Complement-Opsonized Nano-Carriers Are Bound by Dendritic Cells (DC) via Complement Receptor (CR)3, and by B Cell Subpopulations via CR-1/2, and Affect the Activation of DC and B-1 Cells

The development of nanocarriers (NC) for biomedical applications has gained large interest due to their potential to co-deliver drugs in a cell-type-targeting manner. However, depending on their surface characteristics, NC accumulate serum factors, termed protein corona, which may affect their cellular binding. We have previously shown that NC coated with carbohydrates to enable biocompatibility triggered the lectin-dependent complement pathway, resulting in enhanced binding to B cells via complement receptor (CR)1/2. Here we show that such NC also engaged all types of splenic leukocytes known to express CR3 at a high rate when NC were pre-incubated with native mouse serum resulting in complement opsonization. By focusing on dendritic cells (DC) as an important antigen-presenting cell type, we show that CR3 was essential for binding/uptake of complement-opsonized NC, whereas CR4, which in mouse is specifically expressed by DC, played no role. Further, a minor B cell subpopulation (B-1), which is important for first-line pathogen responses, and co-expressed CR1/2 and CR3, in general, engaged NC to a much higher extent than normal B cells. Here, we identified CR-1/2 as necessary for binding of complement-opsonized NC, whereas CR3 was dispensable. Interestingly, the binding of complement-opsonized NC to both DC and B-1 cells affected the expression of activation markers. Our findings may have important implications for the design of nano-vaccines against infectious diseases, which codeliver pathogen-specific protein antigen and adjuvant, aimed to induce a broad adaptive cellular and humoral immune response by inducing cytotoxic T lymphocytes that kill infected cells and pathogen-neutralizing antibodies, respectively. Decoration of nano-vaccines either with carbohydrates to trigger complement activation in vivo or with active complement may result in concomitant targeting of DC and B cells and thereby may strongly enhance the extent of dual cellular/humoral immune responses.

[1]  G. Alter,et al.  Tissues: the unexplored frontier of antibody mediated immunity. , 2021, Current opinion in virology.

[2]  H. Hammad,et al.  CCR2- and Flt3-Dependent Inflammatory Conventional Type 2 Dendritic Cells Are Necessary for the Induction of Adaptive Immunity by the Human Vaccine Adjuvant System AS01 , 2021, Frontiers in Immunology.

[3]  E. Rowan,et al.  Recent advances in therapeutic applications of neutralizing antibodies for virus infections: an overview , 2020, Immunologic Research.

[4]  J. Golay,et al.  The Role of Complement in the Mechanism of Action of Therapeutic Anti-Cancer mAbs , 2020, Antibodies.

[5]  T. Mutis,et al.  Fc-Engineered Antibodies with Enhanced Fc-Effector Function for the Treatment of B-Cell Malignancies , 2020, Cancers.

[6]  M. Bros,et al.  Role of Liver-Mediated Tolerance in Nanoparticle-Based Tumor Therapy , 2020, Cells.

[7]  Joshua A. Jackman,et al.  Cloaking Silica Nanoparticles with Functional Protein Coatings for Reduced Complement Activation and Cellular Uptake. , 2020, ACS nano.

[8]  Xiqun Jiang,et al.  Antibody and antibody fragments for cancer immunotherapy. , 2020, Journal of controlled release : official journal of the Controlled Release Society.

[9]  Zhaowu Ma,et al.  Dendritic cell biology and its role in tumor immunotherapy , 2020, Journal of Hematology & Oncology.

[10]  Xun Sun,et al.  Engineering nanoparticulate vaccines for enhancing antigen cross-presentation. , 2020, Current opinion in biotechnology.

[11]  D. Dudziak,et al.  Harnessing the Complete Repertoire of Conventional Dendritic Cell Functions for Cancer Immunotherapy , 2020, Pharmaceutics.

[12]  Ping Wang,et al.  Therapeutic Potential of B-1a Cells in COVID-19. , 2020, Shock.

[13]  M. I. Tadros,et al.  Eudragit®-S100 Coated PLGA Nanoparticles for Colon Targeting of Etoricoxib: Optimization and Pharmacokinetic Assessments in Healthy Human Volunteers , 2020, International journal of nanomedicine.

[14]  S. Acton,et al.  Molecular mechanisms of dendritic cell migration in immunity and cancer , 2020, Medical Microbiology and Immunology.

[15]  Li Zhao,et al.  Polyglycerol Grafting Shields Nanoparticle from Protein Corona Formation to Avoid Macrophage Uptake. , 2020, ACS nano.

