1
|
Chege Kuria T, Schneider A, Baraka F, Wanzek J, Vogg L, Brey S, Habenicht KM, Winkler TH. In vivo analysis of CRISPR-edited germinal center murine B cells. Front Immunol 2024; 15:1473760. [PMID: 39483469 PMCID: PMC11524869 DOI: 10.3389/fimmu.2024.1473760] [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: 07/31/2024] [Accepted: 09/30/2024] [Indexed: 11/03/2024] Open
Abstract
The germinal center (GC) reaction is crucial for somatic hypermutation, affinity maturation, and the selection of high-affinity B cells, all of which are hallmarks of the humoral immune response. Understanding the distinct roles of various B cell genes is essential for elucidating the selection mechanisms within the GC reaction. Traditionally, studying B cell gene function in the GC reaction involved generating knock-out mice, a highly time-consuming method that necessitates complex vectors. The advent of Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) technology has simplified the creation of knock-out mice. However, even with CRISPR, the generation of knock-out mice still faces challenges, including being time-consuming, costly, having low knock-out efficiency, and raising ethical concerns regarding animal use. To address these challenges, we developed an alternative method to traditional knock-out mouse generation. Our approach entails the ex vivo CRISPR editing of B cells from transgenic donor mice with different B cell receptor affinities followed by their adoptive transfer into recipient mice. We present a cost-effective, rapid, versatile, and adaptable CRISPR-Cas9 method for in vivo loss-of-function studies of individual murine B cell genes within the context of the GC reaction. This method provides a valuable tool for investigating the complex roles of different B cell genes in the GC selection process. As proof of concept, we validated our approach by examining the role of the pro-apoptotic gene Fas in the GC selection process. We adoptively transferred a mix of Fas knock-out (FasKO) low-affinity B cells, Fas wild-type (FasWT) low-affinity B cells, and FasWT high-affinity B cells into recipient mice. From our results, FasKO low-affinity B cells were still outcompeted by the FasWT high-affinity B cells for selection in the GC. An important observation was the accumulation of FasKO low-affinity GC B cells when compared to the FasWT low-affinity B cells, which suggested a role of Fas in the GC selection process.
Collapse
Affiliation(s)
- Timothy Chege Kuria
- Department of Biology, Division of Genetics, Nikolaus-Fiebiger-Center for Molecular
Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
2
|
Xu J, Lu W, Yang J, Liu X. B cells present a double-sided effect in digestive system tumors: a review for tumor microenvironment. Transl Gastroenterol Hepatol 2024; 9:46. [PMID: 39091659 PMCID: PMC11292093 DOI: 10.21037/tgh-23-127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 04/18/2024] [Indexed: 08/04/2024] Open
Abstract
Over the past few years, there has been an increasing interest in investigating tumor-infiltrating lymphocytes. B lymphocytes (B cells) are extensively distributed within tertiary lymphoid structure (TLS) as multifaceted subgroups and are intimately linked to the anti-tumor properties of TLS, as well as the survival and prognostication of individuals. While the investigation of T lymphocytes in the TLS has advanced to the level of clinical practice, the study of B cells remains limited. The principal impediment to the utilization of B cells in immunotherapy is their notable dual impact on tumors. Compared with tumors in other parts and systems, the function of B cells in the microenvironment of digestive system tumors to promote tumors proliferation, differentiation and migration cannot be ignored. Therefore, this review collects the studies of B cell subsets in tumor microenvironments, particularly related single cell sequencing research. The multifaceted role and function of B cells are investigated in esophageal, liver, colorectal, gastric and pancreatic cancers. And through the identification of B cell subsets and specific markers, this review attempts to explain the reasons why B cells produce different tumor-promoting effects in those tumors. The insights gleaned from this review may provide potential help and support the development of B cell-based immunotherapies.
Collapse
Affiliation(s)
- Jiaren Xu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Weiying Lu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Jinpu Yang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaosun Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
3
|
Hartmeier PR, Ostrowski SM, Busch EE, Empey KM, Meng WS. Lymphatic distribution considerations for subunit vaccine design and development. Vaccine 2024; 42:2519-2529. [PMID: 38494411 DOI: 10.1016/j.vaccine.2024.03.033] [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: 09/27/2023] [Revised: 01/30/2024] [Accepted: 03/13/2024] [Indexed: 03/19/2024]
Abstract
Subunit vaccines are an important platform for controlling current and emerging infectious diseases. The lymph nodes are the primary site generating the humoral response and delivery of antigens to these sites is critical to effective immunization. Indeed, the duration of antigen exposure within the lymph node is correlated with the antibody response. While current licensed vaccines are typically given through the intramuscular route, injecting vaccines subcutaneously allows for direct access to lymphatic vessels and therefore can enhance the transfer of antigen to the lymph nodes. However, protein subunit antigen uptake into the lymph nodes is inefficient, and subunit vaccines require adjuvants to stimulate the initial immune response. Therefore, formulation strategies have been developed to enhance the exposure of subunit proteins and adjuvants to the lymph nodes by increasing lymphatic uptake or prolonging the retention at the injection site. Given that lymph node exposure is a crucial consideration in vaccine design, in depth analyses of the pharmacokinetics of antigens and adjuvants should be the focus of future preclinical and clinical studies. This review will provide an overview of formulation strategies for targeting the lymphatics and prolonging antigen exposure and will discuss pharmacokinetic evaluations which can be applied toward vaccine development.
