1
|
Bousbaine D, Obeng EM, Li Z, Fang T, Cheloha RW, Ploegh HL, McCaul N. Site-specific labeling uncovers differences in levels and distribution of B-cell receptors of different isotypes on primary B cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf062. [PMID: 40314988 DOI: 10.1093/jimmun/vkaf062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 02/24/2025] [Indexed: 05/03/2025]
Abstract
Expression of the BCR is essential for survival, development, and effector functions of B cells. Naive B cells express surface IgM and IgD, while surface IgG1 is expressed by class-switched (memory) B cells. Despite similar overall structures, the different BCR isotypes show differences in distribution and expression levels. The dynamics of BCR behavior have been difficult to explore owing to a lack of appropriate tools that can track the BCR without causing concomitant activation. Using CRISPR-Cas9, we inserted a sortase recognition motif (LPETG [LeuProGluThrGly]) at the C-terminus of the OB1 transnuclear ovalbumin-specific Cκ chain (Igκ-LPETG mice). The surface BCR from Igκ-LPETG mice is fully functional and can be labeled site-specifically with biotin or fluorophores. Igκ-LPETG mice show near-normal B-cell development, with an increase in Igλ-producing cells, presumably due to massive contraction of the κ locus V-region cluster upon V-J recombination to generate the OB1 light chain. Using the Igκ-LPETG mice, we compared organization and density of BCRs on the surface of IgM/IgD+ B cells bearing a wild-type (WT) heavy chain locus and IgG1 B cells in the OB1 model. The density of IgG1 BCRs is much reduced compared to IgM/IgD BCRs on primary B cells. Upon activation, IgM/IgD BCRs are found in detergent-insoluble domains, whereas IgG1 BCRs are not. The isotype of the Ig heavy chain thus contributes to surface expression and nanoscale organization of the BCR.
Collapse
Affiliation(s)
- Djenet Bousbaine
- Microbiology Graduate Program, MIT, Cambridge, MA, United States
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, United States
| | - Eugene M Obeng
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, United States
| | - Zeyang Li
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, United States
| | - Tao Fang
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, United States
| | - Ross W Cheloha
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, United States
- Laboratory of Bioorganic Chemistry; National Institute of Diabetes, Digestive, and Kidney Diseases; National Institutes of Health, Bethesda, MD, United States
| | - Hidde L Ploegh
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, United States
| | - Nicholas McCaul
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, United States
| |
Collapse
|
2
|
Degn SE, Tolar P. Towards a unifying model for B-cell receptor triggering. Nat Rev Immunol 2025; 25:77-91. [PMID: 39256626 DOI: 10.1038/s41577-024-01073-x] [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: 07/16/2024] [Indexed: 09/12/2024]
Abstract
Antibodies are exceptionally versatile molecules with remarkable flexibility in their binding properties. Their natural targets range from small-molecule toxins, across viruses of different sizes, to bacteria and large multicellular parasites. The molecular determinants bound by antibodies include proteins, peptides, carbohydrates, nucleic acids, lipids and even synthetic molecules that have never existed in nature. Membrane-anchored antibodies also serve as receptors on the surface of the B cells that produce them. Despite recent structural insights, there is still no unifying molecular mechanism to explain how antibody targets (antigens) trigger the activation of these B-cell receptors (BCRs). After cognate antigen encounter, somatic hypermutation and class-switch recombination allow BCR affinity maturation and immunoglobulin class-specific responses, respectively. This raises the fundamental question of how one receptor activation mechanism can accommodate a plethora of variant receptors and ligands, and how it can ensure that individual B cells remain responsive to antigen after somatic hypermutation and class switching. There is still no definite answer. Here we give a brief historical account of the different models proposed to explain BCR triggering and discuss their merit in the context of the current knowledge of the structure of BCRs, their dynamic membrane distribution, and recent biochemical and cell biological insights.
Collapse
Affiliation(s)
- Søren E Degn
- Laboratory for Lymphocyte Biology, Department of Biomedicine, Aarhus University, Aarhus, Denmark.
- Centre for Cellular Signal Patterns (CellPAT), Aarhus University, Aarhus, Denmark.
| | - Pavel Tolar
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| |
Collapse
|
3
|
Masnikosa R, Cvetković Z, Pirić D. Tumor Biology Hides Novel Therapeutic Approaches to Diffuse Large B-Cell Lymphoma: A Narrative Review. Int J Mol Sci 2024; 25:11384. [PMID: 39518937 PMCID: PMC11545713 DOI: 10.3390/ijms252111384] [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: 07/14/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is a malignancy of immense biological and clinical heterogeneity. Based on the transcriptomic or genomic approach, several different classification schemes have evolved over the years to subdivide DLBCL into clinically (prognostically) relevant subsets, but each leaves unclassified samples. Herein, we outline the DLBCL tumor biology behind the actual and potential drug targets and address the challenges and drawbacks coupled with their (potential) use. Therapeutic modalities are discussed, including small-molecule inhibitors, naked antibodies, antibody-drug conjugates, chimeric antigen receptors, bispecific antibodies and T-cell engagers, and immune checkpoint inhibitors. Candidate drugs explored in ongoing clinical trials are coupled with diverse toxicity issues and refractoriness to drugs. According to the literature on DLBCL, the promise for new therapeutic targets lies in epigenetic alterations, B-cell receptor and NF-κB pathways. Herein, we present putative targets hiding in lipid pathways, ferroptosis, and the gut microbiome that could be used in addition to immuno-chemotherapy to improve the general health status of DLBCL patients, thus increasing the chance of being cured. It may be time to devote more effort to exploring DLBCL metabolism to discover novel druggable targets. We also performed a bibliometric and knowledge-map analysis of the literature on DLBCL published from 2014-2023.
Collapse
Affiliation(s)
- Romana Masnikosa
- Department of Physical Chemistry, Vinca Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia;
| | - Zorica Cvetković
- Department of Hematology, Clinical Hospital Centre Zemun, Vukova 9, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Dr Subotića 8, 11000 Belgrade, Serbia
| | - David Pirić
- Department of Physical Chemistry, Vinca Institute of Nuclear Sciences—National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia;
| |
Collapse
|
4
|
Schultheiß C, Paschold L, Mohebiany AN, Escher M, Kattimani YM, Müller M, Schmidt-Barbo P, Mensa-Vilaró A, Aróstegui JI, Boursier G, de Moreuil C, Hautala T, Willscher E, Jonas H, Chinchuluun N, Grosser B, Märkl B, Klapper W, Oommen PT, Gössling K, Hoffmann K, Tiegs G, Czernilofsky F, Dietrich S, Freeman A, Schwartz DM, Waisman A, Aksentijevich I, Binder M. A20 haploinsufficiency disturbs immune homeostasis and drives the transformation of lymphocytes with permissive antigen receptors. SCIENCE ADVANCES 2024; 10:eadl3975. [PMID: 39167656 PMCID: PMC11338232 DOI: 10.1126/sciadv.adl3975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 07/15/2024] [Indexed: 08/23/2024]
Abstract
Genetic TNFAIP3 (A20) inactivation is a classical somatic lymphoma lesion and the genomic trait in haploinsufficiency of A20 (HA20). In a cohort of 34 patients with HA20, we show that heterozygous TNFAIP3 loss skews immune repertoires toward lymphocytes with classical self-reactive antigen receptors typically found in B and T cell lymphomas. This skewing was mediated by a feed-forward tumor necrosis factor (TNF)/A20/nuclear factor κB (NF-κB) loop that shaped pre-lymphoma transcriptome signatures in clonally expanded B (CD81, BACH2, and NEAT1) or T (GATA3, TOX, and PDCD1) cells. The skewing was reversed by anti-TNF treatment but could also progress to overt lymphoma. Analysis of conditional TNFAIP3 knock-out mice reproduced the wiring of the TNF/A20/NF-κB signaling axis with permissive antigen receptors and suggested a distinct regulation in B and T cells. Together, patients with the genetic disorder HA20 provide an exceptional window into A20/TNF/NF-κB-mediated control of immune homeostasis and early steps of lymphomagenesis that remain clinically unrecognized.
Collapse
Affiliation(s)
- Christoph Schultheiß
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - Lisa Paschold
- Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Alma Nazlie Mohebiany
- Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Moritz Escher
- Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Yogita Mallu Kattimani
- Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Melanie Müller
- Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Paul Schmidt-Barbo
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
- Collaborative Research Institute Intelligent Oncology (CRIION), Freiburg, Germany
| | - Anna Mensa-Vilaró
- Department of Immunology, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Juan Ignacio Aróstegui
- Department of Immunology, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- School of Medicine, University of Barcelona, Barcelona, Spain
| | - Guilaine Boursier
- Department of molecular and cytogenomics, Rare and Autoinflammatory Diseases Laboratory, CHU Montpellier, IRMB, University of Montpellier, INSERM, CEREMAIA, Montpellier, France
| | - Claire de Moreuil
- Department of Internal Medicine, CHU Brest, Université de Bretagne Occidentale, Brest, France
| | - Timo Hautala
- Research Unit of Biomedicine, University of Oulu and Department of Internal Medicine, Oulu University Hospital, Oulu, Finland
| | - Edith Willscher
- Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Hanna Jonas
- Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Namuun Chinchuluun
- Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Bianca Grosser
- Institute for Pathology, University Medical Center Augsburg, Augsburg, Germany
| | - Bruno Märkl
- Institute for Pathology, University Medical Center Augsburg, Augsburg, Germany
| | - Wolfram Klapper
- Institute of Pathology, Hematopathology Section, and Lymph Node Registry, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Prasad Thomas Oommen
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, Center for Child and Adolescent Health, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Katharina Gössling
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, Center for Child and Adolescent Health, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Department of Pediatrics, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Katrin Hoffmann
- Institute for Human Genetics and Molecular Biology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Gisa Tiegs
- Institute for Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Felix Czernilofsky
- Department of Medicine V, Hematology, Oncology, and Rheumatology, University of Heidelberg, Heidelberg, Germany
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), Heidelberg, Germany
| | - Sascha Dietrich
- Department of Medicine V, Hematology, Oncology, and Rheumatology, University of Heidelberg, Heidelberg, Germany
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), Heidelberg, Germany
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Hematolgy, Oncology, and Immunolgy, University Hospital of Düsseldorf, Düsseldorf, Germany
| | - Alexandra Freeman
- Laboratory of Clinical Immunology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Daniella M. Schwartz
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Ivona Aksentijevich
- Inflammatory Disease Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mascha Binder
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
- Collaborative Research Institute Intelligent Oncology (CRIION), Freiburg, Germany
| |
Collapse
|
5
|
Pham CV, Chowdhury R, Patel S, Melke H, Hou Y, Xu H, Jia L, Duan A, Duan W, Xiang D. The role of the size of affinity ligands in the detection and characterization of extracellular vesicles. Biosens Bioelectron 2024; 258:116381. [PMID: 38744116 DOI: 10.1016/j.bios.2024.116381] [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: 02/08/2024] [Revised: 04/15/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024]
Abstract
Surface proteins on the membrane of nano-sized extracellular vesicles (EVs) not only play crucial roles in cell-to-cell communication, but also are specific binding targets for EV detection, isolation and tracking. The low abundance of protein biomarkers on EV surface, the formation of clusters and the complex EV surface network impose significant challenges to the study of EVs. Employing bulky sized affinity ligands, such as antibodies, in the detection and characterization of these vesicles often result in reduced sensitivity of detection or poor quantification of proteins on the EV surface. By virtue of their small size and high specificity, Affibody molecules emerge as a potential alternative to their monoclonal antibody counterparts as robust affinity ligands in EV research. In this study, we present a theoretical framework on the superiority of anti-HER2 Affibodies over anti-HER2 antibodies in labeling and detecting HER2-positive EVs, followed by the demonstration of the advantages of HER2 Affibodies in accessing EV surface and the detection of EVs through multiple types of approaches including fluorescence intensity, colorimetry, and fluorescence polarization. HER2 Affibodies outperformed by 10-fold over three HER2 antibody clones in accessing HER2-positive EVs derived from different human cancer cell lines. Furthermore, HRP-Affibody molecules could detect EVs from cancer cells spiked into human serum with at least a 2-fold higher sensitivity compared with that of their antibody counterparts. In addition, in fluorescence polarization assays in which no separation of free from bound ligand is required, FITC-labeled HER2 Affibodies could sensitively detect HER2-positive EVs with a clinically relevant limit of detection, whilst HER2 antibodies failed to detect EVs in the same conditions. With the demonstrated superiority in accessing and detecting surface targets over bulky-sized antibodies in EVs, Affibodies may become the next-generation of affinity ligands in the precise characterization and quantification of molecular architecture on the surface of EVs.
