Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 130
1.
J Immunol ; 212(11): 1680-1692, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38607278

Plasmacytoid dendritic cells (pDCs) are strongly implicated as a major source of IFN-I in systemic lupus erythematosus (SLE), triggered through TLR-mediated recognition of nucleic acids released from dying cells. However, relatively little is known about how TLR signaling and IFN-I production are regulated in pDCs. In this article, we describe a role for integrin αvß3 in regulating TLR responses and IFN-I production by pDCs in mouse models. We show that αv and ß3-knockout pDCs produce more IFN-I and inflammatory cytokines than controls when stimulated through TLR7 and TLR9 in vitro and in vivo. Increased cytokine production was associated with delayed acidification of endosomes containing TLR ligands, reduced LC3 conjugation, and increased TLR signaling. This dysregulated TLR signaling results in activation of B cells and promotes germinal center (GC) B cell and plasma cell expansion. Furthermore, in a mouse model of TLR7-driven lupus-like disease, deletion of αvß3 from pDCs causes accelerated autoantibody production and pathology. We therefore identify a pDC-intrinsic role for αvß3 in regulating TLR signaling and preventing activation of autoreactive B cells. Because αvß3 serves as a receptor for apoptotic cells and cell debris, we hypothesize that this regulatory mechanism provides important contextual cues to pDCs and functions to limit responses to self-derived nucleic acids.


Autoimmunity , Dendritic Cells , Integrin alphaVbeta3 , Lupus Erythematosus, Systemic , Mice, Knockout , Signal Transduction , Toll-Like Receptor 7 , Animals , Mice , Dendritic Cells/immunology , Integrin alphaVbeta3/immunology , Integrin alphaVbeta3/metabolism , Autoimmunity/immunology , Toll-Like Receptor 7/immunology , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 7/genetics , Lupus Erythematosus, Systemic/immunology , Signal Transduction/immunology , Mice, Inbred C57BL , Cytokines/metabolism , Cytokines/immunology , Toll-Like Receptor 9/immunology , Toll-Like Receptor 9/metabolism , B-Lymphocytes/immunology , Autoantibodies/immunology , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Lymphocyte Activation/immunology , Disease Models, Animal
2.
Angew Chem Int Ed Engl ; 61(10): e202113649, 2022 03 01.
Article En | MEDLINE | ID: mdl-34994999

Precise and effective manipulation of protein functions still faces tremendous challenges. Herein we report a programmable peptide molecule, consisted of targeting and self-assembly modules, that enables specific and highly efficient assembly governed by targeting receptor proteins. Upon binding to the cell membrane receptor, peptide conformation is somewhat stabilized along with decreased self-assembly activation energy, promoting peptide-protein complex oligomerization. We first design a GNNQQNY-RGD peptide (G7-RGD) to recognize integrin αV ß3 receptor for proof-of-concept study. In the presence of αV ß3 protein, the critical assembly concentration of free G7-RGD decreases from 525 to 33 µM and the resultant G7-RGD cluster drives integrin receptor oligomerization. Finally, a bispecific assembling peptide antiCD3-G7-RGD is rationally designed for cancer immunotherapy, which validates CD3 oligomerization and concomitant T cell activation, leading to T cell-mediated cancer cell cytolysis.


Immunotherapy , Integrin alphaVbeta3/analysis , Neoplasms/therapy , Peptides/chemistry , Humans , Integrin alphaVbeta3/immunology , Neoplasms/immunology , Peptides/immunology
3.
J Biol Chem ; 296: 100399, 2021.
Article En | MEDLINE | ID: mdl-33571526

CD40L is a member of the TNF superfamily that participates in immune cell activation. It binds to and signals through several integrins, including αvß3 and α5ß1, which bind to the trimeric interface of CD40L. We previously showed that several integrin ligands can bind to the allosteric site (site 2), which is distinct from the classical ligand-binding site (site 1), raising the question of if CD40L activates integrins. In our explorations of this question, we determined that integrin α4ß1, which is prevalently expressed on the same CD4+ T cells as CD40L, is another receptor for CD40L. Soluble (s)CD40L activated soluble integrins αvß3, α5ß1, and α4ß1 in cell-free conditions, indicating that this activation does not require inside-out signaling. Moreover, sCD40L activated cell-surface integrins in CHO cells that do not express CD40. To learn more about the mechanism of binding, we determined that sCD40L bound to a cyclic peptide from site 2. Docking simulations predicted that the residues of CD40L that bind to site 2 are located outside of the CD40L trimer interface, at a site where four HIGM1 (hyper-IgM syndrome type 1) mutations are clustered. We tested the effect of these mutations, finding that the K143T and G144E mutants were the most defective in integrin activation, providing support that this region interacts with site 2. We propose that allosteric integrin activation by CD40L also plays a role in CD40L signaling, and defective site 2 binding may be related to the impaired CD40L signaling functions of these HIGM1 mutants.