Collapse
Affiliation(s)
- Paul R Hartmeier
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, USA
| | - Sarah M Ostrowski
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, PA 15213, USA
| | - Emelia E Busch
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, USA
| | - Kerry M Empey
- Center for Clinical Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Pharmacy and Therapeutics, School of Pharmacy, University of Pittsburgh, PA 15213, USA; Department of Immunology, School of Medicine University of Pittsburgh, PA 15213, USA
| | - Wilson S Meng
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA 15219, USA.
| |
Collapse
|
4
|
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
|
5
|
Hie BL, Shanker VR, Xu D, Bruun TUJ, Weidenbacher PA, Tang S, Wu W, Pak JE, Kim PS. Efficient evolution of human antibodies from general protein language models. Nat Biotechnol 2024; 42:275-283. [PMID: 37095349 PMCID: PMC10869273 DOI: 10.1038/s41587-023-01763-2] [Citation(s) in RCA: 104] [Impact Index Per Article: 104.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 03/28/2023] [Indexed: 04/26/2023]
Abstract
Natural evolution must explore a vast landscape of possible sequences for desirable yet rare mutations, suggesting that learning from natural evolutionary strategies could guide artificial evolution. Here we report that general protein language models can efficiently evolve human antibodies by suggesting mutations that are evolutionarily plausible, despite providing the model with no information about the target antigen, binding specificity or protein structure. We performed language-model-guided affinity maturation of seven antibodies, screening 20 or fewer variants of each antibody across only two rounds of laboratory evolution, and improved the binding affinities of four clinically relevant, highly mature antibodies up to sevenfold and three unmatured antibodies up to 160-fold, with many designs also demonstrating favorable thermostability and viral neutralization activity against Ebola and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pseudoviruses. The same models that improve antibody binding also guide efficient evolution across diverse protein families and selection pressures, including antibiotic resistance and enzyme activity, suggesting that these results generalize to many settings.
Collapse
Affiliation(s)
- Brian L Hie
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA.
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA.
| | - Varun R Shanker
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Duo Xu
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Theodora U J Bruun
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Payton A Weidenbacher
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Shaogeng Tang
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Wesley Wu
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - John E Pak
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Peter S Kim
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA.
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
6
|
Pelissier A, Stratigopoulou M, Donner N, Dimitriadis E, Bende RJ, Guikema JE, Rodriguez Martinez M, van Noesel CJ. Convergent evolution and B-cell recirculation in germinal centers in a human lymph node. Life Sci Alliance 2023; 6:e202301959. [PMID: 37640448 PMCID: PMC10462906 DOI: 10.26508/lsa.202301959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/31/2023] Open
Abstract
Germinal centers (GCs) play a central role in generating an effective immune response against infectious pathogens, and failures in their regulating mechanisms can lead to the development of autoimmune diseases and cancer. Although previous works study experimental systems of the immune response with mouse models that are immunized with specific antigens, our study focused on a real-life situation, with an ongoing GC response in a human lymph node (LN) involving multiple asynchronized GCs reacting simultaneously to unknown antigens. We combined laser capture microdissection of individual GCs from human LN with next-generation repertoire sequencing to characterize individual GCs as distinct evolutionary spaces. In line with well-characterized GC responses in mice, elicited by immunization with model antigens, we observe a heterogeneous clonal diversity across individual GCs from the same human LN. Still, we identify shared clones in several individual GCs, and phylogenetic tree analysis combined with paratope modeling suggest the re-engagement and rediversification of B-cell clones across GCs and expanded clones exhibiting shared antigen responses across distinct GCs, indicating convergent evolution of the GCs.
Collapse
Affiliation(s)
- Aurelien Pelissier
- IBM Research Europe, Rüschlikon, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Maria Stratigopoulou
- Department of Pathology, Amsterdam University Medical Centers, Location AMC, Lymphoma and Myeloma Center Amsterdam, Amsterdam, Netherlands
| | - Naomi Donner
- Department of Pathology, Amsterdam University Medical Centers, Location AMC, Lymphoma and Myeloma Center Amsterdam, Amsterdam, Netherlands
| | | | - Richard J Bende
- Department of Pathology, Amsterdam University Medical Centers, Location AMC, Lymphoma and Myeloma Center Amsterdam, Amsterdam, Netherlands
| | - Jeroen E Guikema
- Department of Pathology, Amsterdam University Medical Centers, Location AMC, Lymphoma and Myeloma Center Amsterdam, Amsterdam, Netherlands
| | | | - Carel Jm van Noesel
- Department of Pathology, Amsterdam University Medical Centers, Location AMC, Lymphoma and Myeloma Center Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
7
|
Ozulumba T, Montalbine AN, Ortiz-Cárdenas JE, Pompano RR. New tools for immunologists: models of lymph node function from cells to tissues. Front Immunol 2023; 14:1183286. [PMID: 37234163 PMCID: PMC10206051 DOI: 10.3389/fimmu.2023.1183286] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/20/2023] [Indexed: 05/27/2023] Open
Abstract
The lymph node is a highly structured organ that mediates the body's adaptive immune response to antigens and other foreign particles. Central to its function is the distinct spatial assortment of lymphocytes and stromal cells, as well as chemokines that drive the signaling cascades which underpin immune responses. Investigations of lymph node biology were historically explored in vivo in animal models, using technologies that were breakthroughs in their time such as immunofluorescence with monoclonal antibodies, genetic reporters, in vivo two-photon imaging, and, more recently spatial biology techniques. However, new approaches are needed to enable tests of cell behavior and spatiotemporal dynamics under well controlled experimental perturbation, particularly for human immunity. This review presents a suite of technologies, comprising in vitro, ex vivo and in silico models, developed to study the lymph node or its components. We discuss the use of these tools to model cell behaviors in increasing order of complexity, from cell motility, to cell-cell interactions, to organ-level functions such as vaccination. Next, we identify current challenges regarding cell sourcing and culture, real time measurements of lymph node behavior in vivo and tool development for analysis and control of engineered cultures. Finally, we propose new research directions and offer our perspective on the future of this rapidly growing field. We anticipate that this review will be especially beneficial to immunologists looking to expand their toolkit for probing lymph node structure and function.