Collapse
Affiliation(s)
- Cuong Viet Pham
- School of Medicine, Deakin University, and IMPACT Strategic Research Centre, Waurn Ponds, VIC, 3216, Australia
| | - Rocky Chowdhury
- School of Medicine, Deakin University, and IMPACT Strategic Research Centre, Waurn Ponds, VIC, 3216, Australia
| | - Shweta Patel
- School of Medicine, Deakin University, and IMPACT Strategic Research Centre, Waurn Ponds, VIC, 3216, Australia
| | - Haben Melke
- School of Medicine, Deakin University, and IMPACT Strategic Research Centre, Waurn Ponds, VIC, 3216, Australia
| | - Yingchu Hou
- Laboratory of Tumor Molecular and Cellular Biology College of Life Sciences, Shaanxi Normal University 620 West Chang'an Avenue, Xi'an, Shaanxi, 710119, China
| | - Huo Xu
- College of Materials and Chemical Engineering, Minjiang University, Fuzhou, Fujian, 350108, China
| | - Lee Jia
- College of Materials and Chemical Engineering, Minjiang University, Fuzhou, Fujian, 350108, China
| | - Andrew Duan
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Vic, Australia
| | - Wei Duan
- School of Medicine, Deakin University, and IMPACT Strategic Research Centre, Waurn Ponds, VIC, 3216, Australia.
| | - Dongxi Xiang
- State Key Laboratory of Oncogenes and Related Genes, Department of Biliary-Pancreatic Surgery, The Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
6
|
DʼEste E, Lukinavičius G, Lincoln R, Opazo F, Fornasiero EF. Advancing cell biology with nanoscale fluorescence imaging: essential practical considerations. Trends Cell Biol 2024; 34:671-684. [PMID: 38184400 DOI: 10.1016/j.tcb.2023.12.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/06/2023] [Accepted: 12/11/2023] [Indexed: 01/08/2024]
Abstract
Recently, biologists have gained access to several far-field fluorescence nanoscopy (FN) technologies that allow the observation of cellular components with ~20 nm resolution. FN is revolutionizing cell biology by enabling the visualization of previously inaccessible subcellular details. While technological advances in microscopy are critical to the field, optimal sample preparation and labeling are equally important and often overlooked in FN experiments. In this review, we provide an overview of the methodological and experimental factors that must be considered when performing FN. We present key concepts related to the selection of affinity-based labels, dyes, multiplexing, live cell imaging approaches, and quantitative microscopy. Consideration of these factors greatly enhances the effectiveness of FN, making it an exquisite tool for numerous biological applications.
Collapse
Affiliation(s)
- Elisa DʼEste
- Optical Microscopy Facility, Max Planck Institute for Medical Research, Heidelberg 69120, Germany.
| | - Gražvydas Lukinavičius
- Chromatin Labelling and Imaging Group, Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany.
| | - Richard Lincoln
- Department of Optical Nanoscopy, Max Planck Institute for Medical Research, Heidelberg 69120, Germany.
| | - Felipe Opazo
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen (UMG), Göttingen 37073, Germany; Center for Biostructural Imaging of Neurodegeneration (BIN), University Medical Center, Göttingen 37075, Germany; NanoTag Biotechnologies GmbH, Göttingen 37079, Germany.
| | - Eugenio F Fornasiero
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen (UMG), Göttingen 37073, Germany; Department of Life Sciences, University of Trieste, Trieste 34127, Italy.
| |
Collapse
|
7
|
Schmidt-Barbo P, Kalweit G, Naouar M, Paschold L, Willscher E, Schultheiß C, Märkl B, Dirnhofer S, Tzankov A, Binder M, Kalweit M. Detection of disease-specific signatures in B cell repertoires of lymphomas using machine learning. PLoS Comput Biol 2024; 20:e1011570. [PMID: 38954728 PMCID: PMC11249212 DOI: 10.1371/journal.pcbi.1011570] [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: 10/05/2023] [Revised: 07/15/2024] [Accepted: 06/07/2024] [Indexed: 07/04/2024] Open
Abstract
The classification of B cell lymphomas-mainly based on light microscopy evaluation by a pathologist-requires many years of training. Since the B cell receptor (BCR) of the lymphoma clonotype and the microenvironmental immune architecture are important features discriminating different lymphoma subsets, we asked whether BCR repertoire next-generation sequencing (NGS) of lymphoma-infiltrated tissues in conjunction with machine learning algorithms could have diagnostic utility in the subclassification of these cancers. We trained a random forest and a linear classifier via logistic regression based on patterns of clonal distribution, VDJ gene usage and physico-chemical properties of the top-n most frequently represented clonotypes in the BCR repertoires of 620 paradigmatic lymphoma samples-nodular lymphocyte predominant B cell lymphoma (NLPBL), diffuse large B cell lymphoma (DLBCL) and chronic lymphocytic leukemia (CLL)-alongside with 291 control samples. With regard to DLBCL and CLL, the models demonstrated optimal performance when utilizing only the most prevalent clonotype for classification, while in NLPBL-that has a dominant background of non-malignant bystander cells-a broader array of clonotypes enhanced model accuracy. Surprisingly, the straightforward logistic regression model performed best in this seemingly complex classification problem, suggesting linear separability in our chosen dimensions. It achieved a weighted F1-score of 0.84 on a test cohort including 125 samples from all three lymphoma entities and 58 samples from healthy individuals. Together, we provide proof-of-concept that at least the 3 studied lymphoma entities can be differentiated from each other using BCR repertoire NGS on lymphoma-infiltrated tissues by a trained machine learning model.
Collapse
MESH Headings
- Humans
- Machine Learning
- Receptors, Antigen, B-Cell/genetics
- High-Throughput Nucleotide Sequencing/methods
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Computational Biology/methods
- Lymphoma, B-Cell/genetics
- B-Lymphocytes/metabolism
- B-Lymphocytes/immunology
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/classification
- Algorithms
Collapse
Affiliation(s)
- Paul Schmidt-Barbo
- Department of Biomedicine, Translational Immuno-Oncology, University Hospital Basel, Basel, Switzerland
- Collaborative Research Institute Intelligent Oncology (CRIION), Freiburg, Germany
| | - Gabriel Kalweit
- Collaborative Research Institute Intelligent Oncology (CRIION), Freiburg, Germany
- Neurorobotics Lab, University of Freiburg, Freiburg, Germany
| | - Mehdi Naouar
- Collaborative Research Institute Intelligent Oncology (CRIION), Freiburg, Germany
- Neurorobotics Lab, University of Freiburg, Freiburg, Germany
| | - Lisa Paschold
- Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Edith Willscher
- Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Christoph Schultheiß
- Department of Biomedicine, Translational Immuno-Oncology, University Hospital Basel, Basel, Switzerland
| | - Bruno Märkl
- Pathology, University Hospital Augsburg, Augsburg, Germany
| | | | | | - Mascha Binder
- Department of Biomedicine, Translational Immuno-Oncology, University Hospital Basel, Basel, Switzerland
- Collaborative Research Institute Intelligent Oncology (CRIION), Freiburg, Germany
- Medical Oncology, University Hospital Basel, Basel, Switzerland
| | - Maria Kalweit
- Collaborative Research Institute Intelligent Oncology (CRIION), Freiburg, Germany
- Neurorobotics Lab, University of Freiburg, Freiburg, Germany
| |
Collapse
|
8
|
Chen Y, Lin M, Ye D, Wang S, Zuo X, Li M. Functionalized tetrahedral DNA frameworks for the capture of circulating tumor cells. Nat Protoc 2024; 19:985-1014. [PMID: 38316964 DOI: 10.1038/s41596-023-00943-3] [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: 05/10/2023] [Accepted: 10/30/2023] [Indexed: 02/07/2024]
Abstract
Identification and characterization of circulating tumor cells (CTCs) from blood samples of patients with cancer can help monitor parameters such as disease stage, disease progression and therapeutic efficiency. However, the sensitivity and specificity of current multivalent approaches used for CTC capture is limited by the lack of control over the ligands' position. In this Protocol Update, we describe DNA-tetrahedral frameworks anchored with aptamers that can be configured with user-defined spatial arrangements and stoichiometries. The modified tetrahedral DNA frameworks, termed 'n-simplexes', can be used as probes to specifically target receptor-ligand interactions on the cell membrane. Here, we describe the synthesis and use of n-simplexes that target the epithelial cell adhesion molecule expressed on the surface of CTCs. The characterization of the n-simplexes includes measuring the binding affinity to the membrane receptors as a result of the spatial arrangement and stoichiometry of the aptamers. We further detail the capture of CTCs from patient blood samples. The procedure for the preparation and characterization of n-simplexes requires 11.5 h, CTC capture from clinical samples and data processing requires ~5 h per six samples and the downstream analysis of captured cells typically requires 5.5 h. The protocol is suitable for users with basic expertise in molecular biology and handling of clinical samples.
Collapse
Affiliation(s)
- Yirong Chen
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Zhangjiang Institute for Advanced Study, School of Chemistry and Chemical Engineering, and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Meihua Lin
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, China
| | - Dekai Ye
- Zhangjiang Laboratory, Shanghai, China
| | - Shaopeng Wang
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Zhangjiang Institute for Advanced Study, School of Chemistry and Chemical Engineering, and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaolei Zuo
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Zhangjiang Institute for Advanced Study, School of Chemistry and Chemical Engineering, and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Min Li
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Zhangjiang Institute for Advanced Study, School of Chemistry and Chemical Engineering, and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
9
|
Chen J, Sathiaseelan V, Reddy Chilamakuri CS, Roamio Franklin VN, Jakwerth CA, D’Santos C, Ringshausen I. ZAP-70 augments tonic B-cell receptor and CCR7 signaling in IGHV-unmutated chronic lymphocytic leukemia. Blood Adv 2024; 8:1167-1178. [PMID: 38113463 PMCID: PMC10910066 DOI: 10.1182/bloodadvances.2022009557] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 12/08/2023] [Accepted: 12/11/2023] [Indexed: 12/21/2023] Open
Abstract
ABSTRACT Expression of ZAP-70 in a subset of patients with chronic lymphocytic leukemia (CLL) positively correlates with the absence of immunoglobulin heavy-chain gene (IGHV) mutations and is indicative of a more active disease and shorter treatment-free survival. We recently demonstrated that ZAP-70 regulates the constitutive expression of CCL3 and CCL4, activation of AKT, and expression of MYC in the absence of an overt B-cell receptor (BCR) signal, bona fide functions of BCR activation. We, here, provide evidence that these features relate to the presence of a constitutive tonic BCR signal, exclusively found in IGHV-unmutated CLL and dependent on the ZAP-70-mediated activation of AKT and its downstream target GSK-3β. These findings are associated with increased steady-state activation of CD19 and SRC. Notably this tonic BCR signal is not present in IGHV-mutated CLL cells, discordantly expressing ZAP-70. Results of quantitative mass spectrometry and phosphoprotein analyses indicate that this ZAP-70-dependent, tonic BCR signal regulates CLL cell migration through phosphorylation of LCP1 on serine-5. Indeed, we show that CCL19- and CCL21-induced chemotaxis is regulated by and dependent on the expression of ZAP-70 through its function to enhance CCR7 signaling to LCP1. Thus, our data demonstrate that ZAP-70 converges a tonic BCR signal, exclusively present in IGHV-unmutated CLL and CCR7-mediated chemotaxis.
Collapse
Affiliation(s)
- Jingyu Chen
- Department of Biochemistry and Molecular Biology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, People’s Republic of China
- Wellcome/MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Vijitha Sathiaseelan
- Wellcome/MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | | | | | - Constanze A. Jakwerth
- Center of Allergy & Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German Research Center for Environmental Health & German Center for Lung Research (DZL), Munich, Germany
| | - Clive D’Santos
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Ingo Ringshausen
- Wellcome/MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
- University College London, Cancer Institute, London, United Kingdom
| |
Collapse
|
10
|
Daly S, Ferreira Fernandes J, Bruggeman E, Handa A, Peters R, Benaissa S, Zhang B, Beckwith JS, Sanders EW, Sims RR, Klenerman D, Davis SJ, O'Holleran K, Lee SF. High-density volumetric super-resolution microscopy. Nat Commun 2024; 15:1940. [PMID: 38431671 PMCID: PMC10908787 DOI: 10.1038/s41467-024-45828-5] [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: 05/01/2023] [Accepted: 02/01/2024] [Indexed: 03/05/2024] Open
Abstract
Volumetric super-resolution microscopy typically encodes the 3D position of single-molecule fluorescence into a 2D image by changing the shape of the point spread function (PSF) as a function of depth. However, the resulting large and complex PSF spatial footprints reduce biological throughput and applicability by requiring lower labeling densities to avoid overlapping fluorescent signals. We quantitatively compare the density dependence of single-molecule light field microscopy (SMLFM) to other 3D PSFs (astigmatism, double helix and tetrapod) showing that SMLFM enables an order-of-magnitude speed improvement compared to the double helix PSF by resolving overlapping emitters through parallax. We demonstrate this optical robustness experimentally with high accuracy ( > 99.2 ± 0.1%, 0.1 locs μm-2) and sensitivity ( > 86.6 ± 0.9%, 0.1 locs μm-2) through whole-cell (scan-free) imaging and tracking of single membrane proteins in live primary B cells. We also exemplify high-density volumetric imaging (0.15 locs μm-2) in dense cytosolic tubulin datasets.
