significant cause of HCC. Current treatment options for HCC are very limited. Recent evidence highlights B cells as key drivers in MASLD progression toward HCC. However, it remains unclear whether multiple B cell populations or a distinct B cell subset regulates inflammatory responses during liver disease progression. The scope of this study was to define protumorigenic B cell subsets in MASLD and HCC. Methods Multicolor flow cytometry, immunohistochemistry, and immunofluorescence analyses were performed to investigate B cell populations locally (in liver tissue) and systemically (in the blood) in mice with MASLD (n = 6) and HCC (n = 5-6). The results obtained in mice were also verified in patients with MASLD (n = 19) and HCC (n = 16). Results Our study revealed an increase of two regulatory B cell (Breg) subsets, CD19+B220+CD5+CD1d+ (p <0.0001) and CD19-B220+CD5+CD1d- (p <0.0001), both of which highly overexpress IgM/IgD, PD-L1, and IL-10, in the livers of mice with MASLD and HCC. Furthermore, we showed that B-cell depletion therapy in combination with a Listeria-based vaccine decreased CD19-B220+CD5+CD1d- Bregs (p = 0.0103), and improved survival of mice with HCC. We also found CD19+CD5+IL-10+ (p = 0.0167), CD19+CD5+PD-L1+ (p = 0.0333) and CD19+CD5+IgM+IgD+ (p = 0.0317) B cells in human HCCs. In addition, strong overexpression of IgM/IgD, PD-L1, IL-10, were detected on non-switched memory B cells (p = 0.0049) and plasmablasts (p = 0.0020). The examination of blood samples obtained from patients with MASLD showed an increase of total B cells expressing IL-10 (p <0.0001) and IgM/IgD (p = 0.3361), CD19+CD20+CD5+CD1d+ Bregs (p = 0.6424) and CD19+CD20+CD27+ non-switched memory B cells (p = 0.0003). Conclusions Our results provide novel insights into the protumorigenic roles of several B cell subsets, the specific targeting of which could abrogate the progression of liver disease. Impact and implications Hepatocellular carcinoma (HCC) is the primary liver cancer with a constantly rising mortality rate. Metabolic dysfunction-associated steatotic liver disease (MASLD) is an emerging important cause of HCC. Current treatment options for HCC are limited and there is a high risk of recurrence. The study aims to identify new therapeutic strategies by exploring the immunological aspects of MASLD and HCC. Our findings extend the current knowledge on the role of B cells in the progression of MASLD and HCC. This study emphasizes the involvement of IgM+IgD+ regulatory B cells (Bregs) in malignant liver disease progression. These Bregs characterized by a high expression of PD-L1, IL-10, IgM, and IgD. Two other B cell subsets with immunosuppressive phenotype have been found in the study in murine liver disease - plasmablasts and non-switched memory B cells. Targeting these B cells could lead to more effective treatments of HCC.
Collapse
Affiliation(s)
- Nataliia Petriv
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
| | - Huizhen Suo
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
| | - Inga Hochnadel
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
| | - Kai Timrott
- Department of General-, Visceral and Transplantation Surgery, MHH, Hannover, Germany
| | - Nina Bondarenko
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
- Department of Pathological Anatomy, Forensic Medicine and Pathological Physiology, Dnipro State Medical University, Dnipro, Ukraine
- Institute of Pathology, MHH, Hannover, Germany
| | | | - Elena Reinhard
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Nils Jedicke
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
| | - Patrick Kaufhold
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
| | - Carlos Alberto Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | - Michael P. Manns
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
| | - Heike Bantel
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
| | - Tetyana Yevsa
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
| |
Collapse
|
14
|
Hoytema van Konijnenburg DP, Nigrovic PA, Zanoni I. Regional specialization within the mammalian respiratory immune system. Trends Immunol 2024; 45:871-891. [PMID: 39438172 PMCID: PMC11560516 DOI: 10.1016/j.it.2024.09.011] [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: 07/17/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024]
Abstract
The respiratory tract is exposed to infection from inhaled pathogens, including viruses, bacteria, and fungi. So far, a comprehensive assessment that integrates common and distinct aspects of the immune response along different areas of the respiratory tract has been lacking. Here, we discuss key recent findings regarding anatomical, functional, and microbial factors driving regional immune adaptation in the mammalian respiratory system, how they differ between mice and humans, and the similarities and differences with the gastrointestinal tract. We demonstrate that, under evolutionary pressure, mammals evolved spatially organized immune defenses that vary between the upper and lower respiratory tract. Overall, we propose that the functional specialization of the immune response along the respiratory tract has fundamental implications for the management of infectious or inflammatory diseases.
Collapse
Affiliation(s)
| | - Peter A Nigrovic
- Division of Immunology, Boston Children's Hospital, and Harvard Medical School, Boston, MA, USA; Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA
| | - Ivan Zanoni
- Division of Immunology, Boston Children's Hospital, and Harvard Medical School, Boston, MA, USA; Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
15
|
Boudigou M, Frutoso M, Hémon P, Le Dantec C, Chatzis L, Devauchelle V, Jamin C, Cornec D, Pers JO, Le Pottier L, Hillion S. Phenotypic, transcriptomic, and spatial characterization of CD45RB + naïve mature B cells: Implications in Sjögren's disease. Clin Immunol 2024; 268:110378. [PMID: 39393568 DOI: 10.1016/j.clim.2024.110378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/23/2024] [Accepted: 10/05/2024] [Indexed: 10/13/2024]
Abstract
The conventional classification of mature B cells overlooks the diversity within IgD+ CD27- naïve B cells. Here, to identify distinct mature naïve B cells, we categorized CD45RBMEM55- B cells (NA RB-) and CD45RBMEM55+ B cells (NA RB+) and explore their function and localization in circulation and tissues under physiological and pathological conditions. NA RB+ B cells, found in secondary lymphoid organs, differentiate into plasmablasts and secrete IgM. In Sjögren's disease, their numbers decrease, and they show over-activation and abnormal migration, suggesting an adaptive disease response. NA RB+ B cells also appear in inflamed salivary glands, indicating involvement in local immune responses. These findings highlight the distinct roles of NA RB+ B cells in health and Sjögren's disease.
Collapse
Affiliation(s)
| | | | | | | | - Loukas Chatzis
- UMR1227, LBAI, Univ Brest, Inserm, Brest, France; Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | | | | - Divi Cornec
- UMR1227, LBAI, Univ Brest, Inserm, and CHU Brest, Brest, France
| | | | | | - Sophie Hillion
- UMR1227, LBAI, Univ Brest, Inserm, and CHU Brest, Brest, France.
| |
Collapse
|
16
|
Kan C, Tan Z, Wang H, Wang W, Yang J, Zhang Y, Lu X, Cheng Q, Chai L, Peng C, Zhu J, Zhu C, Wang H, Zhan L, Lin K, Liu Y, Zhang L, Fan H, Zheng H. Spatiotemporal Analysis of Mesenchymal Stem Cells Fate Determination by Inflammatory Niche Following Soft Tissue Injury at a Single-Cell Level. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2310282. [PMID: 39308190 DOI: 10.1002/advs.202310282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 09/09/2024] [Indexed: 11/22/2024]
Abstract
Heterotopic ossification (HO), often arising in response to traumatic challenges, results from the aberrant osteochondral differentiation of mesenchymal stem cells (MSCs). Nevertheless, the impact of trauma-induced inflammatory exposure on MSC fate determination remains ambiguous. In this study, the cellular diversity within inflammatory lesions is elucidated, comprising MSCs and several innate and adaptive immune cells. It is observed that quiescent MSCs transition into cycling MSCs, subsequently giving rise to chondrogenic (cMSC) and/or osteogenic (oMSC) lineages within the inflammatory microenvironment following muscle or tendon injuries, as revealed through single-cell RNA sequencing (scRNA-seq), spatial transcriptome and lineage tracing analysis. Moreover, these investigations demonstrate that neutrophils and natural killer (NK) cells enhance transition of quiescent MSCs into cycling MSCs, which is also controlled by M1 macrophages, a subpopulation of macrophages can also stimulate cMSC and oMSC production from cycling MSCs. Additionally, M2 macrophages, CD4+ and CD8+ T lymphocytes are found to promote chondrogenesis. Further analysis demonstrates that immune cells promotes the activation of signaling transducers and activators of transcription (STAT) pathway and phosphoinositide 3 (PI3K)/protein kinase B (AKT) pathway in MSC proliferation and osteochondral progenitors' production, respectively. These findings highlight the dynamics of MSC fate within the inflammatory lesion and unveil the molecular landscape of osteoimmunological interactions, which holds promise for advancing HO treatment.
Collapse
Affiliation(s)
- Chen Kan
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Zhenya Tan
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Haitao Wang
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA
| | - Wei Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Jiazhao Yang
- Department of Orthopedics, The First Affiliated Hospital of USTC, Hefei, 230001, China
| | - Ya Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Xiaoling Lu
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Qirong Cheng
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Lanyi Chai
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Chao Peng
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Jicheng Zhu
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Chenghang Zhu
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Hailin Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Li Zhan
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Keqiong Lin
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Yakun Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Lingqiang Zhang
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China
| | - Haitao Fan
- Department of Orthopedics, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, China
| | - Hong Zheng
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| |
Collapse
|
17
|
Lam N, Lee Y, Farber DL. A guide to adaptive immune memory. Nat Rev Immunol 2024; 24:810-829. [PMID: 38831162 DOI: 10.1038/s41577-024-01040-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2024] [Indexed: 06/05/2024]
Abstract
Immune memory - comprising T cells, B cells and plasma cells and their secreted antibodies - is crucial for human survival. It enables the rapid and effective clearance of a pathogen after re-exposure, to minimize damage to the host. When antigen-experienced, memory T cells become activated, they proliferate and produce effector molecules at faster rates and in greater magnitudes than antigen-inexperienced, naive cells. Similarly, memory B cells become activated and differentiate into antibody-secreting cells more rapidly than naive B cells, and they undergo processes that increase their affinity for antigen. The ability of T cells and B cells to form memory cells after antigen exposure is the rationale behind vaccination. Understanding immune memory not only is crucial for the design of more-efficacious vaccines but also has important implications for immunotherapies in infectious disease and cancer. This 'guide to' article provides an overview of the current understanding of the phenotype, function, location, and pathways for the generation, maintenance and protective capacity of memory T cells and memory B cells.