Collapse
Affiliation(s)
- Tochukwu Ozulumba
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
| | - Alyssa N. Montalbine
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, United States
| | - Jennifer E. Ortiz-Cárdenas
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Department of Bioengineering, Stanford University, Stanford, CA, United States
| | - Rebecca R. Pompano
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
- Carter Immunology Center and University of Virginia (UVA) Cancer Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
8
|
Notch Signaling in Acute Inflammation and Sepsis. Int J Mol Sci 2023; 24:ijms24043458. [PMID: 36834869 PMCID: PMC9967996 DOI: 10.3390/ijms24043458] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/27/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023] Open
Abstract
Notch signaling, a highly conserved pathway in mammals, is crucial for differentiation and homeostasis of immune cells. Besides, this pathway is also directly involved in the transmission of immune signals. Notch signaling per se does not have a clear pro- or anti-inflammatory effect, but rather its impact is highly dependent on the immune cell type and the cellular environment, modulating several inflammatory conditions including sepsis, and therefore significantly impacts the course of disease. In this review, we will discuss the contribution of Notch signaling on the clinical picture of systemic inflammatory diseases, especially sepsis. Specifically, we will review its role during immune cell development and its contribution to the modulation of organ-specific immune responses. Finally, we will evaluate to what extent manipulation of the Notch signaling pathway could be a future therapeutic strategy.
Collapse
|
9
|
Scharf S, Ackermann J, Bender L, Wurzel P, Schäfer H, Hansmann ML, Koch I. Holistic View on the Structure of Immune Response: Petri Net Model. Biomedicines 2023; 11:452. [PMID: 36830988 PMCID: PMC9953182 DOI: 10.3390/biomedicines11020452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/27/2023] [Accepted: 01/29/2023] [Indexed: 02/08/2023] Open
Abstract
The simulation of immune response is a challenging task because quantitative data are scarce. Quantitative theoretical models either focus on specific cell-cell interactions or have to make assumptions about parameters. The broad variation of, e.g., the dimensions and abundance between lymph nodes as well as between individual patients hampers conclusive quantitative modeling. No theoretical model has been established representing a consensus on the set of major cellular processes involved in the immune response. In this paper, we apply the Petri net formalism to construct a semi-quantitative mathematical model of the lymph nodes. The model covers the major cellular processes of immune response and fulfills the formal requirements of Petri net models. The intention is to develop a model taking into account the viewpoints of experienced pathologists and computer scientists in the field of systems biology. In order to verify formal requirements, we discuss invariant properties and apply the asynchronous firing rule of a place/transition net. Twenty-five transition invariants cover the model, and each is assigned to a functional mode of the immune response. In simulations, the Petri net model describes the dynamic modes of the immune response, its adaption to antigens, and its loss of memory.