CD40 Ligand/metabolism , Integrin alpha4beta1/metabolism , Integrin alpha5beta1/metabolism , Integrin alphaVbeta3/metabolism , Receptors, Cell Surface/chemistry , T-Lymphocytes/metabolism , Allosteric Site , Animals , CD40 Ligand/immunology , Cell Line , Cricetinae , Humans , Integrin alpha4beta1/immunology , Integrin alpha5beta1/immunology , Integrin alphaVbeta3/immunology , Molecular Docking Simulation , Protein Binding , Receptors, Cell Surface/metabolism , Signal Transduction , T-Lymphocytes/immunology
4.
Mol Pharm ; 17(10): 3740-3747, 2020 10 05.
Article En | MEDLINE | ID: mdl-32845640

Cyclic RGD (cRGD) peptide-conjugated boronated albumin was developed to direct toward integrin αvß3, which overexpresses on many cancer cells. A stepwise conjugation of c[RGDfK(Mal)] and maleimide-conjugated closo-dodecaborate (MID) to bovine serum albumin (BSA) afforded cRGD-MID-BSA, which was noncytotoxic toward both U87MG and A549 cells. As compared with l-BPA, selective antitumor activity of cRGD-MID-BSA toward U87MG cells overexpressing integrin αvß3 was identified after thermal neutron irradiation. In vivo fluorescence live imaging of Cy5-conjugated cRGD-MID-BSA and MID-BSA revealed that both cRGD-MID-BSA and MID-BSA similarly reached the maximum accumulation during 8-12 h after injection. The selective accumulation and retention of Cy5-cRGD-MID-BSA was more pronounced than Cy5-MID-BSA after 24 h. An in vivo boron neutron capture therapy (BNCT) study revealed that the cRGD peptide ligand combination enhanced accumulation of MID-BSA into tumor cells in U87MG xenograft models. The significant tumor growth suppression was observed in U87MG xenograft models at a dose of 7.5 mg [10B]/kg after neutron irradiation.


Boron Neutron Capture Therapy/methods , Boron/administration & dosage , Drug Carriers/chemistry , Integrin alphaVbeta3/metabolism , Isotopes/administration & dosage , Neoplasms/radiotherapy , Animals , Boron/chemistry , Boron Compounds/administration & dosage , Boron Compounds/chemistry , Cell Line, Tumor , Female , Humans , Integrin alphaVbeta3/immunology , Intravital Microscopy , Isotopes/chemistry , Mice , Neoplasms/diagnostic imaging , Neoplasms/immunology , Neoplasms/pathology , Peptides, Cyclic/chemistry , Serum Albumin, Bovine/chemistry , Xenograft Model Antitumor Assays
5.
Nat Commun ; 11(1): 1242, 2020 03 06.
Article En | MEDLINE | ID: mdl-32144270

Expression of the matricellular protein CCN1 (CYR61) is associated with inflammation and is required for successful wound repair. Here, we show that CCN1 binds bacterial pathogen-associated molecular patterns including peptidoglycans of Gram-positive bacteria and lipopolysaccharides of Gram-negative bacteria. CCN1 opsonizes methicillin-resistant Staphylococcus aureus (MRSA) and Pseudomonas aeruginosa and accelerates their removal by phagocytosis and increased production of bactericidal reactive oxygen species in macrophages through the engagement of integrin αvß3. Mice with myeloid-specific Ccn1 deletion and knock-in mice expressing CCN1 unable to bind αvß3 are more susceptible to infection by S. aureus or P. aeruginosa, resulting in increased mortality and organ colonization. Furthermore, CCN1 binds directly to TLR2 and TLR4 to activate MyD88-dependent signaling, cytokine expression and neutrophil mobilization. CCN1 is therefore a pattern recognition receptor that opsonizes bacteria for clearance and functions as a damage-associated molecular pattern to activate inflammatory responses, activities that contribute to wound healing and tissue repair.