Collapse
Affiliation(s)
- Nora Lam
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - YoonSeung Lee
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Donna L Farber
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Surgery, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
18
|
Kang S, Wu Q, Shen J, Wu C. CD27 is not an ideal marker for human memory B cells and can be modulated by IL-21 upon stimulated by Anti-CD40. Sci Rep 2024; 14:23742. [PMID: 39390111 PMCID: PMC11467254 DOI: 10.1038/s41598-024-75636-2] [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: 02/01/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024] Open
Abstract
B cells play a key role in humoral immune responses by producing antibodies. Although there are numerous research on memory B cells definition markers and cytokines on B cell development, different studies have yielded contradictory conclusions due to species studied, the different cells and stimulating agents used. In the current study, we conducted a detailed characterization of B cells in human CBMCs, PBMCs and tonsil, including expression of Igs, activation and memory markers. Furthermore, we found that considerable amounts of IgA and IgG were expressed by CD27- B cells. These "Atypical" memory B cells corresponded to approximately 50% of IgG+ and IgA+B cells in blood, this proportion even reached 90% in tonsil. In addition, we investigated the effect of IL-21 and TGF-β1 on the membrane-bound form and secreted form of Igs using PBMCs and purified blood B cells. There were actual differences between the effect of cytokines on Igs secretion and surface expression. Our study will be helpful to advance the knowledge and understanding of humoral memory.
Collapse
Affiliation(s)
- Shuangpeng Kang
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, The Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical University, Changsha, People's Republic of China.
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, The Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical University, 1501 Leifeng Road, 410219, Changsha, People's Republic of China.
| | - Qiongli Wu
- Shenzhen Experimental Education School, Shenzhen, People's Republic of China
| | - Juan Shen
- Kingmed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Changyou Wu
- Clinical Research Center of Clifford Hospital, Guangzhou, People's Republic of China.
- Institute of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, 510080, Guangzhou, People's Republic of China.
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, The Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical University, 1501 Leifeng Road, 410219, Changsha, People's Republic of China.
| |
Collapse
|
19
|
Liu JC, Zeng Q, Duan YG, Yeung WSB, Li RHW, Ng EHY, Cheung KW, Zhang Q, Chiu PCN. B cells: roles in physiology and pathology of pregnancy. Front Immunol 2024; 15:1456171. [PMID: 39434884 PMCID: PMC11491347 DOI: 10.3389/fimmu.2024.1456171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
B cells constitute a diverse and adaptable immune cell population with functions that can vary according to the environment and circumstances. The involvement of B cells in pregnancy, as well as the associated molecular pathways, has yet to be investigated. This review consolidates current knowledge on B cell activities and regulation during pregnancy, with a particular focus on the roles of various B cell subsets and the effects of B cell-derived factors on pregnancy outcomes. Moreover, the review examines the significance of B cell-associated autoantibodies, cytokines, and signaling pathways in relation to pregnancy complications such as pregnancy loss, preeclampsia, and preterm birth.
Collapse
Affiliation(s)
- Jin-Chuan Liu
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Qunxiong Zeng
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yong-Gang Duan
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - William S. B. Yeung
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Raymond H. W. Li
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Ernest H. Y. Ng
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Ka-Wang Cheung
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Qingqing Zhang
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Philip C. N. Chiu
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
20
|
Arlt E, Kindermann A, Fritsche AK, Navarrete Santos A, Kielstein H, Bazwinsky-Wutschke I. A Flow Cytometry-Based Examination of the Mouse White Blood Cell Differential in the Context of Age and Sex. Cells 2024; 13:1583. [PMID: 39329764 PMCID: PMC11430320 DOI: 10.3390/cells13181583] [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: 07/31/2024] [Revised: 09/10/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024] Open
Abstract
Analysis of the white blood cell differential as part of a flow cytometry-based approach is a common routine diagnostic tool used in clinics and research. For human blood, the methodological approach, suitable markers, and gating strategies are well-established. However, there is a lack of information regarding the mouse blood count. In this article, we deliver a fast and easy protocol for reprocessing mouse blood for the purpose of flow cytometric analysis, as well as suitable markers and gating strategies. We also present two possible applications: for the analysis of the whole blood count, with blood from a cardiac puncture, and for the analysis of a certain leukocyte subset at multiple time points in the framework of a mouse experiment, using blood from the facial vein. Additionally, we provide orientation values by applying the method to 3-month-old and 24-month-old male and female C57BL/6J mice. Our analyses demonstrate differences in the leukocyte fractions depending on age and sex. We discuss the influencing factors and limitations that can affect the results and that, therefore, need to be considered when applying this method. The present study fills the gap in the knowledge related to the rare information on flow cytometric analysis of mouse blood and, thus, lays the foundation for further investigations in this area.
Collapse
Affiliation(s)
- Elise Arlt
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin-Luther-University Halle-Wittenberg, 06108 Halle (Saale), Germany; (A.K.); (A.-K.F.); (H.K.); (I.B.-W.)
| | - Andrea Kindermann
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin-Luther-University Halle-Wittenberg, 06108 Halle (Saale), Germany; (A.K.); (A.-K.F.); (H.K.); (I.B.-W.)
| | - Anne-Kristin Fritsche
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin-Luther-University Halle-Wittenberg, 06108 Halle (Saale), Germany; (A.K.); (A.-K.F.); (H.K.); (I.B.-W.)
- Institute of Anatomy, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany
| | - Alexander Navarrete Santos
- Core Facility Flow Cytometry, Center for Basic Medical Research, Medical Faculty, Martin-Luther-University Halle-Wittenberg, 06108 Halle (Saale), Germany;
| | - Heike Kielstein
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin-Luther-University Halle-Wittenberg, 06108 Halle (Saale), Germany; (A.K.); (A.-K.F.); (H.K.); (I.B.-W.)
| | - Ivonne Bazwinsky-Wutschke
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin-Luther-University Halle-Wittenberg, 06108 Halle (Saale), Germany; (A.K.); (A.-K.F.); (H.K.); (I.B.-W.)
| |
Collapse
|
21
|
Spencer J, Dionisi C. Immature B cell homing shapes human lymphoid tissue structure and function. J Exp Med 2024; 221:e20240085. [PMID: 39093311 PMCID: PMC11296955 DOI: 10.1084/jem.20240085] [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: 04/11/2024] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024] Open
Abstract
Shortly after the emergence of newly formed human B cells from bone marrow as transitional cells, they diverge along two developmental pathways that can be distinguished by the level of IgM they express and migratory biases. Here, we propose that differential tissue homing of immature B cell subsets contributes to human lymphoid tissue structure and function.
Collapse
Affiliation(s)
- Jo Spencer
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Chiara Dionisi
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, UK
| |
Collapse
|
22
|
Peter B, Rebeaud J, Vigne S, Bressoud V, Phillips N, Ruiz F, Petrova TV, Bernier-Latmani J, Pot C. Perivascular B cells link intestinal angiogenesis to immunity and to the gut-brain axis during neuroinflammation. J Autoimmun 2024; 148:103292. [PMID: 39067313 DOI: 10.1016/j.jaut.2024.103292] [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: 02/06/2024] [Revised: 06/28/2024] [Accepted: 07/15/2024] [Indexed: 07/30/2024]
Abstract
Disruption of gut barrier function and intestinal immune cell homeostasis are increasingly considered critical players in pathogenesis of extra-intestinal inflammatory diseases, including multiple sclerosis (MS) and its prototypical animal model, the experimental autoimmune encephalomyelitis (EAE). Breakdown of epithelial barriers increases intestinal permeability and systemic dissemination of microbiota-derived molecules. However, whether the gut-vascular barrier (GVB) is altered during EAE has not been reported. Here, we demonstrate that endothelial cell proliferation and vessel permeability increase before EAE clinical onset, leading to vascular remodeling and expansion of intestinal villi capillary bed during disease symptomatic phase in an antigen-independent manner. Concomitant to onset of angiogenesis observed prior to neurological symptoms, we identify an increase of intestinal perivascular immune cells characterized by the surface marker lymphatic vessel endothelial hyaluronic acid receptor 1 (LYVE-1). LYVE-1+ is expressed more frequently on B cells that show high levels of CD73 and have proangiogenic properties. B cell depletion was sufficient to mitigate enteric blood endothelial cell proliferation following immunization for EAE. In conclusion, we propose that altered intestinal vasculature driven by a specialized LYVE-1+ B cell subset promotes angiogenesis and that loss of GVB function is implicated in EAE development and autoimmunity.