Collapse
Affiliation(s)
- Sonja Scharf
- Molecular Bioinformatics, Institute of Computer Science, Goethe University Frankfurt, Robert-Mayer Str. 11-15, 60325 Frankfurt am Main, Germany
- Frankfurt Institute for Advanced Studies, Ruth-Moufang-Str. 1, 60438 Frankfurt am Main, Germany
- Institute of General Pharmacology and Toxicology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Jörg Ackermann
- Molecular Bioinformatics, Institute of Computer Science, Goethe University Frankfurt, Robert-Mayer Str. 11-15, 60325 Frankfurt am Main, Germany
| | - Leonie Bender
- Molecular Bioinformatics, Institute of Computer Science, Goethe University Frankfurt, Robert-Mayer Str. 11-15, 60325 Frankfurt am Main, Germany
| | - Patrick Wurzel
- Molecular Bioinformatics, Institute of Computer Science, Goethe University Frankfurt, Robert-Mayer Str. 11-15, 60325 Frankfurt am Main, Germany
- Frankfurt Institute for Advanced Studies, Ruth-Moufang-Str. 1, 60438 Frankfurt am Main, Germany
- Institute of General Pharmacology and Toxicology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Hendrik Schäfer
- Institute of General Pharmacology and Toxicology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
- Institute of Pathology, Corporate Member of Free University of Berlin, Humboldt-University of Berlin, Charity-University Medicine Berlin, Virchowweg 15, 10117 Berlin, Germany
| | - Martin-Leo Hansmann
- Frankfurt Institute for Advanced Studies, Ruth-Moufang-Str. 1, 60438 Frankfurt am Main, Germany
- Institute of General Pharmacology and Toxicology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Ina Koch
- Molecular Bioinformatics, Institute of Computer Science, Goethe University Frankfurt, Robert-Mayer Str. 11-15, 60325 Frankfurt am Main, Germany
| |
Collapse
|
10
|
Yan Z, Qi H, Lan Y. The role of geometric features in a germinal center. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2022; 19:8304-8333. [PMID: 35801467 DOI: 10.3934/mbe.2022387] [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: 06/15/2023]
Abstract
The germinal center (GC) is a self-organizing structure produced in the lymphoid follicle during the T-dependent immune response and is an important component of the humoral immune system. However, the impact of the special structure of GC on antibody production is not clear. According to the latest biological experiments, we establish a spatiotemporal stochastic model to simulate the whole self-organization process of the GC including the appearance of two specific zones: the dark zone (DZ) and the light zone (LZ), the development of which serves to maintain an effective competition among different cells and promote affinity maturation. A phase transition is discovered in this process, which determines the critical GC volume for a successful growth in both the stochastic and the deterministic model. Further increase of the volume does not make much improvement on the performance. It is found that the critical volume is determined by the distance between the activated B cell receptor (BCR) and the target epitope of the antigen in the shape space. The observation is confirmed in both 2D and 3D simulations and explains partly the variability of the observed GC size.
Collapse
Affiliation(s)
- Zishuo Yan
- School of Science, Beijing University of Posts and Telecommunications, Beijing 100876, China
| | - Hai Qi
- Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing 100084, China
| | - Yueheng Lan
- School of Science, Beijing University of Posts and Telecommunications, Beijing 100876, China
| |
Collapse
|
11
|
Vaidehi Narayanan H, Hoffmann A. From Antibody Repertoires to Cell-Cell Interactions to Molecular Networks: Bridging Scales in the Germinal Center. Front Immunol 2022; 13:898078. [PMID: 35603162 PMCID: PMC9114758 DOI: 10.3389/fimmu.2022.898078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 04/08/2022] [Indexed: 01/02/2023] Open
Abstract
Antibody-mediated adaptive immunity must provide effective long-term protection with minimal adverse effects, against rapidly mutating pathogens, in a human population with diverse ages, genetics, and immune histories. In order to grasp and leverage the complexities of the antibody response, we advocate for a mechanistic understanding of the multiscale germinal center (GC) reaction - the process by which precursor B-cells evolve high-affinity antigen-specific antibodies, forming an effector repertoire of plasma and memory cells for decades-long protection. The regulatory dynamics of B-cells within the GC are complex, and unfold across multiple interacting spatial and temporal scales. At the organism scale, over weeks to years, the antibody sequence repertoire formed by various B-cell clonal lineages modulates antibody quantity and quality over time. At the tissue and cellular scale, over hours to weeks, B-cells undergo selection via spatially distributed interactions with local stroma, antigen, and helper T-cells. At the molecular scale, over seconds to days, intracellular signaling, transcriptional, and epigenetic networks modulate B-cell fates and shape their clonal lineages. We summarize our current understanding within each of these scales, and identify missing links in connecting them. We suggest that quantitative multi-scale mathematical models of B-cell and GC reaction dynamics provide predictive frameworks that can apply basic immunological knowledge to practical challenges such as rational vaccine design.
Collapse
Affiliation(s)
| | - Alexander Hoffmann
- Signaling Systems Laboratory, Department of Microbiology, Immunology, and Molecular Genetics, and Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
12
|
Atitey K, Anchang B. Mathematical Modeling of Proliferative Immune Response Initiated by Interactions Between Classical Antigen-Presenting Cells Under Joint Antagonistic IL-2 and IL-4 Signaling. Front Mol Biosci 2022; 9:777390. [PMID: 35155574 PMCID: PMC8831889 DOI: 10.3389/fmolb.2022.777390] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 01/03/2022] [Indexed: 11/13/2022] Open
Abstract
During an adaptive immune response from pathogen invasion, multiple cytokines are produced by various immune cells interacting jointly at the cellular level to mediate several processes. For example, studies have shown that regulation of interleukin-4 (IL-4) correlates with interleukin-2 (IL-2) induced lymphocyte proliferation. This motivates the need to better understand and model the mechanisms driving the dynamic interplay of proliferation of lymphocytes with the complex interaction effects of cytokines during an immune response. To address this challenge, we adopt a hybrid computational approach comprising of continuous, discrete and stochastic non-linear model formulations to predict a system-level immune response as a function of multiple dependent signals and interacting agents including cytokines and targeted immune cells. We propose a hybrid ordinary differential equation-based (ODE) multicellular model system with a stochastic component of antigen microscopic states denoted as Multiscale Multicellular Quantitative Evaluator (MMQE) implemented using MATLAB. MMQE combines well-defined immune response network-based rules and ODE models to capture the complex dynamic interactions between the proliferation levels of different types of communicating lymphocyte agents mediated by joint regulation of IL-2 and IL-4 to predict the emergent global behavior of the system during an immune response. We model the activation of the immune system in terms of different activation protocols of helper T cells by the interplay of independent biological agents of classic antigen-presenting cells (APCs) and their joint activation which is confounded by the exposure time to external pathogens. MMQE quantifies the dynamics of lymphocyte proliferation during pathogen invasion as bivariate distributions of IL-2 and IL-4 concentration levels. Specifically, by varying activation agents such as dendritic cells (DC), B cells and their joint mechanism of activation, we quantify how lymphocyte activation and differentiation protocols boost the immune response against pathogen invasion mediated by a joint downregulation of IL-4 and upregulation of IL-2. We further compare our in-silico results to in-vivo and in-vitro experimental studies for validation. In general, MMQE combines intracellular and extracellular effects from multiple interacting systems into simpler dynamic behaviors for better interpretability. It can be used to aid engineering of anti-infection drugs or optimizing drug combination therapies against several diseases.