Collapse
Affiliation(s)
- Sam Daly
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - João Ferreira Fernandes
- Radcliffe Department of Medicine and MRC Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Ezra Bruggeman
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Anoushka Handa
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Ruby Peters
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, CB2 3EL, UK
| | - Sarah Benaissa
- Cambridge Advanced Imaging Centre, Downing Site, University of Cambridge, Cambridge, CB2 3DY, UK
| | - Boya Zhang
- Cambridge Advanced Imaging Centre, Downing Site, University of Cambridge, Cambridge, CB2 3DY, UK
| | - Joseph S Beckwith
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Edward W Sanders
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Ruth R Sims
- Wavefront-Engineering Microscopy Group, Photonics Department, Institut de la Vision, Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - David Klenerman
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Simon J Davis
- Radcliffe Department of Medicine and MRC Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Kevin O'Holleran
- Cambridge Advanced Imaging Centre, Downing Site, University of Cambridge, Cambridge, CB2 3DY, UK
| | - Steven F Lee
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.
| |
Collapse
|
11
|
Brenner B, Xu F, Zhang Y, Kweon J, Fang R, Sheibani N, Zhang SX, Sun C, Zhang HF. Quantifying nanoscopic alterations associated with mitochondrial dysfunction using three-dimensional single-molecule localization microscopy. BIOMEDICAL OPTICS EXPRESS 2024; 15:1571-1584. [PMID: 38495683 PMCID: PMC10942681 DOI: 10.1364/boe.510351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/12/2024] [Accepted: 01/31/2024] [Indexed: 03/19/2024]
Abstract
Mitochondrial morphology provides unique insights into their integrity and function. Among fluorescence microscopy techniques, 3D super-resolution microscopy uniquely enables the analysis of mitochondrial morphological features individually. However, there is a lack of tools to extract morphological parameters from super-resolution images of mitochondria. We report a quantitative method to extract mitochondrial morphological metrics, including volume, aspect ratio, and local protein density, from 3D single-molecule localization microscopy images, with single-mitochondrion sensitivity. We validated our approach using simulated ground-truth SMLM images of mitochondria. We further tested our morphological analysis on mitochondria that have been altered functionally and morphologically in controlled manners. This work sets the stage to quantitatively analyze mitochondrial morphological alterations associated with disease progression on an individual basis.
Collapse
Affiliation(s)
- Benjamin Brenner
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | - Fengyuanshan Xu
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | - Yang Zhang
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Currently with Program of Polymer and Color Chemistry, Department of Textile Engineering, Chemistry and Science, North Carolina State University, Raleigh, NC, USA
| | - Junghun Kweon
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | - Raymond Fang
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | - Nader Sheibani
- Department of Ophthalmology and Vision Sciences, University of Wisconsin, Madison, WI, USA
| | - Sarah X. Zhang
- Department of Ophthalmology, University at Buffalo, Buffalo, NY, USA
| | - Cheng Sun
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA
| | - Hao F. Zhang
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| |
Collapse
|
12
|
Alsouri S, Ambrose A, Mougios N, Paglilla N, Mayr F, Choi K, Loeber J, Chapuy B, Haeupl B, Opazo F, Oellerich T, Gold M, Engelke M. Actinin-4 controls survival signaling in B cells by limiting the lateral mobility of B-cell antigen receptors. Eur J Immunol 2024; 54:e2350774. [PMID: 38299456 DOI: 10.1002/eji.202350774] [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: 09/14/2023] [Revised: 12/22/2023] [Accepted: 01/04/2024] [Indexed: 02/02/2024]
Abstract
The structure and dynamics of F-actin networks in the cortical area of B cells control the signal efficiency of B-cell antigen receptors (BCRs). Although antigen-induced signaling has been studied extensively, the role of cortical F-actin in antigen-independent tonic BCR signaling is less well understood. Because these signals are essential for the survival of B cells and are consequently exploited by several B-cell lymphomas, we assessed how the cortical F-actin structure influences tonic BCR signal transduction. We employed genetic variants of a primary cell-like B-cell line that can be rendered quiescent to show that cross-linking of actin filaments by α-actinin-4 (ACTN4), but not ACTN1, is required to preserve the dense architecture of F-actin in the cortical area of B cells. The reduced cortical F-actin density in the absence of ACTN4 resulted in increased lateral BCR diffusion. Surprisingly, this was associated with reduced tonic activation of BCR-proximal effector proteins, extracellular signal-regulated kinase, and pro-survival pathways. Accordingly, ACTN4-deficient B-cell lines and primary human B cells exhibit augmented apoptosis. Hence, our findings reveal that cortical F-actin architecture regulates antigen-independent tonic BCR survival signals in human B cells.
Collapse
Affiliation(s)
- Saed Alsouri
- Institute for Cellular and Molecular Immunology, University Medical Center Goettingen, Goettingen, Germany
| | - Ashley Ambrose
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
- Department of Mathematics, University of British Columbia, Vancouver, Canada
| | - Nikolaos Mougios
- Center for Biostructural Imaging of Neurodegeneration (BIN), Goettingen, Germany
- Institute of Neuro- and Sensory Physiology, University Medical Center Goettingen, Goettingen, Germany
| | - Nadia Paglilla
- Institute for Cellular and Molecular Immunology, University Medical Center Goettingen, Goettingen, Germany
| | - Florian Mayr
- Institute for Cellular and Molecular Immunology, University Medical Center Goettingen, Goettingen, Germany
| | - Kate Choi
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
| | - Jens Loeber
- Department of Hematology, Oncology and Cancer Immunology, Charité - University Medical Center Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Björn Chapuy
- Department of Hematology, Oncology and Cancer Immunology, Charité - University Medical Center Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Björn Haeupl
- Frankfurt Cancer Institute, Johann Wolfgang Goethe University Frankfurt, Frankfurt, Germany
- German Cancer Consortium (DKTK), Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Felipe Opazo
- Center for Biostructural Imaging of Neurodegeneration (BIN), Goettingen, Germany
- Institute of Neuro- and Sensory Physiology, University Medical Center Goettingen, Goettingen, Germany
| | - Thomas Oellerich
- Frankfurt Cancer Institute, Johann Wolfgang Goethe University Frankfurt, Frankfurt, Germany
- German Cancer Consortium (DKTK), Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Gold
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
| | - Michael Engelke
- Institute for Cellular and Molecular Immunology, University Medical Center Goettingen, Goettingen, Germany
| |
Collapse
|
13
|
Märkl F, Schultheiß C, Ali M, Chen SS, Zintchenko M, Egli L, Mietz J, Chijioke O, Paschold L, Spajic S, Holtermann A, Dörr J, Stock S, Zingg A, Läubli H, Piseddu I, Anz D, Minden MDV, Zhang T, Nerreter T, Hudecek M, Minguet S, Chiorazzi N, Kobold S, Binder M. Mutation-specific CAR T cells as precision therapy for IGLV3-21 R110 expressing high-risk chronic lymphocytic leukemia. Nat Commun 2024; 15:993. [PMID: 38307904 PMCID: PMC10837166 DOI: 10.1038/s41467-024-45378-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 01/22/2024] [Indexed: 02/04/2024] Open
Abstract
The concept of precision cell therapy targeting tumor-specific mutations is appealing but requires surface-exposed neoepitopes, which is a rarity in cancer. B cell receptors (BCR) of mature lymphoid malignancies are exceptional in that they harbor tumor-specific-stereotyped sequences in the form of point mutations that drive self-engagement of the BCR and autologous signaling. Here, we use a BCR light chain neoepitope defined by a characteristic point mutation (IGLV3-21R110) for selective targeting of a poor-risk subset of chronic lymphocytic leukemia (CLL) with chimeric antigen receptor (CAR) T cells. We develop murine and humanized CAR constructs expressed in T cells from healthy donors and CLL patients that eradicate IGLV3-21R110 expressing cell lines and primary CLL cells, but neither cells expressing the non-pathogenic IGLV3-21G110 light chain nor polyclonal healthy B cells. In vivo experiments confirm epitope-selective cytolysis in xenograft models in female mice using engrafted IGLV3-21R110 expressing cell lines or primary CLL cells. We further demonstrate in two humanized mouse models lack of cytotoxicity towards human B cells. These data provide the basis for advanced approaches of resistance-preventive and biomarker-guided cellular targeting of functionally relevant lymphoma driver mutations sparing normal B cells.
Collapse
Affiliation(s)
- Florian Märkl
- Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany
| | - Christoph Schultheiß
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - Murtaza Ali
- Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Shih-Shih Chen
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | | | - Lukas Egli
- Cellular Immunotherapy, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Juliane Mietz
- Cellular Immunotherapy, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Obinna Chijioke
- Cellular Immunotherapy, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Institute of Pathology and Medical Genetics, University Hospital Basel, Basel, Switzerland
| | - Lisa Paschold
- Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Sebastijan Spajic
- Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany
| | - Anne Holtermann
- Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany
| | - Janina Dörr
- Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany
| | - Sophia Stock
- Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany
| | - Andreas Zingg
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Laboratory of Cancer Immunotherapy, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - Heinz Läubli
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Laboratory of Cancer Immunotherapy, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - Ignazio Piseddu
- Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany
| | - David Anz
- Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany
| | | | - Tianjiao Zhang
- Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Thomas Nerreter
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Michael Hudecek
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Susana Minguet
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical Faculty, Freiburg, Germany
| | - Nicholas Chiorazzi
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Sebastian Kobold
- Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Munich, Research Center for Environmental Health (HMGU), Neuherberg, Germany.
| | - Mascha Binder
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland.
- Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland.
| |
Collapse
|
14
|
Jeong S, Koh D, Gwak E, Srambickal CV, Seo D, Widengren J, Lee JC. Pushing the Resolution Limit of Stimulated Emission Depletion Optical Nanoscopy. Int J Mol Sci 2023; 25:26. [PMID: 38203197 PMCID: PMC10779414 DOI: 10.3390/ijms25010026] [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: 09/15/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 01/12/2024] Open
Abstract
Optical nanoscopy, also known as super-resolution optical microscopy, has provided scientists with the means to surpass the diffraction limit of light microscopy and attain new insights into nanoscopic structures and processes that were previously inaccessible. In recent decades, numerous studies have endeavored to enhance super-resolution microscopy in terms of its spatial (lateral) resolution, axial resolution, and temporal resolution. In this review, we discuss recent efforts to push the resolution limit of stimulated emission depletion (STED) optical nanoscopy across multiple dimensions, including lateral resolution, axial resolution, temporal resolution, and labeling precision. We introduce promising techniques and methodologies building on the STED concept that have emerged in the field, such as MINSTED, isotropic STED, and event-triggered STED, and evaluate their respective strengths and limitations. Moreover, we discuss trade-off relationships that exist in far-field optical microscopy and how they come about in STED optical nanoscopy. By examining the latest developments addressing these aspects, we aim to provide an updated overview of the current state of STED nanoscopy and its potential for future research.
Collapse
Affiliation(s)
- Sejoo Jeong
- Department of New Biology, DGIST, Daegu 42988, Republic of Korea
| | - Dongbin Koh
- School of Undergraduate Studies, DGIST, Daegu 42988, Republic of Korea
| | - Eunha Gwak
- Department of New Biology, DGIST, Daegu 42988, Republic of Korea
| | - Chinmaya V. Srambickal
- Exp. Biomol. Physics, Dept. Applied Physics, KTH—Royal Institute of Technology, 106 91 Stockholm, Sweden
| | - Daeha Seo
- Department of Physics and Chemistry, DGIST, Daegu 42988, Republic of Korea
| | - Jerker Widengren
- Exp. Biomol. Physics, Dept. Applied Physics, KTH—Royal Institute of Technology, 106 91 Stockholm, Sweden
| | - Jong-Chan Lee
- Department of New Biology, DGIST, Daegu 42988, Republic of Korea
- New Biology Research Center, DGIST, Daegu 42988, Republic of Korea
| |
Collapse
|
15
|
Queiroz Zetune Villa Real K, Mougios N, Rehm R, Sograte-Idrissi S, Albert L, Rahimi AM, Maidorn M, Hentze J, Martínez-Carranza M, Hosseini H, Saal KA, Oleksiievets N, Prigge M, Tsukanov R, Stenmark P, Fornasiero EF, Opazo F. A Versatile Synaptotagmin-1 Nanobody Provides Perturbation-Free Live Synaptic Imaging And Low Linkage-Error in Super-Resolution Microscopy. SMALL METHODS 2023; 7:e2300218. [PMID: 37421204 DOI: 10.1002/smtd.202300218] [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: 02/21/2023] [Revised: 06/09/2023] [Indexed: 07/10/2023]
Abstract
Imaging of living synapses has relied for over two decades on the overexpression of synaptic proteins fused to fluorescent reporters. This strategy alters the stoichiometry of synaptic components and ultimately affects synapse physiology. To overcome these limitations, here a nanobody is presented that binds the calcium sensor synaptotagmin-1 (NbSyt1). This nanobody functions as an intrabody (iNbSyt1) in living neurons and is minimally invasive, leaving synaptic transmission almost unaffected, as suggested by the crystal structure of the NbSyt1 bound to Synaptotagmin-1 and by the physiological data. Its single-domain nature enables the generation of protein-based fluorescent reporters, as showcased here by measuring spatially localized presynaptic Ca2+ with a NbSyt1- jGCaMP8 chimera. Moreover, the small size of NbSyt1 makes it ideal for various super-resolution imaging methods. Overall, NbSyt1 is a versatile binder that will enable imaging in cellular and molecular neuroscience with unprecedented precision across multiple spatiotemporal scales.