Collapse
Affiliation(s)
- Benjamin Peter
- Laboratories of Neuroimmunology, Service of Neurology and Neuroscience Research Center, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Epalinges, 1066, Switzerland
| | - Jessica Rebeaud
- Laboratories of Neuroimmunology, Service of Neurology and Neuroscience Research Center, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Epalinges, 1066, Switzerland
| | - Solenne Vigne
- Laboratories of Neuroimmunology, Service of Neurology and Neuroscience Research Center, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Epalinges, 1066, Switzerland
| | - Valentine Bressoud
- Laboratories of Neuroimmunology, Service of Neurology and Neuroscience Research Center, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Epalinges, 1066, Switzerland
| | - Nicholas Phillips
- Laboratories of Neuroimmunology, Service of Neurology and Neuroscience Research Center, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Epalinges, 1066, Switzerland
| | - Florian Ruiz
- Laboratories of Neuroimmunology, Service of Neurology and Neuroscience Research Center, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Epalinges, 1066, Switzerland
| | - Tatiana V Petrova
- Department of Oncology, University of Lausanne and Ludwig Institute for Cancer Research, Epalinges, 1066, Switzerland
| | - Jeremiah Bernier-Latmani
- Department of Oncology, University of Lausanne and Ludwig Institute for Cancer Research, Epalinges, 1066, Switzerland
| | - Caroline Pot
- Laboratories of Neuroimmunology, Service of Neurology and Neuroscience Research Center, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Epalinges, 1066, Switzerland.
| |
Collapse
|
23
|
Yuan Z, Shu L, Fu J, Yang P, Wang Y, Sun J, Zheng M, Liu Z, Yang J, Song J, Song S, Cai Z. Single-Cell RNA Sequencing Deconstructs the Distribution of Immune Cells Within Abdominal Aortic Aneurysms in Mice. Arterioscler Thromb Vasc Biol 2024; 44:1986-2003. [PMID: 39051127 DOI: 10.1161/atvbaha.124.321129] [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: 04/20/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Inflammation is a key component in the development of abdominal aortic aneurysm (AAA), yet insights into the roles of immune cells and their interactions in this process are limited. METHODS Using single-cell RNA transcriptomic analysis, we deconstructed the CD45+ cell population in elastase-induced murine AAA at the single-cell level. We isolated each group of immune cells from murine AAA tissue at different time points and divided them into several subtypes, listed the remarkable differentially expressed genes, explored the developmental trajectories of immune cells, and demonstrated the interactions among them. RESULTS Our findings reveal significant differences in several immune cell subsets, including macrophages, dendritic cells, and T cells, within the AAA microenvironment compared with the normal aorta. Especially, conventional dendritic cell type 1 exclusively existed in the AAA tissue rather than the normal aortas. Via CellChat analysis, we identified several intercellular communication pathways like visfatin, which targets monocyte differentiation and neutrophil extracellular trap-mediated interaction between neutrophils and dendritic cells, which might contribute to AAA development. Some of these pathways were validated in human AAA. CONCLUSIONS Despite the absence of external pathogenic stimuli, AAA tissues develop a complex inflammatory microenvironment involving numerous immune cells. In-depth studies of the inflammatory network shall provide new strategies for patients with AAA.
Collapse
MESH Headings
- Aortic Aneurysm, Abdominal/immunology
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/metabolism
- Animals
- Single-Cell Analysis
- Disease Models, Animal
- Mice, Inbred C57BL
- Aorta, Abdominal/pathology
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/immunology
- Mice
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Humans
- Macrophages/metabolism
- Macrophages/immunology
- Male
- Transcriptome
- RNA-Seq
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Gene Expression Profiling/methods
- Pancreatic Elastase
- Cell Communication
Collapse
Affiliation(s)
- Zhen Yuan
- Departments of Cardiology (Z.Y., L.S., Y.W., Z.C.), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, China (Z.Y., L.S., Y.W., Z.C.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Z.Y., L.S., Y.W., Z.C.)
| | - Li Shu
- Departments of Cardiology (Z.Y., L.S., Y.W., Z.C.), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, China (Z.Y., L.S., Y.W., Z.C.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Z.Y., L.S., Y.W., Z.C.)
| | - Jiantao Fu
- Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, China (J.F., P.Y., J.Y.)
| | - Peipei Yang
- Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, China (J.F., P.Y., J.Y.)
| | - Yidong Wang
- Departments of Cardiology (Z.Y., L.S., Y.W., Z.C.), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, China (Z.Y., L.S., Y.W., Z.C.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Z.Y., L.S., Y.W., Z.C.)
| | - Jie Sun
- Pathology (J. Sun, M.Z.), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mengsha Zheng
- Pathology (J. Sun, M.Z.), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhenjie Liu
- Vascular Surgery (Z.L.), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jin Yang
- Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, China (J.F., P.Y., J.Y.)
| | - Jiangping Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, China (J. Song, S.S.)
| | - Shen Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, China (J. Song, S.S.)
| | - Zhejun Cai
- Departments of Cardiology (Z.Y., L.S., Y.W., Z.C.), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, China (Z.Y., L.S., Y.W., Z.C.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Z.Y., L.S., Y.W., Z.C.)
| |
Collapse
|
24
|
Priest DG, Ebihara T, Tulyeu J, Søndergaard JN, Sakakibara S, Sugihara F, Nakao S, Togami Y, Yoshimura J, Ito H, Onishi S, Muratsu A, Mitsuyama Y, Ogura H, Oda J, Okusaki D, Matsumoto H, Wing JB. Atypical and non-classical CD45RB lo memory B cells are the majority of circulating SARS-CoV-2 specific B cells following mRNA vaccination or COVID-19. Nat Commun 2024; 15:6811. [PMID: 39122676 PMCID: PMC11315995 DOI: 10.1038/s41467-024-50997-4] [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: 11/27/2023] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Resting memory B cells can be divided into classical or atypical groups, but the heterogenous marker expression on activated memory B cells makes similar classification difficult. Here, by longitudinal analysis of mass cytometry and CITE-seq data from cohorts with COVID-19, bacterial sepsis, or BNT162b2 mRNA vaccine, we observe that resting B cell memory consist of classical CD45RB+ memory and CD45RBlo memory, of which the latter contains of two distinct groups of CD11c+ atypical and CD23+ non-classical memory cells. CD45RB levels remain stable in these cells after activation, thereby enabling the tracking of activated B cells and plasmablasts derived from either CD45RB+ or CD45RBlo memory B cells. Moreover, in both COVID-19 patients and mRNA vaccination, CD45RBlo B cells formed the majority of SARS-CoV2 specific memory B cells and correlated with serum antibodies, while CD45RB+ memory are activated by bacterial sepsis. Our results thus identify that stably expressed CD45RB levels can be exploited to trace resting memory B cells and their activated progeny, and suggest that atypical and non-classical CD45RBlo memory B cells contribute to SARS-CoV-2 infection and vaccination.
Collapse
Affiliation(s)
- David G Priest
- Laboratory of Human Single Cell Immunology, World Premier International Research Center Initiative Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Osaka, 563-0793, Japan
| | - Takeshi Ebihara
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Osaka, 565-0871, Japan
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Janyerkye Tulyeu
- Human Single Cell Immunology Team, Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Osaka, 565-0871, Japan
| | - Jonas N Søndergaard
- Human Single Cell Immunology Team, Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Osaka, 565-0871, Japan
| | - Shuhei Sakakibara
- Laboratory of Immune Regulation, IFReC, Osaka University, Suita, Osaka, 563-0793, Japan
- Graduate School of Medical Safety Management, Jikei University of Health Care Sciences, Osaka, 532-0003, Japan
| | - Fuminori Sugihara
- Core Instrumentation Facility, Immunology Frontier Research Center and Research Institute for Microbial Disease, Osaka University, Suita, Osaka, 563-0793, Japan
- Research Institute for Microbial Disease, Osaka University, Suita, Osaka, 563-0793, Japan
| | - Shunichiro Nakao
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Yuki Togami
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Jumpei Yoshimura
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Hiroshi Ito
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Shinya Onishi
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Arisa Muratsu
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Yumi Mitsuyama
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
- Division of Trauma and Surgical Critical Care, Osaka General Medical Center, Osaka, 558-8558, Japan
| | - Hiroshi Ogura
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Osaka, 565-0871, Japan
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Jun Oda
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Osaka, 565-0871, Japan
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Daisuke Okusaki
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Osaka, 565-0871, Japan
- Laboratory of Human Immunology (Single Cell Genomics), WPI-IFReC, Osaka University, Suita, 565-0871, Japan
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, 565-0871, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, 565-0871, Japan
| | - Hisatake Matsumoto
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Osaka, 565-0871, Japan.
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan.
| | - James B Wing
- Laboratory of Human Single Cell Immunology, World Premier International Research Center Initiative Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Osaka, 563-0793, Japan.
- Human Single Cell Immunology Team, Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Osaka, 565-0871, Japan.
- Center for Advanced Modalities and DDS (CAMaD), Osaka University, Osaka, Japan.
| |
Collapse
|
25
|
Tsokos GC. The immunology of systemic lupus erythematosus. Nat Immunol 2024; 25:1332-1343. [PMID: 39009839 DOI: 10.1038/s41590-024-01898-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/17/2024] [Indexed: 07/17/2024]
Abstract
Understanding the pathogenesis and clinical manifestations of systemic lupus erythematosus (SLE) has been a great challenge. Reductionist approaches to understand the nature of the disease have identified many pathogenetic contributors that parallel clinical heterogeneity. This Review outlines the immunological control of SLE and looks to experimental tools and approaches that are improving our understanding of the complex contribution of interacting genetics, environment, sex and immunoregulatory factors and their interface with processes inherent to tissue parenchymal cells. Efforts to advance precision medicine in the care of patients with SLE along with treatment strategies to correct the immune system hold hope and are also examined.
Collapse
Affiliation(s)
- George C Tsokos
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
26
|
Rosen Y, Brbić M, Roohani Y, Swanson K, Li Z, Leskovec J. Toward universal cell embeddings: integrating single-cell RNA-seq datasets across species with SATURN. Nat Methods 2024; 21:1492-1500. [PMID: 38366243 PMCID: PMC11310084 DOI: 10.1038/s41592-024-02191-z] [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: 02/03/2023] [Accepted: 01/22/2024] [Indexed: 02/18/2024]
Abstract
Analysis of single-cell datasets generated from diverse organisms offers unprecedented opportunities to unravel fundamental evolutionary processes of conservation and diversification of cell types. However, interspecies genomic differences limit the joint analysis of cross-species datasets to homologous genes. Here we present SATURN, a deep learning method for learning universal cell embeddings that encodes genes' biological properties using protein language models. By coupling protein embeddings from language models with RNA expression, SATURN integrates datasets profiled from different species regardless of their genomic similarity. SATURN can detect functionally related genes coexpressed across species, redefining differential expression for cross-species analysis. Applying SATURN to three species whole-organism atlases and frog and zebrafish embryogenesis datasets, we show that SATURN can effectively transfer annotations across species, even when they are evolutionarily remote. We also demonstrate that SATURN can be used to find potentially divergent gene functions between glaucoma-associated genes in humans and four other species.