Collapse
|
13
|
Kealy L, Good-Jacobson KL. Advances in understanding the formation and fate of B-cell memory in response to immunization or infection. OXFORD OPEN IMMUNOLOGY 2021; 2:iqab018. [PMID: 36845573 PMCID: PMC8499879 DOI: 10.1093/oxfimm/iqab018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/06/2021] [Accepted: 09/01/2021] [Indexed: 02/07/2023] Open
Abstract
Immunological memory has the potential to provide lifelong protection against recurrent infections. As such, it has been crucial to the success of vaccines. Yet, the recent pandemic has illuminated key gaps in our knowledge related to the factors influencing effective memory formation and the inability to predict the longevity of immune protection. In recent decades, researchers have acquired a number of novel and powerful tools with which to study the factors underpinning humoral memory. These tools have been used to study the B-cell fate decisions that occur within the germinal centre (GC), a site where responding B cells undergo affinity maturation and are one of the major routes for memory B cell and high-affinity long-lived plasma cell formation. The advent of single-cell sequencing technology has provided an enhanced resolution for studying fate decisions within the GC and cutting-edge techniques have enabled researchers to model this reaction with more accuracy both in vitro and in silico. Moreover, modern approaches to studying memory B cells have allowed us to gain a better appreciation for the heterogeneity and adaptability of this vital class of B cells. Together, these studies have facilitated important breakthroughs in our understanding of how these systems operate to ensure a successful immune response. In this review, we describe recent advances in the field of GC and memory B-cell biology in order to provide insight into how humoral memory is formed, as well as the potential for generating lasting immunity to novel pathogens such as severe acute respiratory syndrome coronavirus 2.
Collapse
Affiliation(s)
- Liam Kealy
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia,Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Kim L Good-Jacobson
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia,Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia,Correspondence address. Department of Biochemistry and Molecular Biology, Monash University, Ground floor reception, 23 Innovation Walk (Bldg 77), Clayton, Victoria 3800 Australia. Tel: (+613) 990-29510; E-mail: ; Twitter: @KimLJacobson
| |
Collapse
|
14
|
Abstract
This perspective article gathers the latest developments in mathematical and computational oncology tools that exploit network approaches for the mathematical modelling, analysis, and simulation of cancer development and therapy design. It instigates the community to explore new paths and synergies under the umbrella of the Special Issue “Networks in Cancer: From Symmetry Breaking to Targeted Therapy”. The focus of the perspective is to demonstrate how networks can model the physics, analyse the interactions, and predict the evolution of the multiple processes behind tumour-host encounters across multiple scales. From agent-based modelling and mechano-biology to machine learning and predictive modelling, the perspective motivates a methodology well suited to mathematical and computational oncology and suggests approaches that mark a viable path towards adoption in the clinic.
Collapse
|
15
|
Reimer D, Meyer-Hermann M, Rakhymzhan A, Steinmetz T, Tripal P, Thomas J, Boettcher M, Mougiakakos D, Schulz SR, Urbanczyk S, Hauser AE, Niesner RA, Mielenz D. B Cell Speed and B-FDC Contacts in Germinal Centers Determine Plasma Cell Output via Swiprosin-1/EFhd2. Cell Rep 2021; 32:108030. [PMID: 32783949 DOI: 10.1016/j.celrep.2020.108030] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 04/15/2020] [Accepted: 07/22/2020] [Indexed: 12/20/2022] Open
Abstract
Plasma cells secreting affinity-matured antibodies develop in germinal centers (GCs), where B cells migrate persistently and directionally over defined periods of time. How modes of GC B cell migration influence plasma cell development remained unclear. Through genetic deletion of the F-actin bundling protein Swiprosin-1/EF-hand domain family member 2 (EFhd2) and by two-photon microscopy, we show that EFhd2 restrains B cell speed in GCs and hapten-specific plasma cell output. Modeling the GC reaction reveals that increasing GC B cell speed promotes plasma cell generation. Lack of EFhd2 also reduces contacts of GC B cells with follicular dendritic cells in vivo. Computational modeling uncovers that both GC output and antibody affinity depend quantitatively on contacts of GC B cells with follicular dendritic cells when B cells migrate more persistently. Collectively, our data explain how GC B cells integrate speed and persistence of cell migration with B cell receptor affinity.