Collapse
Affiliation(s)
- Karine Queiroz Zetune Villa Real
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073, Göttingen, Germany
- Center for Biostructural Imaging of Neurodegeneration (BIN), University Medical Center Göttingen, 37075, Göttingen, Germany
| | - Nikolaos Mougios
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073, Göttingen, Germany
- Center for Biostructural Imaging of Neurodegeneration (BIN), University Medical Center Göttingen, 37075, Göttingen, Germany
| | - Ronja Rehm
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073, Göttingen, Germany
| | - Shama Sograte-Idrissi
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073, Göttingen, Germany
- Center for Biostructural Imaging of Neurodegeneration (BIN), University Medical Center Göttingen, 37075, Göttingen, Germany
| | - László Albert
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073, Göttingen, Germany
- Center for Biostructural Imaging of Neurodegeneration (BIN), University Medical Center Göttingen, 37075, Göttingen, Germany
| | - Amir Mohammad Rahimi
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073, Göttingen, Germany
- Center for Biostructural Imaging of Neurodegeneration (BIN), University Medical Center Göttingen, 37075, Göttingen, Germany
| | - Manuel Maidorn
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073, Göttingen, Germany
- Center for Biostructural Imaging of Neurodegeneration (BIN), University Medical Center Göttingen, 37075, Göttingen, Germany
| | - Jannik Hentze
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073, Göttingen, Germany
- Center for Biostructural Imaging of Neurodegeneration (BIN), University Medical Center Göttingen, 37075, Göttingen, Germany
| | - Markel Martínez-Carranza
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Stockholm, SE-10691, Sweden
| | - Hassan Hosseini
- Research Group Neuromodulatory Networks, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany
| | - Kim-Ann Saal
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073, Göttingen, Germany
| | - Nazar Oleksiievets
- III. Institute of Physics - Biophysics, Georg August University, 37077, Göttingen, Germany
| | - Matthias Prigge
- Research Group Neuromodulatory Networks, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany
- Center for Behavioral Brain Sciences, 39118, Magdeburg, Germany
| | - Roman Tsukanov
- III. Institute of Physics - Biophysics, Georg August University, 37077, Göttingen, Germany
| | - Pål Stenmark
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Stockholm, SE-10691, Sweden
| | - Eugenio F Fornasiero
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073, Göttingen, Germany
| | - Felipe Opazo
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073, Göttingen, Germany
- Center for Biostructural Imaging of Neurodegeneration (BIN), University Medical Center Göttingen, 37075, Göttingen, Germany
- NanoTag Biotechnologies GmbH, 37079, Göttingen, Germany
| |
Collapse
|
16
|
Qiao Y, Hu H, Zhao Y, Jin M, Yang D, Yin J, Wu P, Liu W, Li J. Benzene induces spleen injury through the B cell receptor signaling pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 257:114924. [PMID: 37080132 DOI: 10.1016/j.ecoenv.2023.114924] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 03/13/2023] [Accepted: 04/16/2023] [Indexed: 05/03/2023]
Abstract
Benzene is a toxic environmental pollutant that disrupts the immune system in humans. Benzene exposure reduces the abundance of immune cells in multiple immune organs; however, the biological mechanisms underlying benzene-induced immunotoxicity has not been elucidated. In this study, benzene was used to develop mouse model for immune dysfunction. A significant decrease in IgG, IL-2 and IL-6 levels, an increase in oxidative stress and spleen injury were observed after benzene exposure in a dose-dependent manner. Quantitative proteomics revealed that benzene-induced immune dysfunction was associated with deregulation of the B cell receptor (BCR) signaling pathway. Benzene exposure suppressed the expression of CD22, BCL10 and NF-κb p65. Also, a significant decrease in proliferation and an increase in apoptosis of splenic lymphocytes were found after benzene exposure. Moreover, we found that benzene exposure increased mitochondrial reactive oxygen species (mito-ROS) and decreased adenosine triphosphate (ATP). Overall, we revealed the damaging effects of benzene on spleen-related immune function and the underlying biological mechanism, involving the disruption of BCR signaling pathway, NF-κB deactivation, and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Yamei Qiao
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Hui Hu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China; Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, China
| | - Yunyan Zhao
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Min Jin
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Dong Yang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Jing Yin
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Peng Wu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China.
| | - Weili Liu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China.
| | - Junwen Li
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China.
| |
Collapse
|
17
|
Kubagawa H, Clark C, Skopnik CM, Mahmoudi Aliabadi P, Al-Qaisi K, Teuber R, Jani PK, Radbruch A, Melchers F, Engels N, Wienands J. Physiological and Pathophysiological Roles of IgM Fc Receptor (FcµR) Isoforms. Int J Mol Sci 2023; 24:ijms24065728. [PMID: 36982860 PMCID: PMC10058298 DOI: 10.3390/ijms24065728] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/14/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
IgM is the first antibody to emerge during phylogeny, ontogeny, and immune responses and serves as a first line of defense. Effector proteins interacting with the Fc portion of IgM, such as complement and its receptors, have been extensively studied for their functions. IgM Fc receptor (FcµR), identified in 2009, is the newest member of the FcR family and is intriguingly expressed by lymphocytes only, suggesting the existence of distinct functions as compared to the FcRs for switched Ig isotypes, which are expressed by various immune and non-hematopoietic cells as central mediators of antibody-triggered responses by coupling the adaptive and innate immune responses. Results from FcµR-deficient mice suggest a regulatory function of FcµR in B cell tolerance, as evidenced by their propensity to produce autoantibodies of both IgM and IgG isotypes. In this article, we discuss conflicting views about the cellular distribution and potential functions of FcµR. The signaling function of the Ig-tail tyrosine-like motif in the FcµR cytoplasmic domain is now formally shown by substitutional experiments with the IgG2 B cell receptor. The potential adaptor protein associating with FcµR and the potential cleavage of its C-terminal cytoplasmic tail after IgM binding are still enigmatic. Critical amino acid residues in the Ig-like domain of FcµR for interacting with the IgM Cµ4 domain and the mode of interaction are now defined by crystallographic and cryo-electron microscopic analyses. Some discrepancies on these interactions are discussed. Finally, elevated levels of a soluble FcµR isoform in serum samples are described as the consequence of persistent B cell receptor stimulation, as seen in chronic lymphocytic leukemia and probably in antibody-mediated autoimmune disorders.
Collapse
Affiliation(s)
| | - Caren Clark
- Institute of Cellular & Molecular Immunology, University Medical Center, 37073 Göttingen, Germany
| | | | | | | | - Ruth Teuber
- Deutsches Rheuma-Forschungszentrum, 10117 Berlin, Germany
| | - Peter K Jani
- Deutsches Rheuma-Forschungszentrum, 10117 Berlin, Germany
| | | | - Fritz Melchers
- Deutsches Rheuma-Forschungszentrum, 10117 Berlin, Germany
| | - Niklas Engels
- Institute of Cellular & Molecular Immunology, University Medical Center, 37073 Göttingen, Germany
| | - Jürgen Wienands
- Institute of Cellular & Molecular Immunology, University Medical Center, 37073 Göttingen, Germany
| |
Collapse
|
18
|
Ferapontov A, Omer M, Baudrexel I, Nielsen JS, Dupont DM, Juul-Madsen K, Steen P, Eklund AS, Thiel S, Vorup-Jensen T, Jungmann R, Kjems J, Degn SE. Antigen footprint governs activation of the B cell receptor. Nat Commun 2023; 14:976. [PMID: 36813795 PMCID: PMC9947222 DOI: 10.1038/s41467-023-36672-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 02/10/2023] [Indexed: 02/24/2023] Open
Abstract
Antigen binding by B cell receptors (BCR) on cognate B cells elicits a response that eventually leads to production of antibodies. However, it is unclear what the distribution of BCRs is on the naïve B cell and how antigen binding triggers the first step in BCR signaling. Using DNA-PAINT super-resolution microscopy, we find that most BCRs are present as monomers, dimers, or loosely associated clusters on resting B cells, with a nearest-neighbor inter-Fab distance of 20-30 nm. We leverage a Holliday junction nanoscaffold to engineer monodisperse model antigens with precision-controlled affinity and valency, and find that the antigen exerts agonistic effects on the BCR as a function of increasing affinity and avidity. Monovalent macromolecular antigens can activate the BCR at high concentrations, whereas micromolecular antigens cannot, demonstrating that antigen binding does not directly drive activation. Based on this, we propose a BCR activation model determined by the antigen footprint.
Collapse
Affiliation(s)
- Alexey Ferapontov
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark.,Center for Cellular Signal Patterns (CellPAT), Aarhus University, Aarhus C, Denmark
| | - Marjan Omer
- Center for Cellular Signal Patterns (CellPAT), Aarhus University, Aarhus C, Denmark.,Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, Denmark
| | - Isabelle Baudrexel
- Center for Cellular Signal Patterns (CellPAT), Aarhus University, Aarhus C, Denmark.,Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Jesper Sejrup Nielsen
- Center for Cellular Signal Patterns (CellPAT), Aarhus University, Aarhus C, Denmark.,Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, Denmark
| | - Daniel Miotto Dupont
- Center for Cellular Signal Patterns (CellPAT), Aarhus University, Aarhus C, Denmark.,Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, Denmark
| | | | - Philipp Steen
- Max Planck Institute of Biochemistry, Martinsried, Germany.,Faculty of Physics and Center for Nanoscience, Ludwig Maximilian University, Munich, Munich, Germany
| | - Alexandra S Eklund
- Center for Cellular Signal Patterns (CellPAT), Aarhus University, Aarhus C, Denmark.,Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Steffen Thiel
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark.,Center for Cellular Signal Patterns (CellPAT), Aarhus University, Aarhus C, Denmark
| | | | - Ralf Jungmann
- Center for Cellular Signal Patterns (CellPAT), Aarhus University, Aarhus C, Denmark.,Max Planck Institute of Biochemistry, Martinsried, Germany.,Faculty of Physics and Center for Nanoscience, Ludwig Maximilian University, Munich, Munich, Germany
| | - Jørgen Kjems
- Center for Cellular Signal Patterns (CellPAT), Aarhus University, Aarhus C, Denmark.,Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, Denmark
| | - Søren Egedal Degn
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark. .,Center for Cellular Signal Patterns (CellPAT), Aarhus University, Aarhus C, Denmark.
| |
Collapse
|
19
|
Old and New Facts and Speculations on the Role of the B Cell Receptor in the Origin of Chronic Lymphocytic Leukemia. Int J Mol Sci 2022; 23:ijms232214249. [PMID: 36430731 PMCID: PMC9693457 DOI: 10.3390/ijms232214249] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 11/19/2022] Open
Abstract
The engagement of the B cell receptor (BcR) on the surface of leukemic cells represents a key event in chronic lymphocytic leukemia (CLL) since it can lead to the maintenance and expansion of the neoplastic clone. This notion was initially suggested by observations of the CLL BcR repertoire and of correlations existing between certain BcR features and the clinical outcomes of single patients. Based on these observations, tyrosine kinase inhibitors (TKIs), which block BcR signaling, have been introduced in therapy with the aim of inhibiting CLL cell clonal expansion and of controlling the disease. Indeed, the impressive results obtained with these compounds provided further proof of the role of BcR in CLL. In this article, the key steps that led to the determination of the role of BcR are reviewed, including the features of the CLL cell repertoire and the fine mechanisms causing BcR engagement and cell signaling. Furthermore, we discuss the biological effects of the engagement, which can lead to cell survival/proliferation or apoptosis depending on certain intrinsic cell characteristics and on signals that the micro-environment can deliver to the leukemic cells. In addition, consideration is given to alternative mechanisms promoting cell proliferation in the absence of BcR signaling, which can explain in part the incomplete effectiveness of TKI therapies. The role of the BcR in determining clonal evolution and disease progression is also described. Finally, we discuss possible models to explain the selection of a special BcR set during leukemogenesis. The BcR may deliver activation signals to the cells, which lead to their uncontrolled growth, with the possible collaboration of other still-undefined events which are capable of deregulating the normal physiological response of B cells to BcR-delivered stimuli.
Collapse
|
20
|
Shorer Arbel Y, Bronstein Y, Dadosh T, Kamdjou T, Tsuriel S, Shapiro M, Katz BZ, Herishanu Y. Spatial organization and early signaling of the B-cell receptor in CLL. Front Immunol 2022; 13:953660. [PMID: 36016925 PMCID: PMC9398492 DOI: 10.3389/fimmu.2022.953660] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/08/2022] [Indexed: 11/28/2022] Open
Abstract
Most chronic lymphocytic leukemia (CLL) clones express B-cell receptors (BcR) of both IgM/IgD isotypes; however, 5%–10% of CLL cases express isotype-switched immunoglobulin G (IgG). The early signaling and spatial patterning of the various BcRs at steady state and after activation are still fully unresolved. Herein, we show higher expression of the BcR signalosome elements and a more robust constitutive cell-intrinsic proximal BcR signaling in CLL with unmutated IGHV expressing IgM isotype (IgM U-CLL), compared with IGHV-mutated CLL (M-CLL) expressing either IgM or IgG isotypes. IgM in U-CLL is frequently located in the membrane plane in polarized patches, occasionally in caps, and sometimes inside the cells. Among M-CLL, IgM is scattered laterally in the membrane plane in a similar pattern as seen in normal B cells, whereas IgG is dispersed around the cell membrane in smaller clusters than in IgM U-CLL. Upon BcR engagement, both IgG and IgM expressing M-CLL showed attenuated signaling and only slight spatial reorganization dynamics of BcR microclusters and internalization, compared with the extensive reorganization and internalization of the BcR in IgM expressing U-CLL. The global gene signature of IgG M-CLL was closely related to that of IgM M-CLL rather than IgM U-CLL. Overall, we report fundamental differences in the basal composition, biochemical status, and spatial organization of the BcR in the three examined immunogenetic CLL subtypes that correlate with their clinical behavior. On the basis of our findings, IgG class-switched M-CLL likely represents the same disease as IgM M-CLL rather than a different biological and/or clinical entity.