Collapse
Affiliation(s)
- Yanay Rosen
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Maria Brbić
- School of Computer and Communication Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Yusuf Roohani
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Kyle Swanson
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Ziang Li
- Department of Computer Science and Technology, Tsinghua University, Beijing, China
| | - Jure Leskovec
- Department of Computer Science, Stanford University, Stanford, CA, USA.
| |
Collapse
|
27
|
Tsai CY, Oo M, Peh JH, Yeo BCM, Aptekmann A, Lee B, Liu JJJ, Tsao WS, Dick T, Fink K, Gengenbacher M. Splenic marginal zone B cells restrict Mycobacterium tuberculosis infection by shaping the cytokine pattern and cell-mediated immunity. Cell Rep 2024; 43:114426. [PMID: 38959109 PMCID: PMC11307145 DOI: 10.1016/j.celrep.2024.114426] [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: 12/14/2023] [Revised: 04/29/2024] [Accepted: 06/17/2024] [Indexed: 07/05/2024] Open
Abstract
Understanding the role of B cells in tuberculosis (TB) is crucial for developing new TB vaccines. However, the changes in B cell immune landscapes during TB and their functional implications remain incompletely explored. Using high-dimensional flow cytometry to map the immune landscape in response to Mycobacterium tuberculosis (Mtb) infection, our results show an accumulation of marginal zone B (MZB) cells and other unconventional B cell subsets in the lungs and spleen, shaping an unconventional B cell landscape. These MZB cells exhibit activated and memory-like phenotypes, distinguishing their functional profiles from those of conventional B cells. Notably, functional studies show that MZB cells produce multiple cytokines and contribute to systemic protection against TB by shaping cytokine patterns and cell-mediated immunity. These changes in the immune landscape are reversible upon successful TB chemotherapy. Our study suggests that, beyond antibody production, targeting the regulatory function of B cells may be a valuable strategy for TB vaccine development.
Collapse
Affiliation(s)
- Chen-Yu Tsai
- Center for Discovery and Innovation (CDI), Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ 07110, USA
| | - Myo Oo
- Center for Discovery and Innovation (CDI), Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ 07110, USA
| | - Jih Hou Peh
- Biosafety Level 3 Core, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Level 15, Centre for Translational Medicine (MD6), NUS, 14 Medical Drive, Singapore 117599, Singapore
| | - Benjamin C M Yeo
- Infectious Diseases Translational Research Programme and Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Level 2, Blk MD4, 5 Science Drive 2, Singapore 117545, Singapore
| | - Ariel Aptekmann
- Center for Discovery and Innovation (CDI), Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ 07110, USA
| | - Bernett Lee
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research, Biopolis, 8A Biomedical Grove, Level 3 & 4, Immunos Building, Singapore 138648, Singapore; Centre for Biomedical Informatics, Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore; A(∗)STAR Infectious Diseases Labs, Agency for Science, Technology and Research, 8A Biomedical Grove #05-13, Immunos, Singapore 138648, Singapore
| | - Joe J J Liu
- Biosafety Level 3 Core, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Level 15, Centre for Translational Medicine (MD6), NUS, 14 Medical Drive, Singapore 117599, Singapore
| | - Wen-Shan Tsao
- Center for Discovery and Innovation (CDI), Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ 07110, USA
| | - Thomas Dick
- Center for Discovery and Innovation (CDI), Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ 07110, USA; Hackensack Meridian School of Medicine, Nutley, NJ 07110, USA
| | - Katja Fink
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research, Biopolis, 8A Biomedical Grove, Level 3 & 4, Immunos Building, Singapore 138648, Singapore
| | - Martin Gengenbacher
- Center for Discovery and Innovation (CDI), Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ 07110, USA; Hackensack Meridian School of Medicine, Nutley, NJ 07110, USA.
| |
Collapse
|
28
|
Humphries J, Fletcher NL, Sonderegger SE, Bell CA, Kempe K, Thurecht KJ. Mitigating the Effects of Persistent Antipolymer Immune Reactions in Nanomedicine: Evaluating Materials-Based Approaches Using Molecular Imaging. ACS NANO 2024. [PMID: 39037055 DOI: 10.1021/acsnano.4c07317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Poly(ethylene glycol) (PEG) is a hydrophilic polymer ubiquitously used in both medical and nonmedical goods. Recent debate surrounding the observed stimulation of immune responses against PEG has spurred the development of materials that may be suitable replacements for this common polymeric component. The underlying view is that these alternative materials with comparable physicochemical properties can overcome the unfavorable and unpredictable effects of antibody-mediated clearance by being chemically, and therefore antigenically, distinct from PEG. However, this hypothesis has not been thoroughly tested in any defined manner, and the immune response observed against PEG has not been rigorously investigated within the context of these emerging materials. Consequently, it remains unclear whether immunity-mediated discrimination between polymeric entities even occurs in vivo and, if this is the case, how it may be exploited. In this study, we utilize positron emission tomography-computed tomography molecular imaging in mice immunized to develop specific antibody responses to PEG and an alternative polymer in order to visualize and quantify the influence of antipolymer antibodies on the biodistribution of synthetic polymers in vivo as a function of immunization status. Under the conditions of this experiment, mice could be primed to exhibit both innate and adaptive immunity to all of the polymer systems to which they were exposed. We demonstrate that alternating between chemically disparate polymers is a viable approach to extend their efficacy when antipolymer humoral immune responses arise.
Collapse
Affiliation(s)
- James Humphries
- Centre for Advanced Imaging (CAI) and Australian Institute for Bioengineering and Nanotechnology, ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Nicholas L Fletcher
- Centre for Advanced Imaging (CAI) and Australian Institute for Bioengineering and Nanotechnology, ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Stefan E Sonderegger
- Centre for Advanced Imaging (CAI) and Australian Institute for Bioengineering and Nanotechnology, ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Craig A Bell
- Centre for Advanced Imaging (CAI) and Australian Institute for Bioengineering and Nanotechnology, ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Kristian Kempe
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Materials Science and Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Kristofer J Thurecht
- Centre for Advanced Imaging (CAI) and Australian Institute for Bioengineering and Nanotechnology, ARC Training Centre for Innovation in Biomedical Imaging Technology, The University of Queensland, St. Lucia, Queensland 4072, Australia
| |
Collapse
|
29
|
Li M, Yao Z, Wang H, Ma Y, Yang W, Guo Y, Yu G, Shi W, Zhang N, Xu M, Li X, Zhao J, Zhang Y, Xue C, Sun B. Silicon or Calcium Doping Coordinates the Immunostimulatory Effects of Aluminum Oxyhydroxide Nanoadjuvants in Prophylactic Vaccines. ACS NANO 2024; 18:16878-16894. [PMID: 38899978 DOI: 10.1021/acsnano.4c02685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Aluminum salts still remain as the most popular adjuvants in marketed human prophylactic vaccines due to their capability to trigger humoral immune responses with a good safety record. However, insufficient induction of cellular immune responses limits their further applications. In this study, we prepare a library of silicon (Si)- or calcium (Ca)-doped aluminum oxyhydroxide (AlOOH) nanoadjuvants. They exhibit well-controlled physicochemical properties, and the dopants are homogeneously distributed in nanoadjuvants. By using Hepatitis B surface antigen (HBsAg) as the model antigen, doped AlOOH nanoadjuvants mediate higher antigen uptake and promote lysosome escape of HBsAg through lysosomal rupture induced by the dissolution of the dopant in the lysosomes in bone marrow-derived dendritic cells (BMDCs). Additionally, doped nanoadjuvants trigger higher antigen accumulation and immune cell activation in draining lymph nodes. In HBsAg and varicella-zoster virus glycoprotein E (gE) vaccination models, doped nanoadjuvants induce high IgG titer, activations of CD4+ and CD8+ T cells, cytotoxic T lymphocytes, and generations of effector memory T cells. Doping of aluminum salt-based adjuvants with biological safety profiles and immunostimulating capability is a potential strategy to mediate robust humoral and cellular immunity. It potentiates the applications of engineered adjuvants in the development of vaccines with coordinated immune responses.
Collapse
Affiliation(s)
- Min Li
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Zhiying Yao
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Huiyang Wang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Yubin Ma
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Wenqi Yang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Yiyang Guo
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Ge Yu
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Wendi Shi
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Ning Zhang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Muzhe Xu
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Xin Li
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Jiashu Zhao
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Yue Zhang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Changying Xue
- School of Bioengineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| | - Bingbing Sun
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
- Frontiers Science Center for Smart Materials Oriented Chemical Engineering, School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, Dalian 116024, China
| |
Collapse
|
30
|
Huang YH, Yoon CH, Gandhi A, Hanley T, Castrillon C, Kondo Y, Lin X, Kim W, Yang C, Driouchi A, Carroll M, Gray-Owen SD, Wesemann DR, Drake CG, Bertagnolli MM, Beauchemin N, Blumberg RS. High-dimensional mapping of human CEACAM1 expression on immune cells and association with melanoma drug resistance. COMMUNICATIONS MEDICINE 2024; 4:128. [PMID: 38956268 PMCID: PMC11219841 DOI: 10.1038/s43856-024-00525-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 05/08/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Human carcinoembryonic antigen cell adhesion molecule 1 (CEACAM1) is an inhibitory cell surface protein that functions through homophilic and heterophilic ligand binding. Its expression on immune cells in human tumors is poorly understood. METHODS An antibody that distinguishes human CEACAM1 from other highly related CEACAM family members was labeled with 159Tb and inserted into a panel of antibodies that included specificity for programmed cell death protein 1 (PD1) and PD-L1, which are targets of immunotherapy, to gain a data-driven immune cell atlas using cytometry by time-of-flight (CyTOF). A detailed inventory of CEACAM1, PD1, and PD-L1 expression on immune cells in metastatic lesions to lymph node or soft tissues and peripheral blood samples from patients with treatment-naive and -resistant melanoma as well as peripheral blood samples from healthy controls was performed. RESULTS CEACAM1 is absent or at low levels on healthy circulating immune cells but is increased on immune cells in peripheral blood and tumors of melanoma patients. The majority of circulating PD1-positive NK cells, innate T cells, B cells, monocytic cells, dendritic cells, and CD4+ T cells in the peripheral circulation of treatment-resistant disease co-express CEACAM1 and are demonstrable as discrete populations. CEACAM1 is present on distinct types of cells that are unique to the tumor microenvironment and exhibit expression levels that are highest in treatment resistance; this includes tumor-infiltrating CD8+ T cells. CONCLUSIONS To the best of our knowledge, this work represents the first comprehensive atlas of CEACAM1 expression on immune cells in a human tumor and reveals an important correlation with treatment-resistant disease. These studies suggest that agents targeting CEACAM1 may represent appropriate partners for PD1-related pathway therapies.