Collapse
Affiliation(s)
- Dorothea Reimer
- Division of Molecular Immunology, Universitätsklinikum Erlangen, Nikolaus-Fiebiger-Zentrum, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Meyer-Hermann
- Department of Systems Immunology and Braunschweig, Integrated Centre of Systems Biology, Helmholtz Center for Infection Research, Braunschweig, Germany
| | | | - Tobit Steinmetz
- Division of Molecular Immunology, Universitätsklinikum Erlangen, Nikolaus-Fiebiger-Zentrum, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Philipp Tripal
- Optical Imaging Center (OICE), Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Jana Thomas
- Division of Molecular Immunology, Universitätsklinikum Erlangen, Nikolaus-Fiebiger-Zentrum, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Boettcher
- Department of Internal Medicine V, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Dimitrios Mougiakakos
- Department of Internal Medicine V, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Sebastian R Schulz
- Division of Molecular Immunology, Universitätsklinikum Erlangen, Nikolaus-Fiebiger-Zentrum, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Sophia Urbanczyk
- Division of Molecular Immunology, Universitätsklinikum Erlangen, Nikolaus-Fiebiger-Zentrum, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Anja E Hauser
- Deutsches Rheumaforschungszentrum (DRFZ), Berlin, Germany; Charité - University Medicine, Berlin, Germany
| | - Raluca A Niesner
- Deutsches Rheumaforschungszentrum (DRFZ), Berlin, Germany; Dynamic and Functional In Vivo Imaging, Veterinary Medicine, Freie Universität, Berlin, Germany
| | - Dirk Mielenz
- Division of Molecular Immunology, Universitätsklinikum Erlangen, Nikolaus-Fiebiger-Zentrum, FAU Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
16
|
Abstract
Memory B cells (MBCs) are critical for the rapid development of protective immunity following re-infection. MBCs capable of neutralizing distinct subclasses of pathogens, such as influenza and HIV, have been identified in humans. However, efforts to develop vaccines that induce broadly protective MBCs to rapidly mutating pathogens have not yet been successful. Better understanding of the signals regulating MBC development and function are essential to overcome current challenges hindering successful vaccine development. Here, we discuss recent advancements regarding the signals and transcription factors regulating germinal centre-derived MBC development and function.
Collapse
Affiliation(s)
- Brian J Laidlaw
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| | - Jason G Cyster
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
17
|
Merino Tejero E, Lashgari D, García-Valiente R, Gao X, Crauste F, Robert PA, Meyer-Hermann M, Martínez MR, van Ham SM, Guikema JEJ, Hoefsloot H, van Kampen AHC. Multiscale Modeling of Germinal Center Recapitulates the Temporal Transition From Memory B Cells to Plasma Cells Differentiation as Regulated by Antigen Affinity-Based Tfh Cell Help. Front Immunol 2021; 11:620716. [PMID: 33613551 PMCID: PMC7892951 DOI: 10.3389/fimmu.2020.620716] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/21/2020] [Indexed: 01/10/2023] Open
Abstract
Germinal centers play a key role in the adaptive immune system since they are able to produce memory B cells and plasma cells that produce high affinity antibodies for an effective immune protection. The mechanisms underlying cell-fate decisions are not well understood but asymmetric division of antigen, B-cell receptor affinity, interactions between B-cells and T follicular helper cells (triggering CD40 signaling), and regulatory interactions of transcription factors have all been proposed to play a role. In addition, a temporal switch from memory B-cell to plasma cell differentiation during the germinal center reaction has been shown. To investigate if antigen affinity-based Tfh cell help recapitulates the temporal switch we implemented a multiscale model that integrates cellular interactions with a core gene regulatory network comprising BCL6, IRF4, and BLIMP1. Using this model we show that affinity-based CD40 signaling in combination with asymmetric division of B-cells result in switch from memory B-cell to plasma cell generation during the course of the germinal center reaction. We also show that cell fate division is unlikely to be (solely) based on asymmetric division of Ag but that BLIMP1 is a more important factor. Altogether, our model enables to test the influence of molecular modulations of the CD40 signaling pathway on the production of germinal center output cells.