Collapse
MESH Headings
- Humans
- Immunoglobulin G
- Immunoglobulin M
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/metabolism
- Signal Transduction
Collapse
Affiliation(s)
| | - Yotam Bronstein
- Department of Hematology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Tali Dadosh
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot, Israel
| | - Talia Kamdjou
- Department of Hematology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Shlomo Tsuriel
- Department of Pathology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Mika Shapiro
- Department of Hematology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Ben-Zion Katz
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
- Department of Hematology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Yair Herishanu
- Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
- Department of Hematology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- *Correspondence: Yair Herishanu,
| |
Collapse
|
21
|
Brameshuber M, Klotzsch E, Ponjavic A, Sezgin E. Understanding immune signaling using advanced imaging techniques. Biochem Soc Trans 2022; 50:853-866. [PMID: 35343569 PMCID: PMC9162467 DOI: 10.1042/bst20210479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 12/13/2022]
Abstract
Advanced imaging is key for visualizing the spatiotemporal regulation of immune signaling which is a complex process involving multiple players tightly regulated in space and time. Imaging techniques vary in their spatial resolution, spanning from nanometers to micrometers, and in their temporal resolution, ranging from microseconds to hours. In this review, we summarize state-of-the-art imaging methodologies and provide recent examples on how they helped to unravel the mysteries of immune signaling. Finally, we discuss the limitations of current technologies and share our insights on how to overcome these limitations to visualize immune signaling with unprecedented fidelity.
Collapse
Affiliation(s)
- Mario Brameshuber
- Institute of Applied Physics – Biophysics, TU Wien, 1040 Vienna, Austria
| | - Enrico Klotzsch
- Humboldt-Universität zu Berlin, Institut für Biophysik, Experimentelle Biophysik Mechanobiologie, Sitz Invalidenstrasse 42, 10115 Berlin, Germany
| | - Aleks Ponjavic
- School of Physics and Astronomy, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, U.K
- School of Food Science and Nutrition, University of Leeds, Woodhouse Lane, Leeds LS2 9JT, U.K
| | - Erdinc Sezgin
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17165 Solna, Sweden
| |
Collapse
|
22
|
Guo L, Zhang Y, Wang Y, Xie M, Dai J, Qu Z, Zhou M, Cao S, Shi J, Wang L, Zuo X, Fan C, Li J. Directing Multivalent Aptamer-Receptor Binding on the Cell Surface with Programmable Atom-Like Nanoparticles. Angew Chem Int Ed Engl 2022; 61:e202117168. [PMID: 35226386 DOI: 10.1002/anie.202117168] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Indexed: 11/08/2022]
Abstract
Multivalent interactions of biomolecules play pivotal roles in physiological and pathological settings. Whereas the directionality of the interactions is crucial, the state-of-the-art synthetic multivalent ligand-receptor systems generally lack programmable approaches for orthogonal directionality. Here, we report the design of programmable atom-like nanoparticles (aptPANs) to direct multivalent aptamer-receptor binding on the cell interface. The positions of the aptamer motifs can be prescribed on tetrahedral DNA frameworks to realize atom-like orthogonal valence and direction, enabling the construction of multivalent molecules with fixed aptamer copy numbers but different directionality. These directional-yet-flexible aptPAN molecules exhibit the adaptability to the receptor distribution on cell surfaces. We demonstrate the high-affinity tumor cell binding with a linear aptPAN oligomer (≈13-fold improved compared to free aptamers), which leads to ≈50 % suppression of cell growth.
Collapse
Affiliation(s)
- Linjie Guo
- Division of Physical Biology Department, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201800, China.,University of Chinese Academy of Sciences, Beijing, 100049, China.,The Interdisciplinary Research Center, Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201210, China
| | - Yueyue Zhang
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yue Wang
- Division of Physical Biology Department, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201800, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mo Xie
- Division of Physical Biology Department, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201800, China
| | - Jiangbing Dai
- Division of Physical Biology Department, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201800, China
| | - Zhibei Qu
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, 200240, Shanghai, China
| | - Mo Zhou
- Division of Physical Biology Department, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201800, China
| | - Shuting Cao
- Division of Physical Biology Department, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201800, China
| | - Jiye Shi
- Division of Physical Biology Department, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201800, China
| | - Lihua Wang
- The Interdisciplinary Research Center, Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201210, China.,Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Xiaolei Zuo
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, 200240, Shanghai, China
| | - Jiang Li
- Division of Physical Biology Department, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201800, China.,The Interdisciplinary Research Center, Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201210, China.,School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, 200240, Shanghai, China
| |
Collapse
|
23
|
Sun Y, Li X, Wang T, Li W. Core Fucosylation Regulates the Function of Pre-BCR, BCR and IgG in Humoral Immunity. Front Immunol 2022; 13:844427. [PMID: 35401499 PMCID: PMC8990897 DOI: 10.3389/fimmu.2022.844427] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 02/25/2022] [Indexed: 11/20/2022] Open
Abstract
Most of the membrane molecules involved in immune response are glycosylated. N-glycans linked to asparagine (Asn) of immune molecules contribute to the protein conformation, surface expression, stability, and antigenicity. Core fucosylation catalyzed by core fucosyltransferase (FUT8) is the most common post-translational modification. Core fucosylation is essential for evoking a proper immune response, which this review aims to communicate. First, FUT8 deficiency suppressed the interaction between μHC and λ5 during pre-BCR assembly is given. Second, we described the effects of core fucosylation in B cell signal transduction via BCR. Third, we investigated the role of core fucosylation in the interaction between helper T (TH) cells and B cells. Finally, we showed the role of FUT8 on the biological function of IgG. In this review, we discussed recent insights into the sites where core fucosylation is critical for humoral immune responses.
Collapse
Affiliation(s)
- Yuhan Sun
- College of Basic Medical Science, Dalian Medical University, Dalian, China
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Sendai, Japan
| | - Xueying Li
- Research Institute for Microbial Diseases and World Premier International Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Tiantong Wang
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Wenzhe Li
- College of Basic Medical Science, Dalian Medical University, Dalian, China
- *Correspondence: Wenzhe Li,
| |
Collapse
|
24
|
Guo L, Zhang Y, Wang Y, Xie M, Dai J, Qu Z, Zhou M, Cao S, Shi J, Wang L, Zuo X, Fan C, Li J. Directing Multivalent Aptamer‐Receptor Binding on the Cell Surface with Programmable Atom‐Like Nanoparticles. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202117168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Linjie Guo
- Division of Physical Biology Department CAS Key Laboratory of Interfacial Physics and Technology Shanghai Institute of Applied Physics Chinese Academy of Sciences Shanghai 201800 China
- University of Chinese Academy of Sciences Beijing 100049 China
- The Interdisciplinary Research Center Shanghai Synchrotron Radiation Facility Zhangjiang Laboratory Shanghai Advanced Research Institute Chinese Academy of Sciences Shanghai 201210 China
| | - Yueyue Zhang
- Institute of Molecular Medicine Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai 200127 China
| | - Yue Wang
- Division of Physical Biology Department CAS Key Laboratory of Interfacial Physics and Technology Shanghai Institute of Applied Physics Chinese Academy of Sciences Shanghai 201800 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Mo Xie
- Division of Physical Biology Department CAS Key Laboratory of Interfacial Physics and Technology Shanghai Institute of Applied Physics Chinese Academy of Sciences Shanghai 201800 China
| | - Jiangbing Dai
- Division of Physical Biology Department CAS Key Laboratory of Interfacial Physics and Technology Shanghai Institute of Applied Physics Chinese Academy of Sciences Shanghai 201800 China
| | - Zhibei Qu
- School of Chemistry and Chemical Engineering Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine Shanghai Jiao Tong University 200240 Shanghai China
| | - Mo Zhou
- Division of Physical Biology Department CAS Key Laboratory of Interfacial Physics and Technology Shanghai Institute of Applied Physics Chinese Academy of Sciences Shanghai 201800 China
| | - Shuting Cao
- Division of Physical Biology Department CAS Key Laboratory of Interfacial Physics and Technology Shanghai Institute of Applied Physics Chinese Academy of Sciences Shanghai 201800 China
| | - Jiye Shi
- Division of Physical Biology Department CAS Key Laboratory of Interfacial Physics and Technology Shanghai Institute of Applied Physics Chinese Academy of Sciences Shanghai 201800 China
| | - Lihua Wang
- The Interdisciplinary Research Center Shanghai Synchrotron Radiation Facility Zhangjiang Laboratory Shanghai Advanced Research Institute Chinese Academy of Sciences Shanghai 201210 China
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes School of Chemistry and Molecular Engineering East China Normal University Shanghai 200241 China
| | - Xiaolei Zuo
- Institute of Molecular Medicine Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai 200127 China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine Shanghai Jiao Tong University 200240 Shanghai China
| | - Jiang Li
- Division of Physical Biology Department CAS Key Laboratory of Interfacial Physics and Technology Shanghai Institute of Applied Physics Chinese Academy of Sciences Shanghai 201800 China
- The Interdisciplinary Research Center Shanghai Synchrotron Radiation Facility Zhangjiang Laboratory Shanghai Advanced Research Institute Chinese Academy of Sciences Shanghai 201210 China
- School of Chemistry and Chemical Engineering Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine Shanghai Jiao Tong University 200240 Shanghai China
| |
Collapse
|
25
|
Approach to map nanotopography of cell surface receptors. Commun Biol 2022; 5:218. [PMID: 35264712 PMCID: PMC8907216 DOI: 10.1038/s42003-022-03152-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 02/09/2022] [Indexed: 12/18/2022] Open
Abstract
Cells communicate with their environment via surface receptors, but nanoscopic receptor organization with respect to complex cell surface morphology remains unclear. This is mainly due to a lack of accessible, robust and high-resolution methods. Here, we present an approach for mapping the topography of receptors at the cell surface with nanometer precision. The method involves coating glass coverslips with glycine, which preserves the fine membrane morphology while allowing immobilized cells to be positioned close to the optical surface. We developed an advanced and simplified algorithm for the analysis of single-molecule localization data acquired in a biplane detection scheme. These advancements enable direct and quantitative mapping of protein distribution on ruffled plasma membranes with near isotropic 3D nanometer resolution. As demonstrated successfully for CD4 and CD45 receptors, the described workflow is a straightforward quantitative technique to study molecules and their interactions at the complex surface nanomorphology of differentiated metazoan cells. A super-resolution localisation-based method is shown to map receptor topography in immune cells, which allows quantitative study of molecules and their interactions at the complex surface nanomorphology of cells.
Collapse
|
26
|
Nicolò A, Linder AT, Jumaa H, Maity PC. The Determinants of B Cell Receptor Signaling as Prototype Molecular Biomarkers of Leukemia. Front Oncol 2022; 11:771669. [PMID: 34993136 PMCID: PMC8724047 DOI: 10.3389/fonc.2021.771669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 12/02/2021] [Indexed: 12/20/2022] Open
Abstract
Advanced genome-wide association studies (GWAS) identified several transforming mutations in susceptible loci which are recognized as valuable prognostic markers in chronic lymphocytic leukemia (CLL) and B cell lymphoma (BCL). Alongside, robust genetic manipulations facilitated the generation of preclinical mouse models to validate mutations associated with poor prognosis and refractory B cell malignancies. Taken together, these studies identified new prognostic markers that could achieve characteristics of precision biomarkers for molecular diagnosis. On the contrary, the idea of augmented B cell antigen receptor (BCR) signaling as a transforming cue has somewhat receded despite the efficacy of Btk and Syk inhibitors. Recent studies from several research groups pointed out that acquired mutations in BCR components serve as faithful biomarkers, which become important for precision diagnostics and therapy, due to their relevant role in augmented BCR signaling and CLL pathogenesis. For example, we showed that expression of a single point mutated immunoglobulin light chain (LC) recombined through the variable gene segment IGLV3-21, named IGLV3-21R110, marks severe CLL cases. In this perspective, we summarize the molecular mechanisms fine-tuning B cell transformation, focusing on immunoglobulin point mutations and recurrent mutations in tumor suppressors. We present a stochastic model for gain-of-autonomous BCR signaling and subsequent neoplastic transformation. Of note, additional mutational analyses on immunoglobulin heavy chain (HC) derived from non-subset #2 CLL IGLV3-21R110 cases endorses our perspective. Altogether, we propose a model of malignant transformation in which the augmented BCR signaling creates a conducive platform for the appearance of transforming mutations.
Collapse
Affiliation(s)
| | | | - Hassan Jumaa
- Institute of Immunology, Ulm University, Ulm, Germany
| | | |
Collapse
|
27
|
Mazzarello AN, Gentner-Göbel E, Dühren-von Minden M, Tarasenko TN, Nicolò A, Ferrer G, Vergani S, Liu Y, Bagnara D, Rai KR, Burger JA, McGuire PJ, Maity PC, Jumaa H, Chiorazzi N. B-cell receptor isotypes differentially associate with cell signaling, kinetics, and outcome in chronic lymphocytic leukemia. J Clin Invest 2021; 132:149308. [PMID: 34813501 PMCID: PMC8759784 DOI: 10.1172/jci149308] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 11/17/2021] [Indexed: 11/17/2022] Open
Abstract
In chronic lymphocytic leukemia (CLL), the B cell receptor (BCR) plays a critical role in disease development and progression, as indicated by the therapeutic efficacy of drugs blocking BCR signaling. However, the mechanism(s) underlying BCR responsiveness are not completely defined. Selective engagement of membrane IgM or IgD on CLL cells, each coexpressed by more than 90% of cases, leads to distinct signaling events. Since both IgM and IgD carry the same antigen-binding domains, the divergent actions of the receptors are attributed to differences in immunoglobulin (Ig) structure or the outcome of signal transduction. We showed that IgM, not IgD, level and organization associated with CLL-cell birth rate and the type and consequences of BCR signaling in humans and mice. The latter IgM-driven effects were abrogated when BCR signaling was inhibited. Collectively, these studies demonstrated a critical, selective role for IgM in BCR signaling and B cell fate decisions, possibly opening new avenues for CLL therapy.