[16]  Sharan Bobbala,et al.  Surface chemistry-mediated modulation of adsorbed albumin folding state specifies nanocarrier clearance by distinct macrophage subsets , 2020, Nature Communications.

[17]  Zhaolin Hua,et al.  The role of B cell antigen presentation in the initiation of CD4+ T cell response , 2020, Immunological reviews.

[18]  W. Chan,et al.  An analysis of the binding function and structural organization of the protein corona. , 2020, Journal of the American Chemical Society.

[19]  M. Jaafari,et al.  Liposome Circulation Time is Prolonged by CD47 Coating. , 2020, Protein and peptide letters.

[20]  B. Wines,et al.  Harnessing the immune system via FcγR function in immune therapy: a pathway to next‐gen mAbs , 2020, Immunology and cell biology.

[21]  M. Bros,et al.  β2 Integrins—Multi-Functional Leukocyte Receptors in Health and Disease , 2020, International journal of molecular sciences.

[22]  A. Erdei,et al.  Utilization of complement receptors in immune cell - microbe interaction. , 2020, FEBS letters.

[23]  M. Yoshimoto The ontogeny of murine B-1a cells , 2019, International Journal of Hematology.

[24]  M. Maes,et al.  Emerging role of innate B1 cells in the pathophysiology of autoimmune and neuroimmune diseases: association with inflammation, oxidative and nitrosative stress and autoimmune responses. , 2019, Pharmacological research.

[25]  G. Bardi,et al.  Natural Polysaccharide Nanomaterials: An Overview of Their Immunological Properties , 2019, International journal of molecular sciences.

[26]  E. Groman,et al.  Feraheme (Ferumoxytol) Is Recognized by Proinflammatory and Anti-inflammatory Macrophages via Scavenger Receptor Type AI/II. , 2019, Molecular pharmaceutics.

[27]  Yongkui Zhang,et al.  Protein corona of metal-organic framework nanoparticals: Study on the adsorption behavior of protein and cell interaction. , 2019, International journal of biological macromolecules.

[28]  N. Fazilleau,et al.  Regulation of B cell responses by distinct populations of CD4 T cells , 2019, Biomedical journal.

[29]  R. Noubade,et al.  Beyond cDC1: Emerging Roles of DC Crosstalk in Cancer Immunity , 2019, Front. Immunol..

[30]  Hans-Ulrich Humpf,et al.  Serum type and concentration both affect the protein-corona composition of PLGA nanoparticles , 2019, Beilstein journal of nanotechnology.

[31]  R. R. Novaes e Brito,et al.  B-1 cell response in immunity against parasites , 2019, Parasitology Research.

[32]  M. Pallardy,et al.  Immunotoxicity of poly (lactic-co-glycolic acid) nanoparticles: influence of surface properties on dendritic cell activation , 2019, Nanotoxicology.

[33]  H. Simon,et al.  Regulation of the innate immune system by autophagy: monocytes, macrophages, dendritic cells and antigen presentation , 2019, Cell Death & Differentiation.

[34]  N. Baumgarth,et al.  The Multifaceted B Cell Response to Influenza Virus , 2019, The Journal of Immunology.

[35]  T. Vorup-Jensen,et al.  Structural Immunology of Complement Receptors 3 and 4 , 2018, Front. Immunol..

[36]  S. Tenzer,et al.  Protein corona–mediated targeting of nanocarriers to B cells allows redirection of allergic immune responses , 2018, Journal of Allergy and Clinical Immunology.

[37]  K. Landfester,et al.  The Protein Corona as a Confounding Variable of Nanoparticle-Mediated Targeted Vaccine Delivery , 2018, Front. Immunol..

[38]  M. Andersson,et al.  Curvature-dependent effects of nanotopography on classical immune complement activation. , 2018, Acta biomaterialia.

[39]  G. Arrigoni,et al.  C1q-Mediated Complement Activation and C3 Opsonization Trigger Recognition of Stealth Poly(2-methyl-2-oxazoline)-Coated Silica Nanoparticles by Human Phagocytes. , 2018, ACS nano.

[40]  K. Dawson,et al.  Differential Recognition of Nanoparticle Protein Corona and Modified Low-Density Lipoprotein by Macrophage Receptor with Collagenous Structure. , 2018, ACS nano.

[41]  M. Kortylewski,et al.  The revival of CpG oligonucleotide-based cancer immunotherapies , 2018, Contemporary oncology.