Collapse
Affiliation(s)
- Elena Merino Tejero
- Bioinformatics Laboratory, Epidemiology and Data Science, Amsterdam Public Health Research Institute, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Danial Lashgari
- Bioinformatics Laboratory, Epidemiology and Data Science, Amsterdam Public Health Research Institute, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Rodrigo García-Valiente
- Bioinformatics Laboratory, Epidemiology and Data Science, Amsterdam Public Health Research Institute, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Xuefeng Gao
- Department of Hematology and Oncology, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Health Science Center, Shenzhen, China
| | | | - Philippe A Robert
- Department for Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Michael Meyer-Hermann
- Department for Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | | | - S Marieke van Ham
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Jeroen E J Guikema
- Department of Pathology, Lymphoma and Myeloma Center Amsterdam (LYMMCARE), Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Huub Hoefsloot
- Biosystems Data Analysis, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Antoine H C van Kampen
- Bioinformatics Laboratory, Epidemiology and Data Science, Amsterdam Public Health Research Institute, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands.,Biosystems Data Analysis, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
18
|
Wittmann J. Modeling Lymphocytes. SYSTEMS MEDICINE 2021. [DOI: 10.1016/b978-0-12-801238-3.11608-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
19
|
Liang T, Guo Y, Li M, Ding C, Sang S, Zhou T, Shao Q, Liu X, Lu J, Ji Z, Wang T, Kang Q. Cytoskeleton protein 4.1R regulates B-cell fate by modulating the canonical NF-κB pathway. Immunology 2020; 161:314-324. [PMID: 32852059 DOI: 10.1111/imm.13250] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 06/16/2020] [Accepted: 08/12/2020] [Indexed: 11/29/2022] Open
Abstract
During the immune response, B cells can enter the memory pathway, which is characterized by class switch recombination (CSR), or they may undergo plasma cell differentiation (PCD) to secrete immunoglobulin. Both of these processes occur in activated B cells, which are reported to relate to membrane-association proteins and adaptors. Protein 4.1R acts as an adaptor, linking membrane proteins to the cytoskeleton, and is involved in many cell events such as cell activation and differentiation, and cytokine secretion. However, the effect of 4.1R on regulating B-cell fate is unclear. Here, we show an important association between B-cell fate and 4.1R. In vitro, primary B cells were stimulated with lipopolysaccharide combined with interleukin-4; results showed that 4.1R-deficient (4.1R-/- ) cells compared with wild-type (4.1R+/+ ) B cells augmented expression of activation-induced cytidine deaminase and germline, resulting in increased IgG1+ B cells, whereas the secretion of IgG1 and IgM was reduced, and CD138+ B cells were also decreased. Throughout the process, 4.1R regulated canonical nuclear factor (NF-κB) rather than non-canonical NF-κB to promote the expression of CSR complex components, leading to up-regulation of B-cell CSR. In contrast, 4.1R-deficient B cells showed reduced expression of Blimp-1, which caused B cells to down-regulate PCD. Furthermore, over-activation of canonical NF-κB may induce apoptosis signaling to cause PCD apoptosis to reduce PCD number. In summary, our results suggest that 4.1R acts as a B-cell fate regulator by inhibiting the canonical NF-κB signaling pathway.
Collapse
Affiliation(s)
- Taotao Liang
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Yuying Guo
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengjia Li
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Cong Ding
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Siyao Sang
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Tingting Zhou
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Qi Shao
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Xin Liu
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Jike Lu
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Zhenyu Ji
- Henan Academy of Medical and Pharmaceutical Sciences, Zhengzhou, China
| | - Ting Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Qiaozhen Kang
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
20
|
Pélissier A, Akrout Y, Jahn K, Kuipers J, Klein U, Beerenwinkel N, Rodríguez Martínez M. Computational Model Reveals a Stochastic Mechanism behind Germinal Center Clonal Bursts. Cells 2020; 9:E1448. [PMID: 32532145 PMCID: PMC7349200 DOI: 10.3390/cells9061448] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/29/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023] Open
Abstract
Germinal centers (GCs) are specialized compartments within the secondary lymphoid organs where B cells proliferate, differentiate, and mutate their antibody genes in response to the presence of foreign antigens. Through the GC lifespan, interclonal competition between B cells leads to increased affinity of the B cell receptors for antigens accompanied by a loss of clonal diversity, although the mechanisms underlying clonal dynamics are not completely understood. We present here a multi-scale quantitative model of the GC reaction that integrates an intracellular component, accounting for the genetic events that shape B cell differentiation, and an extracellular stochastic component, which accounts for the random cellular interactions within the GC. In addition, B cell receptors are represented as sequences of nucleotides that mature and diversify through somatic hypermutations. We exploit extensive experimental characterizations of the GC dynamics to parameterize our model, and visualize affinity maturation by means of evolutionary phylogenetic trees. Our explicit modeling of B cell maturation enables us to characterise the evolutionary processes and competition at the heart of the GC dynamics, and explains the emergence of clonal dominance as a result of initially small stochastic advantages in the affinity to antigen. Interestingly, a subset of the GC undergoes massive expansion of higher-affinity B cell variants (clonal bursts), leading to a loss of clonal diversity at a significantly faster rate than in GCs that do not exhibit clonal dominance. Our work contributes towards an in silico vaccine design, and has implications for the better understanding of the mechanisms underlying autoimmune disease and GC-derived lymphomas.