Collapse
Affiliation(s)
- Andrea N Mazzarello
- Karches Center for Oncology Research, The Feinstein Institute for Medical Research, Manhasset, United States of America
| | | | | | - Tatyana N Tarasenko
- Metabolism, Infection and Immunity Section, National Institutes of Health, Bethesda, United States of America
| | | | - Gerardo Ferrer
- Karches Center for Oncology Research, The Feinstein Institute for Medical Research, Manhasset, United States of America
| | - Stefano Vergani
- Karches Center for Oncology Research, The Feinstein Institute for Medical Research, Manhasset, United States of America
| | - Yun Liu
- Karches Center for Oncology Research, The Feinstein Institute for Medical Research, Manhasset, United States of America
| | - Davide Bagnara
- Karches Center for Oncology Research, The Feinstein Institute for Medical Research, Manhasset, United States of America
| | - Kanti R Rai
- Karches Center for Oncology Research, The Feinstein Institute for Medical Research, Manhasset, United States of America
| | - Jan A Burger
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Peter J McGuire
- National Institutes of Health, Bethesda, United States of America
| | - Palash C Maity
- Institute for Immunology, University Hospital Ulm, Ulm, Germany
| | - Hassan Jumaa
- Institute for Immunology, University Hospital Ulm, Ulm, Germany
| | - Nicholas Chiorazzi
- Karches Center for Oncology Research, The Feinstein Institute for Medical Research, Manhasset, United States of America
| |
Collapse
|
28
|
Glowacki SK, Gomes de Castro MA, Yip KM, Asadpour O, Münchhalfen M, Engels N, Opazo F. A fluorescent probe for STED microscopy to study NIP-specific B cells. Analyst 2021; 146:4744-4747. [PMID: 34226908 DOI: 10.1039/d1an00601k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We have developed a series of monovalent fluorophore-conjugated affinity probes based on the hapten 3-nitro-4-hydroxy-5-iodophenylacetyl (NIP), which is widely used as a model antigen to study B lymphocytes and the functional principles of B cell antigen receptors (BCRs). We successfully used them in flow-cytometry, confocal and super-resolution microscopy techniques.
Collapse
Affiliation(s)
- Selda Kabatas Glowacki
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073, Göttingen, Germany
| | | | | | | | | | | | | |
Collapse
|
29
|
Valli J, Garcia-Burgos A, Rooney LM, Vale de Melo E Oliveira B, Duncan RR, Rickman C. Seeing beyond the limit: A guide to choosing the right super-resolution microscopy technique. J Biol Chem 2021; 297:100791. [PMID: 34015334 PMCID: PMC8246591 DOI: 10.1016/j.jbc.2021.100791] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 05/14/2021] [Accepted: 05/14/2021] [Indexed: 02/06/2023] Open
Abstract
Super-resolution microscopy has become an increasingly popular and robust tool across the life sciences to study minute cellular structures and processes. However, with the increasing number of available super-resolution techniques has come an increased complexity and burden of choice in planning imaging experiments. Choosing the right super-resolution technique to answer a given biological question is vital for understanding and interpreting biological relevance. This is an often-neglected and complex task that should take into account well-defined criteria (e.g., sample type, structure size, imaging requirements). Trade-offs in different imaging capabilities are inevitable; thus, many researchers still find it challenging to select the most suitable technique that will best answer their biological question. This review aims to provide an overview and clarify the concepts underlying the most commonly available super-resolution techniques as well as guide researchers through all aspects that should be considered before opting for a given technique.
Collapse
Affiliation(s)
- Jessica Valli
- Edinburgh Super Resolution Imaging Consortium (ESRIC), Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Edinburgh, United Kingdom.
| | - Adrian Garcia-Burgos
- Edinburgh Super Resolution Imaging Consortium (ESRIC), Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Edinburgh, United Kingdom
| | - Liam M Rooney
- Edinburgh Super Resolution Imaging Consortium (ESRIC), Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Edinburgh, United Kingdom
| | - Beatriz Vale de Melo E Oliveira
- Edinburgh Super Resolution Imaging Consortium (ESRIC), Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Edinburgh, United Kingdom
| | - Rory R Duncan
- Edinburgh Super Resolution Imaging Consortium (ESRIC), Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Edinburgh, United Kingdom
| | - Colin Rickman
- Edinburgh Super Resolution Imaging Consortium (ESRIC), Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Edinburgh, United Kingdom.
| |
Collapse
|
30
|
He H, Liu L, Chen X, Wang Q, Wang X, Nau WM, Huang F. Carbon Dot Blinking Enables Accurate Molecular Counting at Nanoscale Resolution. Anal Chem 2021; 93:3968-3975. [DOI: 10.1021/acs.analchem.0c04885] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Hua He
- State Key Laboratory of Heavy Oil Processing and College of Chemical Engineering China, University of Petroleum (East China), Qingdao 266580, China
| | - Lihua Liu
- State Key Laboratory of Heavy Oil Processing and College of Chemical Engineering China, University of Petroleum (East China), Qingdao 266580, China
| | - Xiaoliang Chen
- State Key Laboratory of Heavy Oil Processing and College of Chemical Engineering China, University of Petroleum (East China), Qingdao 266580, China
| | - Qian Wang
- State Key Laboratory of Heavy Oil Processing and College of Chemical Engineering China, University of Petroleum (East China), Qingdao 266580, China
| | - Xiaojuan Wang
- State Key Laboratory of Heavy Oil Processing and College of Chemical Engineering China, University of Petroleum (East China), Qingdao 266580, China
| | - Werner M. Nau
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Campus Ring 1, Bremen 28759, Germany
| | - Fang Huang
- State Key Laboratory of Heavy Oil Processing and College of Chemical Engineering China, University of Petroleum (East China), Qingdao 266580, China
| |
Collapse
|
31
|
A quantitative view on multivalent nanomedicine targeting. Adv Drug Deliv Rev 2021; 169:1-21. [PMID: 33264593 DOI: 10.1016/j.addr.2020.11.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 11/11/2020] [Accepted: 11/21/2020] [Indexed: 12/17/2022]
Abstract
Although the concept of selective delivery has been postulated over 100 years ago, no targeted nanomedicine has been clinically approved so far. Nanoparticles modified with targeting ligands to promote the selective delivery of therapeutics towards a specific cell population have been extensively reported. However, the rational design of selective particles is still challenging. One of the main reasons for this is the lack of quantitative theoretical and experimental understanding of the interactions involved in cell targeting. In this review, we discuss new theoretical models and experimental methods that provide a quantitative view of targeting. We show the new advancements in multivalency theory enabling the rational design of super-selective nanoparticles. Furthermore, we present the innovative approaches to obtain key targeting parameters at the single-cell and single molecule level and their role in the design of targeting nanoparticles. We believe that the combination of new theoretical multivalent design and experimental methods to quantify receptors and ligands aids in the rational design and clinical translation of targeted nanomedicines.
Collapse
|
32
|
Caveolin-1, tetraspanin CD81 and flotillins in lymphocyte cell membrane organization, signaling and immunopathology. Biochem Soc Trans 2020; 48:2387-2397. [PMID: 33242069 DOI: 10.1042/bst20190387] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/08/2020] [Accepted: 10/12/2020] [Indexed: 12/24/2022]
Abstract
The adaptive immune system relies on B and T lymphocytes to ensure a specific and long-lasting protection of an individual from a wide range of potential pathogenic hits. Lymphocytes are highly potent and efficient in eliminating pathogens. However, lymphocyte activation must be tightly regulated to prevent incorrect activity that could result in immunopathologies, such as autoimmune disorders or cancers. Comprehensive insight into the molecular events underlying lymphocyte activation is of enormous importance to better understand the function of the immune system. It provides the basis to design therapeutics to regulate lymphocyte activation in pathological scenarios. Most reported defects in immunopathologies affect the regulation of intracellular signaling pathways. This highlights the importance of these molecules, which control lymphocyte activation and homeostasis impacting lymphocyte tolerance to self, cytokine production and responses to infections. Most evidence for these defects comes from studies of disease models in genetically engineered mice. There is an increasing number of studies focusing on lymphocytes derived from patients which supports these findings. Many indirectly involved proteins are emerging as unexpected regulators of the immune system. In this mini-review, we focus in proteins that regulate plasma membrane (PM) compartmentalization and thereby impact the steady state and the activation of immunoreceptors, namely the T cell antigen receptor (TCR) and the B cell antigen receptor (BCR). Some of these membrane proteins are shown to be involved in immune abnormalities; others, however, are not thoroughly investigated in the context of immune pathogenesis. We aim to highlight them and stimulate future research avenues.
Collapse
|
33
|
Thurner L, Hartmann S, Neumann F, Hoth M, Stilgenbauer S, Küppers R, Preuss KD, Bewarder M. Role of Specific B-Cell Receptor Antigens in Lymphomagenesis. Front Oncol 2020; 10:604685. [PMID: 33363034 PMCID: PMC7756126 DOI: 10.3389/fonc.2020.604685] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/02/2020] [Indexed: 12/22/2022] Open
Abstract
The B-cell receptor (BCR) signaling pathway is a crucial pathway of B cells, both for their survival and for antigen-mediated activation, proliferation and differentiation. Its activation is also critical for the genesis of many lymphoma types. BCR-mediated lymphoma proliferation may be caused by activating BCR-pathway mutations and/or by active or tonic stimulation of the BCR. BCRs of lymphomas have frequently been described as polyreactive. In this review, the role of specific target antigens of the BCRs of lymphomas is highlighted. These antigens have been found to be restricted to specific lymphoma entities. The antigens can be of infectious origin, such as H. pylori in gastric MALT lymphoma or RpoC of M. catarrhalis in nodular lymphocyte predominant Hodgkin lymphoma, or they are autoantigens. Examples of such autoantigens are the BCR itself in chronic lymphocytic leukemia, LRPAP1 in mantle cell lymphoma, hyper-N-glycosylated SAMD14/neurabin-I in primary central nervous system lymphoma, hypo-phosphorylated ARS2 in diffuse large B-cell lymphoma, and hyper-phosphorylated SLP2, sumoylated HSP90 or saposin C in plasma cell dyscrasia. Notably, atypical posttranslational modifications are often responsible for the immunogenicity of many autoantigens. Possible therapeutic approaches evolving from these specific antigens are discussed.
Collapse
Affiliation(s)
- Lorenz Thurner
- Department of Internal Medicine I, José Carreras Center for Immuno- and Gene Therapy, Saarland University Medical School, Homburg, Germany
| | - Sylvia Hartmann
- Dr. Senckenberg Institute of Pathology, Goethe University, Frankfurt a. Main, Germany
| | - Frank Neumann
- Department of Internal Medicine I, José Carreras Center for Immuno- and Gene Therapy, Saarland University Medical School, Homburg, Germany
| | - Markus Hoth
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, Homburg, Germany
| | - Stephan Stilgenbauer
- Department of Internal Medicine I, José Carreras Center for Immuno- and Gene Therapy, Saarland University Medical School, Homburg, Germany
| | - Ralf Küppers
- Medical School, Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Essen, Germany.,Deutsches Konsortium für translationale Krebsforschung (DKTK), Partner Site Essen, Essen, Germany
| | - Klaus-Dieter Preuss
- Department of Internal Medicine I, José Carreras Center for Immuno- and Gene Therapy, Saarland University Medical School, Homburg, Germany
| | - Moritz Bewarder
- Department of Internal Medicine I, José Carreras Center for Immuno- and Gene Therapy, Saarland University Medical School, Homburg, Germany
| |
Collapse
|
34
|
van Deventer S, Arp AB, van Spriel AB. Dynamic Plasma Membrane Organization: A Complex Symphony. Trends Cell Biol 2020; 31:119-129. [PMID: 33248874 DOI: 10.1016/j.tcb.2020.11.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 11/02/2020] [Accepted: 11/06/2020] [Indexed: 01/20/2023]
Abstract
Membrane protein organization is essential for proper cellular functioning and the result of a dynamic exchange between protein monomers, nanoscale protein clusters, and microscale higher-order structures. This exchange is affected by both lipid bilayer intrinsic factors, such as lipid rafts and tetraspanins, and extrinsic factors, such as cortical actin and galectins. Because membrane organizers act jointly like instruments in a symphony, it is challenging to define the 'key' organizers. Here, we posit, for the first time, definitions of key intrinsic and extrinsic membrane organizers. Tetraspanin nanodomains are key organizers that are often overlooked. We discuss how different key organizers can collaborate, which is important to get a full grasp of plasma membrane biology.