Cysteine-Rich Protein 61/metabolism , Opsonin Proteins/metabolism , Pseudomonas Infections/immunology , Staphylococcal Infections/immunology , Toll-Like Receptors/metabolism , Animals , Cysteine-Rich Protein 61/genetics , Cysteine-Rich Protein 61/immunology , Disease Models, Animal , Disease Susceptibility , Female , Gene Knock-In Techniques , Gene Knockdown Techniques , Humans , Integrin alphaVbeta3/immunology , Integrin alphaVbeta3/metabolism , Male , Methicillin-Resistant Staphylococcus aureus , Mice , Mice, Inbred C57BL , Mice, Transgenic , Opsonin Proteins/genetics , Pathogen-Associated Molecular Pattern Molecules/metabolism , Phagocytosis/immunology , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/immunology , Sf9 Cells , Signal Transduction/immunology , Staphylococcal Infections/microbiology , Toll-Like Receptors/immunology
6.
Thromb Haemost ; 119(11): 1807-1815, 2019 Nov.
Article En | MEDLINE | ID: mdl-31587244

BACKGROUND: Fetal/neonatal alloimmune thrombocytopenia (FNAIT) results from maternal alloantibodies (abs) reacting with fetal platelets expressing paternal human platelet antigens (HPAs), mostly HPA-1a. Anti-HPA-1a abs, are the most frequent cause of severe thrombocytopenia and intracranial hemorrhage (ICH). OBJECTIVES: Titration of anti-HPA-1a in maternal serum using standard National Institute for Biological Standards and Control (NIBSC) 03/152 is one diagnostic approach to predict the severity of FNAIT. Recently, we found three anti-HPA-1a subtypes reacting with the ß3 subunit independently or dependently from complexes with αIIb and αv. Endothelial cell-reactive anti-αvß3 abs were found predominantly in cases with ICH. Our aim was to assess whether available standard material represents all anti-HPA-1a subtypes. MATERIALS AND METHODS: In this study, anti-HPA-1a sera (NIBSC 03/152) and human monoclonal antibodies (moabs) against HPA-1a (moabs 26.4 and 813) were evaluated using transfected cell lines expressing αIIbß3, αvß3 or monomeric cß3. RESULTS: Flow cytometry analyses with well-characterized murine moabs recognizing αIIbß3, αvß3, or ß3 alone demonstrated that AP3 reacts compound-independently, whereas compound-dependent moabs Gi5 and 23C6 reacted only with complexes. NIBSC 03/152, moabs 26.4, and 813 against HPA-1a reacted like AP3, same results were obtained with monomeric cß3 in immunoblotting. Antigen capture assay targeting endothelial cells showed anti-HPA-1a reactivity disappearance after cß3 beads adsorption. Furthermore, in contrast to anti-HPA-1a abs from ICH cases, none of NIBSC 03/152, 26.4, and 813 inhibited tube formation. CONCLUSION: These results suggest that current anti-HPA-1a standard material contains only the anti-ß3 subtype. The absence of anti-αvß3 makes NIBSC 03/152 less suitable as standard to predict the severity of FNAIT.


Antibodies, Monoclonal/immunology , Antibody Specificity , Antigens, Human Platelet/immunology , Immunologic Tests , Integrin alphaVbeta3/immunology , Integrin beta3/immunology , Isoantibodies/immunology , Platelet Glycoprotein GPIIb-IIIa Complex/immunology , Thrombocytopenia, Neonatal Alloimmune/diagnosis , Endothelial Cells/immunology , HEK293 Cells , Humans , Isoantibodies/blood , Neovascularization, Physiologic , Predictive Value of Tests , Reproducibility of Results , Severity of Illness Index , Thrombocytopenia, Neonatal Alloimmune/blood , Thrombocytopenia, Neonatal Alloimmune/immunology
7.
JCI Insight ; 4(18)2019 09 19.
Article En | MEDLINE | ID: mdl-31534047