Collapse
Affiliation(s)
- Yu-Hwa Huang
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Charles H Yoon
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Amit Gandhi
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Thomas Hanley
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Carlos Castrillon
- Program in Cellular and Molecular Medicine, Children's Hospital Medical Center, Harvard Medical School, Boston, MA, USA
| | - Yasuyuki Kondo
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Internal Medicine, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Xi Lin
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Walter Kim
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Chao Yang
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Amine Driouchi
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Michael Carroll
- Program in Cellular and Molecular Medicine, Children's Hospital Medical Center, Harvard Medical School, Boston, MA, USA
| | - Scott D Gray-Owen
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Duane R Wesemann
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Allergy and Immunology, Division of Genetics, Brigham and Women's Hospital and Ragon Institute of MGH, MIT and Harvard, Boston, MA, USA
| | - Charles G Drake
- Herbert Irving Comprehensive Cancer Center, Columbia University School of Medicine, New York, NY, USA
- Janssen R&D, Springhouse, PA, USA
| | - Monica M Bertagnolli
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- National Institutes of Health, Bethesda, MD, USA
| | - Nicole Beauchemin
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Richard S Blumberg
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
31
|
Fooksman DR, Jing Z, Park R. New insights into the ontogeny, diversity, maturation and survival of long-lived plasma cells. Nat Rev Immunol 2024; 24:461-470. [PMID: 38332373 DOI: 10.1038/s41577-024-00991-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2024] [Indexed: 02/10/2024]
Abstract
Plasma cells are unique immune effectors, capable of producing large amounts of high-affinity antibodies that protect against pathogenic infections. Although most plasma cells have short lifespans, certain conditions or vaccinations can give rise to long-lived plasma cells (LLPCs) that provide individuals with lifelong protection against pathogen exposure. The nature of these LLPCs is poorly understood; however, recent studies have shed new light on the ontogeny, diversity, maturation and survival of these unique cells. Whereas LLPCs had been thought to arise preferentially from germinal centres, novel genetic tools have revealed that they can originate from various stages throughout the humoral response. Furthermore, new single-cell analyses have shown that mouse and human plasma cells are heterogeneous and may undergo further maturation in situ in the bone marrow niche. Finally, plasma cells were previously considered to be sessile cells maintained in fixed survival niches, but new data show that plasma cell subsets can differentially migrate and organize into clusters that may be associated with survival niches. These descriptive findings provide new insights into how cell-intrinsic programmes and extrinsic factors may regulate the longevity of plasma cells in various contexts, which suggest new research avenues for their functional validation.
Collapse
Affiliation(s)
- David R Fooksman
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Zhixin Jing
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Rosa Park
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
32
|
Elsner RA, Smita S, Shlomchik MJ. IL-12 induces a B cell-intrinsic IL-12/IFNγ feed-forward loop promoting extrafollicular B cell responses. Nat Immunol 2024; 25:1283-1295. [PMID: 38862796 PMCID: PMC11992614 DOI: 10.1038/s41590-024-01858-1] [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: 05/12/2023] [Accepted: 04/26/2024] [Indexed: 06/13/2024]
Abstract
While some infections elicit germinal centers, others produce only extrafollicular responses. The mechanisms controlling these dichotomous fates are poorly understood. We identify IL-12 as a cytokine switch, acting directly on B cells to promote extrafollicular and suppress germinal center responses. IL-12 initiates a B cell-intrinsic feed-forward loop between IL-12 and IFNγ, amplifying IFNγ production, which promotes proliferation and plasmablast differentiation from mouse and human B cells, in synergy with IL-12. IL-12 sustains the expression of a portion of IFNγ-inducible genes. Together, they also induce unique gene changes, reflecting both IFNγ amplification and cooperative effects between both cytokines. In vivo, cells lacking both IL-12 and IFNγ receptors are more impaired in plasmablast production than those lacking either receptor alone. Further, B cell-derived IL-12 enhances both plasmablast responses and T helper 1 cell commitment. Thus, B cell-derived IL-12, acting on T and B cells, determines the immune response mode, with implications for vaccines, pathogen protection and autoimmunity.
Collapse
Affiliation(s)
- Rebecca A Elsner
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Shuchi Smita
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mark J Shlomchik
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
33
|
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
|
34
|
Xu Z, Li R, Wang L, Wu Y, Tian Y, Su Y, Ma Y, Li R, Wei Y, Zhang C, Han S, Duan S, Peng H, Xue J. Pathogenic role of different phenotypes of immune cells in airway allergic diseases: a study based on Mendelian randomization. Front Immunol 2024; 15:1349470. [PMID: 38812518 PMCID: PMC11133742 DOI: 10.3389/fimmu.2024.1349470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 04/25/2024] [Indexed: 05/31/2024] Open
Abstract
Background Airway allergic disease (AAD) is a class of autoimmune diseases with predominantly Th2-type inflammation, mainly including allergic rhinitis (AR), allergic asthma (AS), and chronic sinusitis (CRS). There are very complex regulatory mechanisms between immune cells and AAD; however, previous reports found that the functions of the same immune cells in AAD are not identical. Objective The aim of this study was to explore the causal relationship between different phenotypic immune cells and their association with AAD. Method Utilizing the publicly available Genome-Wide Association Studies (GWAS) database, this study conducted a bidirectional Mendelian randomization (MR) to assess the causal relationship between immune cells of 731 different immunophenotypes and AAD. The primary assessment methods included inverse variance weighting, weighted median, and MR Egger. Additionally, sensitivity analyses such as MR-PRESSO, leave-one-out, and scatter plots were employed to eliminate the interference of heterogeneity and pleiotropy, ensuring the stability of the causal inference. Result A total of 38 immune cells with different immunophenotypes were found to be positively and causally associated with AR, of which 26 were protective factors and 12 were risk factors. Positive associations were found between 33 immune cells and AS, of which 14 were protective factors and 19 were risk factors, as well as between 39 immune cells and CRS, of which 22 were protective factors and 17 were risk factors. Finally, the results of all relevant immune cells for the three diseases were taken and intersected, and it was found that CD3 on CD39+-activated Treg (IVWAR = 0.001, IVWCRS = 0.043, IVWAS = 0.027) may be the key immune cell that inhibits the development of AAD (ORAR = 0.940, ORAS = 0.967, ORCRS = 0.976). Conclusion This study reveals that different immune phenotypes of immune cells are closely related to AAD at the genetic level, which provides a theoretical basis for future clinical studies.
Collapse
Affiliation(s)
- Zhihan Xu
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Airway Inflammatory Diseases Neuroimmunity Laboratory, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ren Li
- Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Leigang Wang
- Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yisha Wu
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Airway Inflammatory Diseases Neuroimmunity Laboratory, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yuhe Tian
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Airway Inflammatory Diseases Neuroimmunity Laboratory, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yilin Su
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Airway Inflammatory Diseases Neuroimmunity Laboratory, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yuqiang Ma
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Airway Inflammatory Diseases Neuroimmunity Laboratory, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ruiying Li
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Airway Inflammatory Diseases Neuroimmunity Laboratory, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yao Wei
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Airway Inflammatory Diseases Neuroimmunity Laboratory, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Medical University, Taiyuan, Shanxi, China
| | - Chen Zhang
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Airway Inflammatory Diseases Neuroimmunity Laboratory, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Medical University, Taiyuan, Shanxi, China
| | - Shikai Han
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Airway Inflammatory Diseases Neuroimmunity Laboratory, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Medical University, Taiyuan, Shanxi, China
| | - Siyu Duan
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Airway Inflammatory Diseases Neuroimmunity Laboratory, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Medical University, Taiyuan, Shanxi, China
| | - Haiyi Peng
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Airway Inflammatory Diseases Neuroimmunity Laboratory, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jinmei Xue
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Airway Inflammatory Diseases Neuroimmunity Laboratory, Head & Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
35
|
Chi H, Pepper M, Thomas PG. Principles and therapeutic applications of adaptive immunity. Cell 2024; 187:2052-2078. [PMID: 38670065 PMCID: PMC11177542 DOI: 10.1016/j.cell.2024.03.037] [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: 01/02/2024] [Revised: 03/01/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024]
Abstract
Adaptive immunity provides protection against infectious and malignant diseases. These effects are mediated by lymphocytes that sense and respond with targeted precision to perturbations induced by pathogens and tissue damage. Here, we review key principles underlying adaptive immunity orchestrated by distinct T cell and B cell populations and their extensions to disease therapies. We discuss the intracellular and intercellular processes shaping antigen specificity and recognition in immune activation and lymphocyte functions in mediating effector and memory responses. We also describe how lymphocytes balance protective immunity against autoimmunity and immunopathology, including during immune tolerance, response to chronic antigen stimulation, and adaptation to non-lymphoid tissues in coordinating tissue immunity and homeostasis. Finally, we discuss extracellular signals and cell-intrinsic programs underpinning adaptive immunity and conclude by summarizing key advances in vaccination and engineering adaptive immune responses for therapeutic interventions. A deeper understanding of these principles holds promise for uncovering new means to improve human health.