Collapse
Affiliation(s)
- Sjoerd van Deventer
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Abbey B Arp
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Annemiek B van Spriel
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
35
|
Chen H, Xie X, Chen TY. Single-molecule microscopy for in-cell quantification of protein oligomeric stoichiometry. Curr Opin Struct Biol 2020; 66:112-118. [PMID: 33242727 DOI: 10.1016/j.sbi.2020.10.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 10/14/2020] [Accepted: 10/19/2020] [Indexed: 12/11/2022]
Abstract
Protein organization modification plays a vital role in initiating signaling pathways, transcriptional regulation, and cell apoptosis regulation. Simultaneous quantification of oligomeric state and cellular parameters in the same cell, even though challenging, is required to understand their correlation at the molecular level. Recent advances of fluorescence protein and single-molecule localization microscopy enables the determination of localizations and oligomeric states of target proteins in cells. We reviewed the fluorescence intensity-based, localization-based, and photophysical property-based approaches for in-cell quantification of protein oligomeric stoichiometry. We discussed their working principles, applications, advantages, and limitations. These results also imply the combination of methodologies targeting different biological parameters at the single-cell level is essential to uncover the structure-function relationship at the molecular level.
Collapse
Affiliation(s)
- Huanhuan Chen
- Department of Chemistry, University of Houston, Houston, TX 77204, United States
| | - Xihong Xie
- Department of Chemistry, University of Houston, Houston, TX 77204, United States
| | - Tai-Yen Chen
- Department of Chemistry, University of Houston, Houston, TX 77204, United States.
| |
Collapse
|
36
|
Haselager MV, Kater AP, Eldering E. Proliferative Signals in Chronic Lymphocytic Leukemia; What Are We Missing? Front Oncol 2020; 10:592205. [PMID: 33134182 PMCID: PMC7578574 DOI: 10.3389/fonc.2020.592205] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 09/18/2020] [Indexed: 12/23/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) cells cycle between lymphoid tissue sites where they actively proliferate, and the peripheral blood (PB) where they become quiescent. Strong evidence exists for a crucial role of B cell receptor (BCR) triggering, either by (self-)antigen or by receptor auto-engagement in the lymph node (LN) to drive CLL proliferation and provide adhesion. The clinical success of Bruton's tyrosine kinase (BTK) inhibitors is widely accepted to be based on blockade of the BCR signal. Additional signals in the LN that support CLL survival derive from surrounding cells, such as CD40L-presenting T helper cells, myeloid and stromal cells. It is not quite clear if and to what extent these non-BCR signals contribute to proliferation in situ. In vitro BCR triggering, in contrast, leads to low-level activation and does not result in cell division. Various combinations of non-BCR signals delivered via co-stimulatory receptors, Toll-like receptors (TLRs), and/or soluble cytokines are applied, leading to comparatively modest and short-lived CLL proliferation in vitro. Thus, an unresolved gap exists between the condition in the patient as we now understand it and applicable knowledge that can be harnessed in the laboratory for future therapeutic applications. Even in this era of targeted drugs, CLL remains largely incurable with frequent relapses and emergence of resistance. Therefore, we require better insight into all aspects of CLL growth and potential rewiring of signaling pathways. We aim here to provide an overview of in vivo versus in vitro signals involved in CLL proliferation, point out areas of missing knowledge and suggest future directions for research.
Collapse
Affiliation(s)
- Marco V. Haselager
- Department of Experimental Immunology, Academic University Medical Center, location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Lymphoma and Myeloma Center Amsterdam, LYMMCARE, Amsterdam, Netherlands
- Cancer Center Amsterdam, LYMMCARE, Amsterdam, Netherlands
- Amsterdam Infection & Immunity Institute, Amsterdam, Netherlands
| | - Arnon P. Kater
- Cancer Center Amsterdam, LYMMCARE, Amsterdam, Netherlands
- Amsterdam Infection & Immunity Institute, Amsterdam, Netherlands
- Department of Hematology, Academic University Medical Center, location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Eric Eldering
- Department of Experimental Immunology, Academic University Medical Center, location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
- Lymphoma and Myeloma Center Amsterdam, LYMMCARE, Amsterdam, Netherlands
- Cancer Center Amsterdam, LYMMCARE, Amsterdam, Netherlands
- Amsterdam Infection & Immunity Institute, Amsterdam, Netherlands
| |
Collapse
|
37
|
Jing Y, Cai M, Zhou L, Jiang J, Gao J, Wang H. Aptamer AS1411 utilized for super-resolution imaging of nucleolin. Talanta 2020; 217:121037. [PMID: 32498876 DOI: 10.1016/j.talanta.2020.121037] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 04/08/2020] [Accepted: 04/11/2020] [Indexed: 12/14/2022]
Abstract
Nucleolin (NCL) is a multifunctional protein that mainly localizes in the nucleolus and also distributes in the nucleoplasm, cytoplasm and cell membrane. Most studies focus on its biofunctions in cell activities and diseases, however, its detailed distribution and organization pattern in situ remains obscure. Moreover, antibodies were commonly used to investigate NCL in cells. It is worth noting that antibody labeling of intracellular proteins needs detergents to permeabilize the membrane, which could disrupt the membrane structure and proteins. The emergence of aptamer AS1411 provides us a good choice to recognize the NCL without permeabilization owing to its superior cellular uptake and enhanced stability. Therefore, we applied aptamer AS1411 to super-resolution imaging to visualize the distribution of NCL at a nanometer level. Aptamer achieved a better recognition of intracellular NCL and displayed the detailed structure of NCL in different parts of cells. Significantly, cytoplasmic and membrane NCL have higher expression and larger clusters in cancer cells than that in normal cells. Our work presented a detailed organization of NCL in cells and revealed the distribution differences between cancer cells and normal cells, which promote the understanding of its functions in physiology and pathology.
Collapse
Affiliation(s)
- Yingying Jing
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China; University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Mingjun Cai
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Lulu Zhou
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Junguang Jiang
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Jing Gao
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China.
| | - Hongda Wang
- State Key Laboratory of Electroanalytical Chemistry, Research Center of Biomembranomics, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China; University of Science and Technology of China, Hefei, Anhui, 230027, China; Laboratory for Marine Biology and Biotechnology, Qing Dao National Laboratory for Marine Science and Technology, Wenhai Road, Aoshanwei, Jimo, Qingdao, Shandong, 266237, China.
| |
Collapse
|
38
|
Orinska Z, Hagemann PM, Halova I, Draber P. Tetraspanins in the regulation of mast cell function. Med Microbiol Immunol 2020; 209:531-543. [PMID: 32507938 PMCID: PMC7395004 DOI: 10.1007/s00430-020-00679-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 05/06/2020] [Indexed: 12/13/2022]
Abstract
Mast cells (MCs) are long-living immune cells highly specialized in the storage and release of different biologically active compounds and are involved in the regulation of innate and adaptive immunity. MC degranulation and replacement of MC granules are accompanied by active membrane remodelling. Tetraspanins represent an evolutionary conserved family of transmembrane proteins. By interacting with lipids and other membrane and intracellular proteins, they are involved in organisation of membrane protein complexes and act as "molecular facilitators" connecting extracellular and cytoplasmic signaling elements. MCs express different tetraspanins and MC degranulation is accompanied by changes in membrane organisation. Therefore, tetraspanins are very likely involved in the regulation of MC exocytosis and membrane reorganisation after degranulation. Antiviral response and production of exosomes are further aspects of MC function characterized by dynamic changes of membrane organization. In this review, we pay a particular attention to tetraspanin gene expression in different human and murine MC populations, discuss tetraspanin involvement in regulation of key MC signaling complexes, and analyze the potential contribution of tetraspanins to MC antiviral response and exosome production. In-depth knowledge of tetraspanin-mediated molecular mechanisms involved in different aspects of the regulation of MC response will be beneficial for patients with allergies, characterized by overwhelming MC reactions.
Collapse
Affiliation(s)
- Zane Orinska
- Division of Experimental Pneumology, Research Center Borstel, Leibniz Lungenzentrum, Airway Research Center North, German Center for Lung Research (DZL), Borstel, Germany.
| | - Philipp M Hagemann
- Division of Experimental Pneumology, Research Center Borstel, Leibniz Lungenzentrum, Airway Research Center North, German Center for Lung Research (DZL), Borstel, Germany
| | - Ivana Halova
- Department of Signal Transduction, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Petr Draber
- Department of Signal Transduction, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
39
|
Mechanisms of B Cell Receptor Activation and Responses to B Cell Receptor Inhibitors in B Cell Malignancies. Cancers (Basel) 2020; 12:cancers12061396. [PMID: 32481736 PMCID: PMC7352865 DOI: 10.3390/cancers12061396] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/22/2020] [Accepted: 05/25/2020] [Indexed: 12/27/2022] Open
Abstract
The B cell receptor (BCR) pathway has been identified as a potential therapeutic target in a number of common B cell malignancies, including chronic lymphocytic leukemia, diffuse large B cell lymphoma, Burkitt lymphoma, follicular lymphoma, mantle cell lymphoma, marginal zone B cell lymphoma, and Waldenstrom's macroglobulinemia. This finding has resulted in the development of numerous drugs that target this pathway, including various inhibitors of the kinases BTK, PI3K, and SYK. Several of these drugs have been approved in recent years for clinical use, resulting in a profound change in the way these diseases are currently being treated. However, the response rates and durability of responses vary largely across the different disease entities, suggesting a different proportion of patients with an activated BCR pathway and different mechanisms of BCR pathway activation. Indeed, several antigen-dependent and antigen-independent mechanisms have recently been described and shown to result in the activation of distinct downstream signaling pathways. The purpose of this review is to provide an overview of the mechanisms responsible for the activation of the BCR pathway in different B cell malignancies and to correlate these mechanisms with clinical responses to treatment with BCR inhibitors.
Collapse
|
40
|
Sograte-Idrissi S, Schlichthaerle T, Duque-Afonso CJ, Alevra M, Strauss S, Moser T, Jungmann R, Rizzoli SO, Opazo F. Circumvention of common labelling artefacts using secondary nanobodies. NANOSCALE 2020; 12:10226-10239. [PMID: 32356544 DOI: 10.1039/d0nr00227e] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
A standard procedure to study cellular elements is via immunostaining followed by optical imaging. This methodology typically requires target-specific primary antibodies (1.Abs), which are revealed by secondary antibodies (2.Abs). Unfortunately, the antibody bivalency, polyclonality, and large size can result in a series of artifacts. Alternatively, small, monovalent probes, such as single-domain antibodies (nanobodies) have been suggested to minimize these limitations. The discovery and validation of nanobodies against specific targets are challenging, thus only a minimal amount of them are currently available. Here, we used STED, DNA-PAINT, and light-sheet microscopy, to demonstrate that secondary nanobodies (1) increase localization accuracy compared to 2.Abs; (2) allow direct pre-mixing with 1.Abs before staining, reducing experimental time, and enabling the use of multiple 1.Abs from the same species; (3) penetrate thick tissues more efficiently; and (4) avoid probe-induced clustering of target molecules observed with conventional 2.Abs in living or poorly fixed samples. Altogether, we show how secondary nanobodies are a valuable alternative to 2.Abs.
Collapse
Affiliation(s)
- Shama Sograte-Idrissi
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073 Göttingen, Germany. and Center for Biostructural Imaging of Neurodegeneration (BIN), University of Göttingen Medical Center, 37075 Göttingen, Germany and International Max Planck Research School for Molecular Biology, Göttingen, Germany
| | - Thomas Schlichthaerle
- Faculty of Physics and Center for Nanoscience, LMU Munich, 80539, Munich, Germany and Max Planck Institute of Biochemistry, 82152, Martinsried, Germany
| | - Carlos J Duque-Afonso
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany and Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany and Multiscale Bioimaging Cluster of Excellence (MBExC), University of Göttingen, 37075 Göttingen, Germany and University of Göttingen, 37075, Göttingen, Germany
| | - Mihai Alevra
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073 Göttingen, Germany.
| | - Sebastian Strauss
- Faculty of Physics and Center for Nanoscience, LMU Munich, 80539, Munich, Germany and Max Planck Institute of Biochemistry, 82152, Martinsried, Germany
| | - Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany and Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany and Multiscale Bioimaging Cluster of Excellence (MBExC), University of Göttingen, 37075 Göttingen, Germany and University of Göttingen, 37075, Göttingen, Germany
| | - Ralf Jungmann
- Faculty of Physics and Center for Nanoscience, LMU Munich, 80539, Munich, Germany and Max Planck Institute of Biochemistry, 82152, Martinsried, Germany
| | - Silvio O Rizzoli
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073 Göttingen, Germany. and Center for Biostructural Imaging of Neurodegeneration (BIN), University of Göttingen Medical Center, 37075 Göttingen, Germany and Multiscale Bioimaging Cluster of Excellence (MBExC), University of Göttingen, 37075 Göttingen, Germany
| | - Felipe Opazo
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073 Göttingen, Germany. and Center for Biostructural Imaging of Neurodegeneration (BIN), University of Göttingen Medical Center, 37075 Göttingen, Germany and NanoTag Biotechnologies GmbH, 37079, Göttingen, Germany
| |
Collapse
|
41
|
Feng Y, Wang Y, Zhang S, Haneef K, Liu W. Structural and immunogenomic insights into B-cell receptor activation. J Genet Genomics 2020; 47:27-35. [PMID: 32111437 DOI: 10.1016/j.jgg.2019.12.003] [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: 07/17/2019] [Revised: 11/10/2019] [Accepted: 12/09/2019] [Indexed: 02/08/2023]
Abstract
B cells express B-cell receptors (BCRs) which recognize antigen to trigger signaling cascades for B-cell activation and subsequent antibody production. BCR activation has a crucial influence on B-cell fate. How BCR is activated upon encountering antigen remains to be solved, although tremendous progresses have been achieved in the past few years. Here, we summarize the models that have been proposed to explain BCR activation, including the cross-linking model, the conformation-induced oligomerization model, the dissociation activation model, and the conformational change model. Especially, we elucidate the partially resolved structures of antibodies and/or BCRs by far and discusse how these current structural and further immunogenomic messages and more importantly the future studies may shed light on the explanation of BCR activation and the relevant diseases in the case of dysregulation.