Osteoarthritis (OA) is the leading cause of joint failure, yet the underlying mechanisms remain elusive, and no approved therapies that slow progression exist. Dysregulated integrin function was previously implicated in OA pathogenesis. However, the roles of integrin αVß3 and the integrin-associated receptor CD47 in OA remain largely unknown. Here, transcriptomic and proteomic analyses of human and murine osteoarthritic tissues revealed dysregulated expression of αVß3, CD47, and their ligands. Using genetically deficient mice and pharmacologic inhibitors, we showed that αVß3, CD47, and the downstream signaling molecules Fyn and FAK are crucial to OA pathogenesis. MicroPET/CT imaging of a mouse model showed elevated ligand-binding capacities of integrin αVß3 and CD47 in osteoarthritic joints. Further, our in vitro studies demonstrated that chondrocyte breakdown products, derived from articular cartilage of individuals with OA, induced αVß3/CD47-dependent expression of inflammatory and degradative mediators, and revealed the downstream signaling network. Our findings identify a central role for dysregulated αVß3 and CD47 signaling in OA pathogenesis and suggest that activation of αVß3 and CD47 signaling in many articular cell types contributes to inflammation and joint destruction in OA. Thus, the data presented here provide a rationale for targeting αVß3, CD47, and their signaling pathways as a disease-modifying therapy.


CD47 Antigen/metabolism , Cartilage, Articular/pathology , Integrin alphaVbeta3/metabolism , Osteoarthritis/immunology , Signal Transduction/immunology , Animals , CD47 Antigen/genetics , CD47 Antigen/immunology , Cartilage, Articular/cytology , Cartilage, Articular/diagnostic imaging , Cartilage, Articular/immunology , Cells, Cultured , Chondrocytes , Datasets as Topic , Disease Models, Animal , Disease Progression , Gene Expression Profiling , Humans , Integrin alphaVbeta3/genetics , Integrin alphaVbeta3/immunology , Male , Mice , Osteoarthritis/diagnostic imaging , Osteoarthritis/pathology , Positron Emission Tomography Computed Tomography , Primary Cell Culture , Proteomics , Synovial Membrane/cytology , Synovial Membrane/immunology , Synovial Membrane/pathology , Synoviocytes , X-Ray Microtomography
8.
Cancer Res ; 79(19): 5048-5059, 2019 10 01.
Article En | MEDLINE | ID: mdl-31416839

Tumor-associated macrophages (TAM) are highly expressed within the tumor microenvironment of a wide range of cancers, where they exert a protumor phenotype by promoting tumor cell growth and suppressing antitumor immune function. Here, we show that TAM accumulation in human and mouse tumors correlates with tumor cell expression of integrin αvß3, a known driver of epithelial cancer progression and drug resistance. A monoclonal antibody targeting αvß3 (LM609) exploited the coenrichment of αvß3 and TAMs to not only eradicate highly aggressive drug-resistant human lung and pancreas cancers in mice, but also to prevent the emergence of circulating tumor cells. Importantly, this antitumor activity in mice was eliminated following macrophage depletion. Although LM609 had no direct effect on tumor cell viability, it engaged macrophages but not natural killer (NK) cells to induce antibody-dependent cellular cytotoxicity (ADCC) of αvß3-expressing tumor cells despite their expression of the CD47 "don't eat me" signal. In contrast to strategies designed to eliminate TAMs, these findings suggest that anti-αvß3 represents a promising immunotherapeutic approach to redirect TAMs to serve as tumor killers for late-stage or drug-resistant cancers. SIGNIFICANCE: Therapeutic antibodies are commonly engineered to optimize engagement of NK cells as effectors. In contrast, LM609 targets αvß3 to suppress tumor progression and enhance drug sensitivity by exploiting TAMs to trigger ADCC.


Antibodies, Monoclonal/pharmacology , Antibody-Dependent Cell Cytotoxicity/drug effects , Integrin alphaVbeta3/immunology , Macrophages/immunology , Neoplasms, Glandular and Epithelial/immunology , Animals , Antineoplastic Agents/pharmacology , Disease Progression , Humans , Mice , Neoplasms, Glandular and Epithelial/pathology , Phagocytosis/drug effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
9.
Anal Chem ; 91(17): 11045-11054, 2019 09 03.
Article En | MEDLINE | ID: mdl-31361124