Collapse
Affiliation(s)
- Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Marion Pepper
- Department of Immunology, University of Washington, Seattle, WA, USA.
| | - Paul G Thomas
- Department of Host-Microbe Interactions and Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
36
|
Etesami NS, Barker KA, Shenoy AT, De Ana CL, Arafa EI, Grifno GN, Matschulat AM, Vannini ME, Pihl RMF, Breen MP, Soucy AM, Goltry WN, Ha CT, Betsuyaku H, Browning JL, Varelas X, Traber KE, Jones MR, Quinton LJ, Maglione PJ, Nia HT, Belkina AC, Mizgerd JP. B cells in the pneumococcus-infected lung are heterogeneous and require CD4 + T cell help including CD40L to become resident memory B cells. Front Immunol 2024; 15:1382638. [PMID: 38715601 PMCID: PMC11074383 DOI: 10.3389/fimmu.2024.1382638] [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: 02/06/2024] [Accepted: 04/01/2024] [Indexed: 05/12/2024] Open
Abstract
Recovery from respiratory pneumococcal infections generates lung-localized protection against heterotypic bacteria, mediated by resident memory lymphocytes. Optimal protection in mice requires re-exposure to pneumococcus within days of initial infection. Serial surface marker phenotyping of B cell populations in a model of pneumococcal heterotypic immunity revealed that bacterial re-exposure stimulates the immediate accumulation of dynamic and heterogeneous populations of B cells in the lung, and is essential for the establishment of lung resident memory B (BRM) cells. The B cells in the early wave were activated, proliferating locally, and associated with both CD4+ T cells and CXCL13. Antagonist- and antibody-mediated interventions were implemented during this early timeframe to demonstrate that lymphocyte recirculation, CD4+ cells, and CD40 ligand (CD40L) signaling were all needed for lung BRM cell establishment, whereas CXCL13 signaling was not. While most prominent as aggregates in the loose connective tissue of bronchovascular bundles, morphometry and live lung imaging analyses showed that lung BRM cells were equally numerous as single cells dispersed throughout the alveolar septae. We propose that CD40L signaling from antigen-stimulated CD4+ T cells in the infected lung is critical to establishment of local BRM cells, which subsequently protect the airways and parenchyma against future potential infections.
Collapse
Affiliation(s)
- Neelou S. Etesami
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Kimberly A. Barker
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Anukul T. Shenoy
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Carolina Lyon De Ana
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Emad I. Arafa
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Gabrielle N. Grifno
- Department of Biomedical Engineering, Boston University College of Engineering, Boston, MA, United States
| | - Adeline M. Matschulat
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Michael E. Vannini
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Riley M. F. Pihl
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Michael P. Breen
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Alicia M. Soucy
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Wesley N. Goltry
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Catherine T. Ha
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Hanae Betsuyaku
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Jeffrey L. Browning
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Xaralabos Varelas
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Katrina E. Traber
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Matthew R. Jones
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Lee J. Quinton
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Division of Infectious Diseases and Immunology, University of Massachusetts Chan Medical School, Worcester, MA, United States
- Department of Pathology and Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Paul J. Maglione
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Hadi T. Nia
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Biomedical Engineering, Boston University College of Engineering, Boston, MA, United States
| | - Anna C. Belkina
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Pathology and Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Flow Cytometry Core Facility, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Joseph P. Mizgerd
- Pulmonary Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Virology, Immunology, and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| |
Collapse
|
37
|
Zhu X, Hong S, Bu J, Liu Y, Liu C, Li R, Zhang T, Zhang Z, Li L, Zhou X, Hua Z, Zhu B, Hou B. Antiviral memory B cells exhibit enhanced innate immune response facilitated by epigenetic memory. SCIENCE ADVANCES 2024; 10:eadk0858. [PMID: 38552009 PMCID: PMC10980274 DOI: 10.1126/sciadv.adk0858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 02/26/2024] [Indexed: 04/01/2024]
Abstract
The long-lasting humoral immunity induced by viral infections or vaccinations depends on memory B cells with greatly increased affinity to viral antigens, which are evolved from germinal center (GC) responses. However, it is unclear whether antiviral memory B cells represent a distinct subset among the highly heterogeneous memory B cell population. Here, we examined memory B cells induced by a virus-mimicking antigen at both transcriptome and epigenetic levels and found unexpectedly that antiviral memory B cells exhibit an enhanced innate immune response, which appeared to be facilitated by the epigenetic memory that is established through the memory B cell development. In addition, T-bet is associated with the altered chromatin architecture and is required for the formation of the antiviral memory B cells. Thus, antiviral memory B cells are distinct from other GC-derived memory B cells in both physiological functions and epigenetic landmarks.
Collapse
Affiliation(s)
- Xiping Zhu
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Sheng Hong
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jiachen Bu
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yingping Liu
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Can Liu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Runhan Li
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tiantian Zhang
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhuqiang Zhang
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Liping Li
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xuyu Zhou
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhaolin Hua
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bing Zhu
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- New Cornerstone Science Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Baidong Hou
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
38
|
Wang YX, Zhou CP, Wang DT, Ma J, Sun XH, Wang Y, Zhang YM. Unraveling the causal role of immune cells in gastrointestinal tract cancers: insights from a Mendelian randomization study. Front Immunol 2024; 15:1343512. [PMID: 38533503 PMCID: PMC10963466 DOI: 10.3389/fimmu.2024.1343512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 02/21/2024] [Indexed: 03/28/2024] Open
Abstract
Background Despite early attempts, the relationship between immune characteristics and gastrointestinal tract cancers remains incompletely elucidated. Hence, rigorous and further investigations in this domain hold significant clinical relevance for the development of novel potential immunotherapeutic targets. Methods We conducted a two-sample Mendelian randomization (MR) analysis using the tools available in the "TwoSampleMR" R package. The GWAS data for these 731 immune traits were sourced from the GWAS Catalog database. Concurrently, data on gastrointestinal tract cancers, encompassing malignant tumors in the esophagus, stomach, small intestine, colon, and rectum, were extracted from the FinnGen database. The immune traits subjected to MR analysis predominantly fall into four categories: median fluorescence intensities (MFI), relative cell (RC), absolute cell (AC), and morphological parameters (MP). To ensure the reliability of our findings, sensitivity analyses were implemented to address robustness, account for heterogeneity, and alleviate the impact of horizontal pleiotropy. Results A total of 78 immune traits causally linked to gastrointestinal tract cancers were identified, encompassing esophageal cancer (12 traits), gastric cancer (13 traits), small intestine cancer (22 traits), colon cancer (12 traits), and rectal cancer (19 traits). Additionally, 60 immune traits were recognized as protective factors associated with gastrointestinal tract cancers, distributed across esophageal cancer (14 traits), gastric cancer (16 traits), small intestine cancer (7 traits), colon cancer (14 traits), and rectal cancer (9 traits). Furthermore, it was observed that seven immune traits are causally related to gastrointestinal tract cancers in at least two locations. These traits include "CCR2 on CD14- CD16+ monocyte," "CD19 on IgD+ CD38-," "CD19 on IgD+ CD38- naive," "CD25hi CD45RA+ CD4 not Treg AC," "CD27 on unsw mem," "CD28 on CD39+ activated Treg," and "CD45 on CD4+." Conclusion This study elucidates a causal link between immune cells and gastrointestinal tract cancers at various sites through genetic investigation. The findings of this research open up new perspectives and resources for exploring tumor prevention strategies and immunotherapeutic targets.
Collapse
Affiliation(s)
- Yu-xiang Wang
- Department of General Surgery, Anqing Municipal Hospital, Anqing, Anhui, China
| | - Chao-ping Zhou
- Department of General Surgery, Anqing Municipal Hospital, Anqing, Anhui, China
| | - Da-tian Wang
- Department of General Surgery, Anqing Municipal Hospital, Anqing, Anhui, China
| | - Jun Ma
- Department of General Surgery, Anqing Municipal Hospital, Anqing, Anhui, China
| | - Xue-hu Sun
- Department of Emergency Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yao Wang
- Department of Digestive Endoscopy, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ya-ming Zhang
- Department of General Surgery, Anqing Municipal Hospital, Anqing, Anhui, China
| |
Collapse
|
39
|
Syeda MZ, Hong T, Huang C, Huang W, Mu Q. B cell memory: from generation to reactivation: a multipronged defense wall against pathogens. Cell Death Discov 2024; 10:117. [PMID: 38453885 PMCID: PMC10920759 DOI: 10.1038/s41420-024-01889-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 03/09/2024] Open
Abstract
Development of B cell memory is a conundrum that scientists are still exploring. Studies have been conducted in vitro and using advanced animal models to elucidate the mechanism underlying the generation of memory B cells (MBCs), the precise roles of MBCs against pathogens, and their protective functions against repeated infections throughout life. Lifelong immunity against invading diseases is mainly the result of overcoming a single infection. This protection is largely mediated by the two main components of B cell memory-MBCs and long-lived plasma cells (PCs). The chemical and cellular mechanisms that encourage fat selection for MBCs or long-lived PCs are an area of active research. Despite the fact that nearly all available vaccinations rely on the capacity to elicit B-cell memory, we have yet to develop successful vaccines that can induce broad-scale protective MBCs against some of the deadliest diseases, including malaria and AIDS. A deeper understanding of the specific cellular and molecular pathways that govern the generation, function, and reactivation of MBCs is critical for overcoming the challenges associated with vaccine development. Here, we reviewed literature on the development of MBCs and their reactivation, interaction with other cell types, strategies against invading pathogens, and function throughout life and discussed the recent advances regarding the key signals and transcription factors which regulate B cell memory and their relevance to the quest for vaccine development.