[42]  Reinhold Förster,et al.  Mechanisms and Dynamics of T Cell-Mediated Cytotoxicity In Vivo. , 2017, Trends in immunology.

[43]  K. Toh,et al.  Molecularly Imprinted Nanogels Acquire Stealth In Situ by Cloaking Themselves with Native Dysopsonic Proteins. , 2017, Angewandte Chemie.

[44]  H. Acha‐Orbea,et al.  CD11b regulates the Treg/Th17 balance in murine arthritis via IL‐6 , 2017, European journal of immunology.

[45]  V. M. Holers,et al.  In Vitro and In Vivo Differences in Murine Third Complement Component (C3) Opsonization and Macrophage/Leukocyte Responses to Antibody-Functionalized Iron Oxide Nanoworms , 2017, Front. Immunol..

[46]  N. Hanagata CpG oligodeoxynucleotide nanomedicines for the prophylaxis or treatment of cancers, infectious diseases, and allergies , 2017, International journal of nanomedicine.

[47]  G. Ferrari,et al.  Beyond Viral Neutralization. , 2017, AIDS research and human retroviruses.

[48]  S. Seok,et al.  GM-CSF Grown Bone Marrow Derived Cells Are Composed of Phenotypically Different Dendritic Cells and Macrophages , 2016, Molecules and cells.

[49]  H. Schild,et al.  Vaccination with trifunctional nanoparticles that address CD8+ dendritic cells inhibits growth of established melanoma. , 2016, Nanomedicine.

[50]  Scott N. Mueller,et al.  CD4+ T‐cell help amplifies innate signals for primary CD8+ T‐cell immunity , 2016, Immunological reviews.

[51]  S. Kent,et al.  Human immune cell targeting of protein nanoparticles--caveospheres. , 2016, Nanoscale.

[52]  Justine D Mintern,et al.  Targeting dendritic cells: a promising strategy to improve vaccine effectiveness , 2016, Clinical & translational immunology.

[53]  J. Vishwanatha,et al.  Rationalizing the use of functionalized poly-lactic-co-glycolic acid nanoparticles for dendritic cell-based targeted anticancer therapy. , 2016, Nanomedicine.

[54]  S. Cherry,et al.  Infection-induced type I interferons activate CD11b on B-1 cells for subsequent lymph node accumulation , 2015, Nature Communications.

[55]  Guoquan Zhang,et al.  Role of B Cells in Host Defense against Primary Coxiella burnetii Infection , 2015, Infection and Immunity.

[56]  Ping Wang,et al.  The role of B-1 cells in inflammation , 2015, Immunologic research.

[57]  Lubka T. Roumenina,et al.  Complement System Part II: Role in Immunity , 2015, Front. Immunol..

[58]  C. Solans,et al.  Interactions of PLGA nanoparticles with blood components: protein adsorption, coagulation, activation of the complement system and hemolysis studies. , 2015, Nanoscale.

[59]  S. Eberhardt,et al.  Iron-based superparamagnetic nanoparticle contrast agents for MRI of infection and inflammation. , 2015, AJR. American journal of roentgenology.

[60]  M. Carroll,et al.  Follicular dendritic cells: dynamic antigen libraries , 2014, Nature Reviews Immunology.

[61]  Darren J. Martin,et al.  Cryptic epitopes of albumin determine mononuclear phagocyte system clearance of nanomaterials. , 2014, ACS nano.

[62]  Xiaoling Hu,et al.  Integrin CD11b negatively regulates BCR signalling to maintain autoreactive B cell tolerance , 2013, Nature Communications.

[63]  F. Broere,et al.  Peritoneal cavity B‐1a cells promote peripheral CD4+ T‐cell activation , 2013, European journal of immunology.

[64]  I. Tsigelny,et al.  Direct recognition of superparamagnetic nanocrystals by macrophage scavenger receptor SR-AI. , 2013, ACS nano.

[65]  J. Hamerman,et al.  β2 integrins inhibit TLR responses by regulating NF‐κB pathway and p38 MAPK activation , 2013, European journal of immunology.

[66]  J. Buyon,et al.  Complement Receptor 3 Influences Toll-like Receptor 7/8-Dependent Inflammation , 2013, The Journal of Biological Chemistry.

[67]  Junling Wang,et al.  PU.1 Is Essential for CD11c Expression in CD8+/CD8− Lymphoid and Monocyte-Derived Dendritic Cells during GM-CSF or FLT3L-Induced Differentiation , 2012, PloS one.