Collapse
Affiliation(s)
- Aurélien Pélissier
- IBM Research Zurich, 8803 Rüschlikon, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland; (K.J.); (J.K.); (N.B.)
| | | | - Katharina Jahn
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland; (K.J.); (J.K.); (N.B.)
| | - Jack Kuipers
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland; (K.J.); (J.K.); (N.B.)
| | - Ulf Klein
- Leeds Institute of Medical Research at St. James’s, University of Leeds, Leeds LS9 7TF, UK;
| | - Niko Beerenwinkel
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland; (K.J.); (J.K.); (N.B.)
| | | |
Collapse
|
21
|
Zhang J, Kodali S, Chen M, Wang J. Maintenance of Germinal Center B Cells by Caspase-9 through Promotion of Apoptosis and Inhibition of Necroptosis. THE JOURNAL OF IMMUNOLOGY 2020; 205:113-120. [PMID: 32434938 DOI: 10.4049/jimmunol.2000359] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 04/28/2020] [Indexed: 12/17/2022]
Abstract
In response to T cell-dependent Ag encounter, naive B cells develop into germinal center (GC) B cells, which can further differentiate into Ab-secreting plasma cells or memory B cells. GC B cells are short lived and are prone to caspase-mediated apoptosis. However, how apoptotic caspases regulate GC B cell fate has not been fully characterized. In this study, we show that mice with B cell-specific knockout of caspase-9 had decreases in GC B cells and Ab production after immunization. Caspase-9-deficient B cells displayed defects in caspase-dependent apoptosis but increases in necroptosis signaling. Additional deletion of Ripk3 restored GC B cells and Ab production in mice with B cell-specific knockout of caspase-9. Our results indicate that caspase-9 plays an important role in the maintenance of Ab responses by promoting apoptosis and inhibiting necroptosis in B cells.
Collapse
Affiliation(s)
- Jingting Zhang
- Immunobiology and Transplant Science Center, Houston Methodist Research Institute, Houston, TX 77030
| | - Srikanth Kodali
- Immunobiology and Transplant Science Center, Houston Methodist Research Institute, Houston, TX 77030.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030
| | - Min Chen
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, TX 77030; and
| | - Jin Wang
- Immunobiology and Transplant Science Center, Houston Methodist Research Institute, Houston, TX 77030; .,Department of Surgery, Weill Cornell Medical College, Cornell University, New York, NY 10065
| |
Collapse
|
22
|
Mitchell S. What Will B Will B: Identifying Molecular Determinants of Diverse B-Cell Fate Decisions Through Systems Biology. Front Cell Dev Biol 2020; 8:616592. [PMID: 33511125 PMCID: PMC7835399 DOI: 10.3389/fcell.2020.616592] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 12/02/2020] [Indexed: 12/25/2022] Open
Abstract
B-cells are the poster child for cellular diversity and heterogeneity. The diverse repertoire of B lymphocytes, each expressing unique antigen receptors, provides broad protection against pathogens. However, B-cell diversity goes beyond unique antigen receptors. Side-stepping B-cell receptor (BCR) diversity through BCR-independent stimuli or engineered organisms with monoclonal BCRs still results in seemingly identical B-cells reaching a wide variety of fates in response to the same challenge. Identifying to what extent the molecular state of a B-cell determines its fate is key to gaining a predictive understanding of B-cells and consequently the ability to control them with targeted therapies. Signals received by B-cells through transmembrane receptors converge on intracellular molecular signaling networks, which control whether each B-cell divides, dies, or differentiates into a number of antibody-secreting distinct B-cell subtypes. The signaling networks that interpret these signals are well known to be susceptible to molecular variability and noise, providing a potential source of diversity in cell fate decisions. Iterative mathematical modeling and experimental studies have provided quantitative insight into how B-cells achieve distinct fates in response to pathogenic stimuli. Here, we review how systems biology modeling of B-cells, and the molecular signaling networks controlling their fates, is revealing the key determinants of cell-to-cell variability in B-cell destiny.
Collapse
|
23
|
Arulraj T, Binder SC, Robert PA, Meyer-Hermann M. Synchronous Germinal Center Onset Impacts the Efficiency of Antibody Responses. Front Immunol 2019; 10:2116. [PMID: 31555300 PMCID: PMC6742702 DOI: 10.3389/fimmu.2019.02116] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 08/22/2019] [Indexed: 12/25/2022] Open
Abstract
The germinal center reaction is an important target for modulating antibody responses. Antibody production from germinal centers is regulated by a negative feedback mechanism termed antibody feedback. By imposing antibody feedback, germinal centers can interact and regulate the output of other germinal centers. Using an agent-based model of the germinal center reaction, we studied the impact of antibody feedback on kinetics and efficiency of a germinal center. Our simulations predict that high feedback of antibodies from germinal centers reduces the production of plasma cells and subsequently the efficiency of the germinal center reaction by promoting earlier termination. Affinity maturation is only weakly improved by increased antibody feedback and ultimately interrupted because of premature termination of the reaction. The model predicts that the asynchronous onset and changes in number of germinal centers could alter the efficiency of antibody response due to changes in feedback by soluble antibodies. Consequently, late initialized germinal centers have a compromised output due to higher antibody feedback from the germinal centers formed earlier. The results demonstrate potential effects of germinal center intercommunication and highlight the importance of understanding germinal center interactions for optimizing the antibody response, in particular, in the elderly and in the context of vaccination.
Collapse
Affiliation(s)
- Theinmozhi Arulraj
- Department of Systems Immunology, Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Sebastian C Binder
- Department of Systems Immunology, Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Centre for Individualized Infection Medicine (CIIM), Hanover, Germany
| | - Philippe A Robert
- Department of Systems Immunology, Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Michael Meyer-Hermann
- Department of Systems Immunology, Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Centre for Individualized Infection Medicine (CIIM), Hanover, Germany.,Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| |
Collapse
|