Collapse
Affiliation(s)
- Madiha Zahra Syeda
- The People's Hospital of Gaozhou, Guangdong Medical University, Maoming, 525200, China
- School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Tu Hong
- The First Affiliated Hospital, Zhejiang University, School of Medicine, 310058, Hangzhou, China
| | - Chunming Huang
- The People's Hospital of Gaozhou, Guangdong Medical University, Maoming, 525200, China.
| | - Wenhua Huang
- School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Qingchun Mu
- The People's Hospital of Gaozhou, Guangdong Medical University, Maoming, 525200, China.
| |
Collapse
|
40
|
Woodruff MC, Faliti CE, Sanz I. Systems biology of B cells in COVID-19. Semin Immunol 2024; 72:101875. [PMID: 38489999 PMCID: PMC11988200 DOI: 10.1016/j.smim.2024.101875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 03/17/2024]
Abstract
The integration of multi-'omic datasets into complex systems-wide assessments has become a mainstay in immunologic investigation. This focus on high-dimensional data collection and analysis was on full display in the investigation of COVID-19, the respiratory illness resulting from infection by the novel coronavirus SARS-CoV-2. Particularly in the area of B cell biology, tremendous efforts in both cellular and serologic investigation have resulted in an increasingly detailed mapping of the coordinated effector, memory, and antibody secreting cell responses that underpin the development of humoral immunity in response to primary viral infection. Further, the rapid development and deployment of effective vaccines has allowed for the assessment of developing memory responses across a wide variety of immune contexts, including in patients with compromised immune function. The result has been a period of rapid gains in the understanding of B cell biology unrestricted to the study of COVID-19. Here, we outline the systems-level technologies that have been routinely implemented in these investigations throughout the pandemic, and discuss how their use has led to clear and applicable gains in pursuance of the amelioration of human infectious disease and beyond.
Collapse
Affiliation(s)
- Matthew C Woodruff
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA; Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA.
| | - Caterina E Faliti
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA; Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA.
| | - Ignacio Sanz
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA; Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| |
Collapse
|
41
|
Callahan D, Smita S, Joachim S, Hoehn K, Kleinstein S, Weisel F, Chikina M, Shlomchik M. Memory B cell subsets have divergent developmental origins that are coupled to distinct imprinted epigenetic states. Nat Immunol 2024; 25:562-575. [PMID: 38200277 PMCID: PMC12036331 DOI: 10.1038/s41590-023-01721-9] [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: 02/14/2023] [Accepted: 11/28/2023] [Indexed: 01/12/2024]
Abstract
Memory B cells (MBCs) are phenotypically and functionally diverse, but their developmental origins remain undefined. Murine MBCs can be divided into subsets by expression of CD80 and PD-L2. Upon re-immunization, CD80/PD-L2 double-negative (DN) MBCs spawn germinal center B cells (GCBCs), whereas CD80/PD-L2 double-positive (DP) MBCs generate plasmablasts but not GCBCs. Using multiple approaches, including generation of an inducible GCBC-lineage reporter mouse, we demonstrate in a T cell-dependent response that DN cells formed independently of the germinal center (GC), whereas DP cells exhibited either extrafollicular (DPEX) or GCBC (DPGC) origins. Chromatin and transcriptional profiling revealed similarity of DN cells with an early memory precursor. Reciprocally, GCBC-derived DP cells shared distinct genomic features with GCBCs, while DPEX cells had hybrid features. Upon restimulation, DPEX cells were more prone to divide, while DPGC cells differentiated toward IgG1+ plasmablasts. Thus, MBC functional diversity is generated through distinct developmental histories, which imprint characteristic epigenetic patterns onto their progeny, thereby programming them for divergent functional responses.
Collapse
Affiliation(s)
- Derrick Callahan
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Shuchi Smita
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Stephen Joachim
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kenneth Hoehn
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Steven Kleinstein
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Florian Weisel
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Maria Chikina
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mark Shlomchik
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
42
|
Koenig JFE, Knudsen NPH, Phelps A, Bruton K, Hoof I, Lund G, Libera DD, Lund A, Christensen LH, Glass DR, Walker TD, Fang A, Waserman S, Jordana M, Andersen PS. Type 2-polarized memory B cells hold allergen-specific IgE memory. Sci Transl Med 2024; 16:eadi0944. [PMID: 38324637 DOI: 10.1126/scitranslmed.adi0944] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 12/18/2023] [Indexed: 02/09/2024]
Abstract
Allergen-specific immunoglobulin E (IgE) antibodies mediate pathology in diseases such as allergic rhinitis and food allergy. Memory B cells (MBCs) contribute to circulating IgE by regenerating IgE-producing plasma cells upon allergen encounter. Here, we report a population of type 2-polarized MBCs defined as CD23hi, IL-4Rαhi, and CD32low at both the transcriptional and surface protein levels. These MBC2s are enriched in IgG1- and IgG4-expressing cells while constitutively expressing germline transcripts for IgE. Allergen-specific B cells from patients with allergic rhinitis and food allergy were enriched in MBC2s. Furthermore, MBC2s generated allergen-specific IgE during sublingual immunotherapy, thereby identifying these cells as a major reservoir for IgE. The identification of MBC2s provides insights into the maintenance of IgE memory, which is detrimental in allergic diseases but could be beneficial in protection against venoms and helminths.
Collapse
Affiliation(s)
- Joshua F E Koenig
- Schroeder Allergy and Immunology Research Institute, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | | | - Allyssa Phelps
- Schroeder Allergy and Immunology Research Institute, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Kelly Bruton
- Schroeder Allergy and Immunology Research Institute, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Ilka Hoof
- ALK-Abelló A/S, 2970 Hørsholm, Denmark
| | | | - Danielle Della Libera
- Schroeder Allergy and Immunology Research Institute, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | | | | | - David R Glass
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Tina D Walker
- Schroeder Allergy and Immunology Research Institute, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Allison Fang
- Schroeder Allergy and Immunology Research Institute, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Susan Waserman
- Schroeder Allergy and Immunology Research Institute, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Manel Jordana
- Schroeder Allergy and Immunology Research Institute, Faculty of Health Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | | |
Collapse
|
43
|
White TLA, Jin Y, Roberts SDA, Gable MJ, Morel PA. Phosphorylation of hnRNP A1-Serine 199 Is Not Required for T Cell Differentiation and Function. Immunohorizons 2024; 8:136-146. [PMID: 38334757 PMCID: PMC10916359 DOI: 10.4049/immunohorizons.2300074] [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: 09/11/2023] [Accepted: 01/05/2024] [Indexed: 02/10/2024] Open
Abstract
hnRNP A1 is an important RNA-binding protein that influences many stages of RNA processing, including transcription, alternative splicing, mRNA nuclear export, and RNA stability. However, the role of hnRNP A1 in immune cells, specifically CD4+ T cells, remains unclear. We previously showed that Akt phosphorylation of hnRNP A1 was dependent on TCR signal strength and was associated with Treg differentiation. To explore the impact of hnRNP A1 phosphorylation by Akt on CD4+ T cell differentiation, our laboratory generated a mutant mouse model, hnRNP A1-S199A (A1-MUT) in which the major Akt phosphorylation site on hnRNP A1 was mutated to alanine using CRISPR Cas9 technology. Immune profiling of A1-MUT mice revealed changes in the numbers of Tregs in the mesenteric lymph node. We found no significant differences in naive CD4+ T cell differentiation into Th1, Th2, Th17, or T regulatory cells (Tregs) in vitro. In vivo, Treg differentiation assays using OTII-A1-Mut CD4+ T cells exposed to OVA food revealed migration and homing defects in the A1-MUT but no change in Treg induction. A1-MUT mice were immunized with NP- keyhole limpet hemocyanin, and normal germinal center development, normal numbers of NP-specific B cells, and no change in Tfh numbers were observed. In conclusion, Akt phosphorylation of hnRNP A1 S199 does not play a role in CD4+ T cell fate or function in the models tested. This hnRNP A1-S199A mouse model should be a valuable tool to study the role of Akt phosphorylation of hnRNP A1-S199 in different cell types or other mouse models of human disease.
Collapse
Affiliation(s)
- Tristan L. A. White
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Ye Jin
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Sean D. A. Roberts
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Matthew J. Gable
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Penelope A. Morel
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
44
|
Quast I. B Cell Receptor Transgenic Mice as Tools to Study Memory B Cells. Methods Mol Biol 2024; 2826:131-139. [PMID: 39017890 DOI: 10.1007/978-1-0716-3950-4_10] [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: 07/18/2024]
Abstract
B cell receptor (BCR) transgenic mice allow the control of the initial target (antigen) specificity of naïve B cells and to investigate their properties following activation. Here, I describe how BCR transgenic B cells can be used in combination with adoptive cell transfer and immunization models to study memory B cell formation and reactivation.
Collapse
Affiliation(s)
- Isaak Quast
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
45
|
Gao F, Litchfield B, Wu H. Adipose tissue lymphocytes and obesity. THE JOURNAL OF CARDIOVASCULAR AGING 2024; 4:5. [PMID: 38455510 PMCID: PMC10919906 DOI: 10.20517/jca.2023.38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
Obesity is associated with chronic inflammation in adipose tissue (AT), mainly evidenced by infiltration and phenotypic changes of various types of immune cells. Macrophages are the major innate immune cells and represent the predominant immune cell population within AT. Lymphocytes, including T cells and B cells, are adaptive immune cells and constitute another important immune cell population in AT. In obesity, CD8+ effector memory T cells, CD4+ Th1 cells, and B2 cells are increased in AT and promote AT inflammation, while regulatory T cells and Th2 cells, which usually function as immune regulatory or type 2 inflammatory cells, are reduced in AT. Immune cells may regulate the metabolism of adipocytes and other cells through various mechanisms, contributing to the development of metabolic diseases, including insulin resistance and type 2 diabetes. Efforts targeting immune cells and inflammation to prevent and treat obesity-linked metabolic disease have been explored, but have not yielded significant success in clinical studies. This review provides a concise overview of the changes in lymphocyte populations within AT and their potential role in AT inflammation and the regulation of metabolic functions in the context of obesity.