[68]  I. Tsigelny,et al.  Recognition of dextran-superparamagnetic iron oxide nanoparticle conjugates (Feridex) via macrophage scavenger receptor charged domains. , 2012, Bioconjugate chemistry.

[69]  T. Rothstein,et al.  A small CD11b+ human B1 cell subpopulation stimulates T cells and is expanded in lupus , 2011, The Journal of experimental medicine.

[70]  A. Salomon,et al.  Lectin Site Ligation of CR3 Induces Conformational Changes and Signaling* , 2011, The Journal of Biological Chemistry.

[71]  Nan Li,et al.  Integrin CD11b negatively regulates TLR-triggered inflammatory responses by activating Syk and promoting degradation of MyD88 and TRIF via Cbl-b , 2010, Nature Immunology.

[72]  R. Ludwig Therapeutic use of heparin beyond anticoagulation. , 2009, Current drug discovery technologies.

[73]  J. Prechl,et al.  Expression and role of CR1 and CR2 on B and T lymphocytes under physiological and autoimmune conditions. , 2009, Molecular immunology.

[74]  Heather N. Hayenga,et al.  Differential Regulation of Neutrophil CD18 Integrin Function by Di- and Tri-Valent Cations: Manganese vs. Gadolinium , 2008, Annals of Biomedical Engineering.

[75]  R. Roozendaal,et al.  Complement receptors CD21 and CD35 in humoral immunity , 2007, Immunological reviews.

[76]  C. Bode,et al.  Superparamagnetic iron oxide binding and uptake as imaged by magnetic resonance is mediated by the integrin receptor Mac-1 (CD11b/CD18): implications on imaging of atherosclerotic plaques. , 2007, Atherosclerosis.

[77]  Debra K. Shivers,et al.  Complement Receptor 3 Ligation of Dendritic Cells Suppresses Their Stimulatory Capacity1 , 2007, The Journal of Immunology.

[78]  D. Vestweber,et al.  Active MAC-1 (CD11b/CD18) on DCs inhibits full T-cell activation. , 2007, Blood.

[79]  C. Vauthier,et al.  Complement Activation by Core–Shell Poly(isobutylcyanoacrylate)–Polysaccharide Nanoparticles: Influences of Surface Morphology, Length, and Type of Polysaccharide , 2006, Pharmaceutical Research.

[80]  Simon C Watkins,et al.  Internalization of circulating apoptotic cells by splenic marginal zone dendritic cells: dependence on complement receptors and effect on cytokine production. , 2003, Blood.

[81]  L. Schwarzfischer,et al.  Comparative analysis of integrin expression on monocyte‐derived macrophages and monocyte‐derived dendritic cells , 2000, Immunology.

[82]  G. Kelsoe,et al.  Antibody response to a T-dependent antigen requires B cell expression of complement receptors , 1996, The Journal of experimental medicine.

[83]  N. Cooper,et al.  Modulation of signaling via the B cell antigen receptor by CD21, the receptor for C3dg and EBV. , 1994, Journal of immunology.

[84]  V. Vetvicka,et al.  CR3 (CD11b, CD18): a phagocyte and NK cell membrane receptor with multiple ligand specificities and functions , 1993, Clinical and experimental immunology.

[85]  D. Golenbock,et al.  Lipopolysaccharide-induced stimulation of CD11b/CD18 expression on neutrophils. Evidence of specific receptor-based response and inhibition by lipid A-based antagonists. , 1991, Journal of immunology.

[86]  R. Steinman,et al.  The distinct leukocyte integrins of mouse spleen dendritic cells as identified with new hamster monoclonal antibodies , 1990, The Journal of experimental medicine.

[87]  Jamal S. Lewis,et al.  Macrophage integrins modulate response to ultra-high molecular weight polyethylene particles and direct particle-induced osteolysis. , 2017, Biomaterials.

[88]  Meredith O'Keeffe,et al.  Monitoring dendritic cell activation and maturation. , 2013, Methods in molecular biology.

[89]  Ying S. Chao,et al.  Role of carbohydrate receptors in the macrophage uptake of dextran-coated iron oxide nanoparticles. , 2012, Advances in experimental medicine and biology.

[90]  Istvan Toth,et al.  Nanoparticle-induced unfolding of fibrinogen promotes Mac-1 receptor activation and inflammation. , 2011, Nature nanotechnology.

[91]  M. Büchler,et al.  Release of iC3b from apoptotic tumor cells induces tolerance by binding to immature dendritic cells in vitro and in vivo , 2005, Cancer Immunology, Immunotherapy.