Collapse
Affiliation(s)
- Yangyang Feng
- MOE Key Laboratory of Protein Sciences, Center for Life Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing, 100084, China
| | - Yu Wang
- MOE Key Laboratory of Protein Sciences, Center for Life Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing, 100084, China
| | - Shaocun Zhang
- MOE Key Laboratory of Protein Sciences, Center for Life Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing, 100084, China
| | - Kabeer Haneef
- MOE Key Laboratory of Protein Sciences, Center for Life Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing, 100084, China
| | - Wanli Liu
- MOE Key Laboratory of Protein Sciences, Center for Life Sciences, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Life Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
42
|
Jing Y, Zhou L, Chen J, Xu H, Sun J, Cai M, Jiang J, Gao J, Wang H. Quantitatively Mapping the Assembly Pattern of EpCAM on Cell Membranes with Peptide Probes. Anal Chem 2019; 92:1865-1873. [DOI: 10.1021/acs.analchem.9b03901] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Yingying Jing
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Research Center of Biomembranomics, Changchun, Jilin 130022, China
- University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Lulu Zhou
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Research Center of Biomembranomics, Changchun, Jilin 130022, China
| | - Junling Chen
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Research Center of Biomembranomics, Changchun, Jilin 130022, China
| | - Haijiao Xu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Research Center of Biomembranomics, Changchun, Jilin 130022, China
| | - Jiayin Sun
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Research Center of Biomembranomics, Changchun, Jilin 130022, China
| | - Mingjun Cai
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Research Center of Biomembranomics, Changchun, Jilin 130022, China
| | - Junguang Jiang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Research Center of Biomembranomics, Changchun, Jilin 130022, China
| | - Jing Gao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Research Center of Biomembranomics, Changchun, Jilin 130022, China
| | - Hongda Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Research Center of Biomembranomics, Changchun, Jilin 130022, China
- University of Science and Technology of China, Hefei, Anhui 230027, China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Wenhai Road, Aoshanwei, Jimo, Qingdao, Shandong 266237, China
| |
Collapse
|
43
|
Li M, Ding H, Lin M, Yin F, Song L, Mao X, Li F, Ge Z, Wang L, Zuo X, Ma Y, Fan C. DNA Framework-Programmed Cell Capture via Topology-Engineered Receptor-Ligand Interactions. J Am Chem Soc 2019; 141:18910-18915. [PMID: 31691568 DOI: 10.1021/jacs.9b11015] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Receptor-ligand interactions (RLIs) that play pivotal roles in living organisms are often depicted with the classic keys-and-locks model. Nevertheless, RLIs on the cell surface are generally highly complex and nonlinear, partially due to the noncontinuous and dynamic distribution of receptors on extracellular membranes. Here, we develop a tetrahedral DNA framework (TDF)-programmed approach to topologically engineer RLIs on the cell membrane, which enables active recruitment-binding of clustered receptors for high-affinity capture of circulating tumor cells (CTCs). The four vertices of a TDF afford orthogonal anchoring of ligands with spatial organization, based on which we synthesized n-simplexes harboring 1-3 aptamers targeting epithelial cell adhesion molecule (EpCAM) that are overexpressed on the membrane of tumor cells. The 2-simplex with three aptamers not only shows increased binding affinity (∼19-fold) but prevents endocytosis by cells. By using 2-simplex as the capture probe, we demonstrate the high-efficiency CTC capture, which is challenged in real clinical breast cancer patient samples. This TDF-programmed platform thus provides a powerful means for studying RLIs in physiological settings and for cancer diagnosis.
Collapse
Affiliation(s)
- Min Li
- Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine and School of Chemistry and Chemical Engineering , Shanghai Jiao Tong University , Shanghai 200127 , China
| | - Hongming Ding
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, School of Physical Science and Technology , Soochow University , Suzhou 215006 , China
| | - Meihua Lin
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry , China University of Geosciences , Wuhan 430074 , China
| | - Fangfei Yin
- Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine and School of Chemistry and Chemical Engineering , Shanghai Jiao Tong University , Shanghai 200127 , China.,Division of Physical Biology and Bioimaging Center, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics , Chinese Academy of Sciences , Shanghai 201800 , China
| | - Lu Song
- Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine and School of Chemistry and Chemical Engineering , Shanghai Jiao Tong University , Shanghai 200127 , China.,Division of Physical Biology and Bioimaging Center, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics , Chinese Academy of Sciences , Shanghai 201800 , China
| | - Xiuhai Mao
- Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine and School of Chemistry and Chemical Engineering , Shanghai Jiao Tong University , Shanghai 200127 , China
| | - Fan Li
- Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine and School of Chemistry and Chemical Engineering , Shanghai Jiao Tong University , Shanghai 200127 , China
| | - Zhilei Ge
- Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine and School of Chemistry and Chemical Engineering , Shanghai Jiao Tong University , Shanghai 200127 , China
| | - Lihua Wang
- Division of Physical Biology and Bioimaging Center, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics , Chinese Academy of Sciences , Shanghai 201800 , China
| | - Xiaolei Zuo
- Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine and School of Chemistry and Chemical Engineering , Shanghai Jiao Tong University , Shanghai 200127 , China
| | - Yuqiang Ma
- National Laboratory of Solid State Microstructures and Department of Physics, Collaborative Innovation Center of Advanced Microstructures , Nanjing University , Nanjing 210093 , China
| | - Chunhai Fan
- Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine and School of Chemistry and Chemical Engineering , Shanghai Jiao Tong University , Shanghai 200127 , China
| |
Collapse
|
44
|
Dietz MS, Heilemann M. Optical super-resolution microscopy unravels the molecular composition of functional protein complexes. NANOSCALE 2019; 11:17981-17991. [PMID: 31573593 DOI: 10.1039/c9nr06364a] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Optical super-resolution microscopy has revolutionized our understanding of cell biology. Next to visualizing cellular structures with near-molecular spatial resolution, an additional benefit is the molecular characterization of biomolecular complexes directly in an intact cell. Single-molecule localization microscopy, as one technology out of the toolbox of super-resolution methods, generates images by detecting the position of single fluorophore labels and is particularly suited for molecular quantification. We review imaging and analysis methods employing single-molecule localization microscopy and extract molecule numbers.
Collapse
Affiliation(s)
- Marina S Dietz
- Single Molecule Biophysics, Institute of Physical and Theoretical Chemistry, Goethe University Frankfurt, 60438 Frankfurt, Germany.
| | | |
Collapse
|
45
|
Ziegler CGK, Kim J, Piersanti K, Oyler-Yaniv A, Argyropoulos KV, van den Brink MRM, Palomba ML, Altan-Bonnet N, Altan-Bonnet G. Constitutive Activation of the B Cell Receptor Underlies Dysfunctional Signaling in Chronic Lymphocytic Leukemia. Cell Rep 2019; 28:923-937.e3. [PMID: 31340154 PMCID: PMC8018719 DOI: 10.1016/j.celrep.2019.06.069] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/18/2019] [Accepted: 06/19/2019] [Indexed: 12/31/2022] Open
Abstract
In cancer biology, the functional interpretation of genomic alterations is critical to achieve the promise of genomic profiling in the clinic. For chronic lymphocytic leukemia (CLL), a heterogeneous disease of B-lymphocytes maturing under constitutive B cell receptor (BCR) stimulation, the functional role of diverse clonal mutations remains largely unknown. Here, we demonstrate that alterations in BCR signaling dynamics underlie the progression of B cells toward malignancy. We reveal emergent dynamic features—bimodality, hypersensitivity, and hysteresis—in the BCR signaling pathway of primary CLL B cells. These signaling abnormalities in CLL quantitatively derive from BCR clustering and constitutive signaling with positive feedback reinforcement, as demonstrated through single-cell analysis of phospho-responses, computational modeling, and super-resolution imaging. Such dysregulated signaling segregates CLL patients by disease severity and clinical presentation. These findings provide a quantitative framework and methodology to assess complex and heterogeneous leukemia pathology and to inform therapeutic strategies in parallel with genomic profiling. Using phospho-flow cytometry and computational modeling, Ziegler et al. find that B cell receptor clustering and positive feedback through SYK and LYN drive signaling hypersensitivity, bistability, and hysteresis in chronic lymphocytic leukemic B cells. Super-resolution microscopy confirms membrane auto-aggregation in leukemic B cells, and variability in signaling dysfunction predicts disease severity.
Collapse
Affiliation(s)
- Carly G K Ziegler
- ImmunoDynamics Group, Programs in Computational Biology and Immunology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Center for Cancer Systems Biology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Joel Kim
- ImmunoDynamics Group, Programs in Computational Biology and Immunology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Center for Cancer Systems Biology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Kelly Piersanti
- Center for Cancer Systems Biology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Alon Oyler-Yaniv
- ImmunoDynamics Group, Programs in Computational Biology and Immunology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Physics Department, Ben Gurion University, Beer-Sheva, Israel
| | - Kimon V Argyropoulos
- Center for Cancer Systems Biology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Marcel R M van den Brink
- Center for Cancer Systems Biology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - M Lia Palomba
- Center for Cancer Systems Biology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | - Grégoire Altan-Bonnet
- ImmunoDynamics Group, Programs in Computational Biology and Immunology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Center for Cancer Systems Biology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
46
|
Tenspolde M, Zimmermann K, Weber LC, Hapke M, Lieber M, Dywicki J, Frenzel A, Hust M, Galla M, Buitrago-Molina LE, Manns MP, Jaeckel E, Hardtke-Wolenski M. Regulatory T cells engineered with a novel insulin-specific chimeric antigen receptor as a candidate immunotherapy for type 1 diabetes. J Autoimmun 2019; 103:102289. [PMID: 31176558 DOI: 10.1016/j.jaut.2019.05.017] [Citation(s) in RCA: 152] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/23/2019] [Accepted: 05/26/2019] [Indexed: 12/14/2022]
Abstract
Adoptive immunotherapy with ex vivo expanded, polyspecific regulatory T cells (Tregs) is a promising treatment for graft-versus-host disease. Animal transplantation models used by us and others have demonstrated that the adoptive transfer of allospecific Tregs offers greater protection from graft rejection than that of polyclonal Tregs. This finding is in contrast to those of autoimmune models, where adoptive transfer of polyspecific Tregs had very limited effects, while antigen-specific Tregs were promising. However, antigen-specific Tregs in autoimmunity cannot be isolated in sufficient numbers. Chimeric antigen receptors (CARs) can modify T cells and redirect their specificity toward needed antigens and are currently clinically used in leukemia patients. A major benefit of CAR technology is its "off-the-shelf" usability in a translational setting in contrast to major histocompatibility complex (MHC)-restricted T cell receptors. We used CAR technology to redirect T cell specificity toward insulin and redirect T effector cells (Teffs) to Tregs by Foxp3 transduction. Our data demonstrate that our converted, insulin-specific CAR Tregs (cTregs) were functional stable, suppressive and long-lived in vivo. This is a proof of concept for both redirection of T cell specificity and conversion of Teffs to cTregs.
Collapse
Affiliation(s)
- Michel Tenspolde
- Dept. of Gastroenterology, Hepatology & Endocrinology, Hannover Medical School, Hannover, Germany
| | - Katharina Zimmermann
- Dept. of Gastroenterology, Hepatology & Endocrinology, Hannover Medical School, Hannover, Germany
| | - Leonie C Weber
- Dept. of Gastroenterology and Hepatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Martin Hapke
- Dept. of Gastroenterology, Hepatology & Endocrinology, Hannover Medical School, Hannover, Germany
| | - Maren Lieber
- Dept. of Gastroenterology, Hepatology & Endocrinology, Hannover Medical School, Hannover, Germany
| | - Janine Dywicki
- Dept. of Gastroenterology, Hepatology & Endocrinology, Hannover Medical School, Hannover, Germany
| | - Andre Frenzel
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany; YUMAB GmbH, Science Campus Braunschweig-Süd, Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Michael Hust
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany; YUMAB GmbH, Science Campus Braunschweig-Süd, Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Melanie Galla
- Institute of Experimental Haematology, Hannover Medical School, Hannover, Germany
| | - Laura E Buitrago-Molina
- Dept. of Gastroenterology and Hepatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Michael P Manns
- Dept. of Gastroenterology, Hepatology & Endocrinology, Hannover Medical School, Hannover, Germany
| | - Elmar Jaeckel
- Dept. of Gastroenterology, Hepatology & Endocrinology, Hannover Medical School, Hannover, Germany
| | - Matthias Hardtke-Wolenski
- Dept. of Gastroenterology, Hepatology & Endocrinology, Hannover Medical School, Hannover, Germany; Dept. of Gastroenterology and Hepatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|