Collapse
Affiliation(s)
- Feng Gao
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Huaizhu Wu
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
46
|
Hanson CH, Henry B, Andhare P, Lin FJ, Pak H, Turner JS, Adams LJ, Liu T, Fremont DH, Ellebedy AH, Laidlaw BJ. CD62L expression marks a functionally distinct subset of memory B cells. Cell Rep 2023; 42:113542. [PMID: 38060451 PMCID: PMC10842417 DOI: 10.1016/j.celrep.2023.113542] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/26/2023] [Accepted: 11/20/2023] [Indexed: 12/30/2023] Open
Abstract
The memory B cell response consists of phenotypically distinct subsets that differ in their ability to respond upon antigen re-encounter. However, the pathways regulating the development and function of memory B cell subsets are poorly understood. Here, we show that CD62L and CD44 are progressively expressed on mouse memory B cells and identify transcriptionally and functionally distinct memory B cell subsets. Bcl6 is important in regulating memory B cell subset differentiation with overexpression of Bcl6 resulting in impaired CD62L+ memory B cell development. Bcl6 regulates memory B cell subset development through control of a network of genes, including Bcl2 and Zeb2. Overexpression of Zeb2 impairs the development of CD62L+ memory B cells. Importantly, CD62L is also differentially expressed on human memory B cells following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination and identifies phenotypically distinct populations. Together, these data indicate that CD62L expression marks functionally distinct memory B cell subsets.
Collapse
Affiliation(s)
- Christopher H Hanson
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Brittany Henry
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Pradhnesh Andhare
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Frank J Lin
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Haley Pak
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Jackson S Turner
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Lucas J Adams
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tom Liu
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Daved H Fremont
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA; Department of Biochemistry and Molecular Biophysics, 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; The Andrew M. and Jane M. Bursky Center for Human Immunology & Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO, USA; Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, Saint Louis, MO, USA
| | - Brian J Laidlaw
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
47
|
Reyes RA, Batugedara G, Dutta P, Reers AB, Garza R, Ssewanyana I, Jagannathan P, Feeney ME, Greenhouse B, Bol S, Ay F, Bunnik EM. Atypical B cells consist of subsets with distinct functional profiles. iScience 2023; 26:108496. [PMID: 38098745 PMCID: PMC10720271 DOI: 10.1016/j.isci.2023.108496] [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: 08/30/2023] [Revised: 10/30/2023] [Accepted: 11/16/2023] [Indexed: 12/17/2023] Open
Abstract
Atypical B cells are a population of activated B cells that are commonly enriched in individuals with chronic immune activation but are also part of a normal immune response to infection or vaccination. To better define the role of atypical B cells in the human adaptive immune response, we performed single-cell sequencing of transcriptomes, cell surface markers, and B cell receptors in individuals with chronic exposure to the malaria parasite Plasmodium falciparum, a condition known to lead to accumulation of circulating atypical B cells. We identified three previously uncharacterized populations of atypical B cells with distinct transcriptional and functional profiles and observed marked differences among these three subsets in their ability to produce immunoglobulin G upon T-cell-dependent activation. Our findings help explain the conflicting observations in prior studies regarding the function of atypical B cells and highlight their different roles in the adaptive immune response in chronic inflammatory conditions.
Collapse
Affiliation(s)
- Raphael A. Reyes
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Gayani Batugedara
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Paramita Dutta
- Centers for Cancer Immunotherapy and Autoimmunity, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Ashley B. Reers
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Rolando Garza
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Isaac Ssewanyana
- Infectious Disease Research Collaboration, Kampala, Uganda
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Prasanna Jagannathan
- Department of Medicine, Division of Infectious Diseases, Stanford University, Stanford, CA 94305, USA
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, USA
| | - Margaret E. Feeney
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Bryan Greenhouse
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Sebastiaan Bol
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Ferhat Ay
- Centers for Cancer Immunotherapy and Autoimmunity, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | - Evelien M. Bunnik
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
48
|
Kare AJ, Nichols L, Zermeno R, Raie MN, Tumbale SK, Ferrara KW. OMIP-095: 40-Color spectral flow cytometry delineates all major leukocyte populations in murine lymphoid tissues. Cytometry A 2023; 103:839-850. [PMID: 37768325 PMCID: PMC10843696 DOI: 10.1002/cyto.a.24788] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 07/26/2023] [Accepted: 08/18/2023] [Indexed: 09/29/2023]
Abstract
High-dimensional immunoprofiling is essential for studying host response to immunotherapy, infection, and disease in murine model systems. However, the difficulty of multiparameter panel design combined with a lack of existing murine tools has prevented the comprehensive study of all major leukocyte phenotypes in a single assay. Herein, we present a 40-color flow cytometry panel for deep immunophenotyping of murine lymphoid tissues, including the spleen, blood, Peyer's patches, inguinal lymph nodes, bone marrow, and thymus. This panel uses a robust set of surface markers capable of differentiating leukocyte subsets without the use of intracellular staining, thus allowing for the use of cells in downstream functional experiments or multiomic analyses. Our panel classifies T cells, B cells, natural killer cells, innate lymphoid cells, monocytes, macrophages, dendritic cells, basophils, neutrophils, eosinophils, progenitors, and their functional subsets by using a series of co-stimulatory, checkpoint, activation, migration, and maturation markers. This tool has a multitude of systems immunology applications ranging from serial monitoring of circulating blood signatures to complex endpoint analysis, especially in pre-clinical settings where treatments can modulate leukocyte abundance and/or function. Ultimately, this 40-color panel resolves a diverse array of immune cells on the axes of time, tissue, and treatment, filling the niche for a modern tool dedicated to murine immunophenotyping.
Collapse
Affiliation(s)
- Aris J. Kare
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Lisa Nichols
- Stanford Shared FACS Facility, Stanford University, Stanford, CA 94305, USA
| | - Ricardo Zermeno
- Stanford Shared FACS Facility, Stanford University, Stanford, CA 94305, USA
| | - Marina N. Raie
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | | | | |
Collapse
|
49
|
Horiuchi S, Koike T, Takebuchi H, Hoshino K, Sasaki I, Fukuda-Ohta Y, Kaisho T, Kitamura D. SpiB regulates the expression of B-cell-related genes and increases the longevity of memory B cells. Front Immunol 2023; 14:1250719. [PMID: 37965309 PMCID: PMC10641807 DOI: 10.3389/fimmu.2023.1250719] [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: 06/30/2023] [Accepted: 09/18/2023] [Indexed: 11/16/2023] Open
Abstract
Generation of memory B cells is one of the key features of adaptive immunity as they respond rapidly to re-exposure to the antigen and generate functional antibodies. Although the functions of memory B cells are becoming clearer, the regulation of memory B cell generation and maintenance is still not well understood. Here we found that transcription factor SpiB is expressed in some germinal center (GC) B cells and memory B cells and participates in the maintenance of memory B cells. Overexpression and knockdown analyses revealed that SpiB suppresses plasma cell differentiation by suppressing the expression of Blimp1 while inducing Bach2 in the in-vitro-induced germinal center B (iGB) cell culture system, and that SpiB facilitates in-vivo appearance of memory-like B cells derived from the iGB cells. Further analysis in IgG1+ cell-specific SpiB conditional knockout (cKO) mice showed that function of SpiB is critical for the generation of late memory B cells but not early memory B cells or GC B cells. Gene expression analysis suggested that SpiB-dependent suppression of plasma cell differentiation is independent of the expression of Bach2. We further revealed that SpiB upregulates anti-apoptosis and autophagy genes to control the survival of memory B cells. These findings indicate the function of SpiB in the generation of long-lasting memory B cells to maintain humoral memory.
Collapse
Affiliation(s)
- Shu Horiuchi
- Division of Cancer Cell Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Takuya Koike
- Division of Cancer Cell Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Hirofumi Takebuchi
- Division of Cancer Cell Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Katsuaki Hoshino
- Department of Immunology, Faculty of Medicine, Kagawa University, Miki-cho, Kagawa, Japan
- Laboratory for Human Disease Models, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Izumi Sasaki
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yuri Fukuda-Ohta
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Tsuneyasu Kaisho
- Laboratory for Human Disease Models, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Daisuke Kitamura
- Division of Cancer Cell Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| |
Collapse
|
50
|
Tummolo A, Melpignano L. The Reciprocal Interplay between Infections and Inherited Metabolic Disorders. Microorganisms 2023; 11:2545. [PMID: 37894204 PMCID: PMC10608884 DOI: 10.3390/microorganisms11102545] [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: 08/31/2023] [Revised: 10/06/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Infections represent the main cause of acute metabolic derangements and/or the worsening of the clinical course of many inherited metabolic disorders (IMDs). The basic molecular mechanisms behind the role of infections in these conditions have not been completely clarified. This review points out the different mechanisms behind the relationship between IMDs and infections, providing an overview of this still-under-investigated area. Classically, infections have been considered as the consequence of a compromised immune system due to a biochemical defect of energy production. An adjunctive pathogenetic mechanism is related to a genetically altered protein-attached glycans composition, due to congenital glycosilation defects. In addition, a dietary regimen with a reduced intake of both micro- and macronutrients can potentially compromise the ability of the immune system to deal with an infection. There is recent pre-clinical evidence showing that during infections there may be a disruption of substrates of various metabolic pathways, leading to further cellular metabolic alteration. Therefore, infective agents may affect cellular metabolic pathways, by mediation or not of an altered immune system. The data reviewed here strongly suggest that the role of infections in many types of IMDs deserves greater attention for a better management of these disorders and a more focused therapeutic approach.
Collapse
Affiliation(s)
- Albina Tummolo
- Department of Metabolic Diseases, Clinical Genetics and Diabetology, Giovanni XXIII Children Hospital, Azienda Ospedaliero-Universitaria Consorziale, 70126 Bari, Italy
| | - Livio Melpignano
- Medical Direction, Giovanni XXIII Children Hospital, Azienda Ospedaliero-Universitaria Consorziale, 70126 Bari, Italy;
| |
Collapse
|