Early precise diagnosis of cancers is crucial to realize more effective therapeutic interventions with minimal toxic effects. Cancer phenotypes may also alter greatly among patients and within individuals over the therapeutic process. The identification and characterization of specific biomarkers expressed on tumor cells are in high demand for diagnosis and treatment, but they are still a challenge. Herein, we designed three new bioorthogonal surface-enhanced Raman scattering (SERS) nanoprobes and successfully applied the cocktail of them for MDA-MB-231 and MCF-7 breast cancer multiplex phenotype detection. The SERS nanoprobes containing Raman reporters with diynl, azide, or cyano moieties demonstrated apparent Raman shift peaks in 2205, 2120, and 2230 cm-1, respectively, in the biologically Raman-silent region. Three target ligands, including oligonucleotide aptamer (AS1411), arginine-glycine-aspatic acid (RGD) peptide, and homing cell adhesion molecule antibody (anti-CD44), were separately conjugated to the nanoprobes for active recognition capability. The cocktail of the nanoprobes manifested minimal cytotoxicity and simultaneously multiplex phenotype imaging of MDA-MB-231 and MCF-7 cells. Quantitative measurement of cellular uptake by inductively coupled plasma mass spectrometry (ICPMS) verified that MDA-MB-231 cells harbored a much higher expression level of CD44 receptor than MCF-7 cells. For in vivo SERS detection, Raman shift peaks of 2120, 2205, and 2230 cm-1 in the micro-tumor were clearly observed, representing the existence of three specific biomarkers of nucleolin, integrin αvß3, and CD44 reporter, which could be used for early cancer phenotype identification. The biodistribution results indicated that target ligand modified nanoprobes exhibited much more accumulation in tumors than those nanoprobes without target ligands. The multicolor cocktail of bioorthogonal SERS nanoprobes offers an attractive and insightful strategy for early cancer multiplex phenotype targeting and diagnosis in vivo that is noninvasive and has low cross-talk, unique spectral-molecular signature, high sensitivity, and negligible background interference.


Biomarkers, Tumor/genetics , Breast Neoplasms/diagnostic imaging , Immunoconjugates/chemistry , Molecular Probes/chemistry , Nanoparticles/chemistry , Spectrum Analysis, Raman/methods , Animals , Antibodies/chemistry , Aptamers, Nucleotide/chemistry , Biomarkers, Tumor/immunology , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Female , Gene Expression , Heterografts , Humans , Hyaluronan Receptors/antagonists & inhibitors , Hyaluronan Receptors/genetics , Hyaluronan Receptors/immunology , Immunoconjugates/administration & dosage , Integrin alphaVbeta3/genetics , Integrin alphaVbeta3/immunology , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Probes/administration & dosage , Nanoparticles/administration & dosage , Oligopeptides/chemistry , Phosphoproteins/genetics , Phosphoproteins/immunology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Spectrophotometry, Atomic , Nucleolin
10.
Nat Commun ; 10(1): 1482, 2019 04 01.
Article En | MEDLINE | ID: mdl-30931941

Integrins are components of cell-matrix adhesions, and function as scaffolds for various signal transduction pathways. So far no lipid ligand for integrin has been reported. Here we show that a lipid, oxysterol 25-hydroxycholesterol (25HC), directly binds to α5ß1 and αvß3 integrins to activate integrin-focal adhesion kinase (FAK) signaling. Treatment of macrophages and epithelial cells with 25HC results in an increase in activated αvß3 integrin in podosome and focal adhesion matrix adhesion sites. Moreover, activation of pattern recognition receptor on macrophages induces secretion of 25HC, triggering integrin signaling and the production of proinflammatory cytokines such as TNF and IL-6. Thus, the lipid molecule 25HC is a physiologically relevant activator of integrins and is involved in positively regulating proinflammatory responses. Our data suggest that extracellular 25HC links innate immune inflammatory response with integrin signaling.


Focal Adhesion Protein-Tyrosine Kinases/metabolism , Hydroxycholesterols/metabolism , Immunity, Innate/immunology , Integrin alpha5beta1/immunology , Integrin alphaVbeta3/immunology , Macrophages/immunology , Animals , Focal Adhesions , Inflammation , Integrin alpha5beta1/metabolism , Integrin alphaVbeta3/metabolism , Interleukin-6/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Receptors, Pattern Recognition/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/immunology
11.
J Clin Invest ; 129(4): 1713-1726, 2019 04 01.
Article En | MEDLINE | ID: mdl-30747722

Soluble urokinase receptor (suPAR) is a circulatory molecule that activates αvß3 integrin on podocytes, causes foot process effacement, and contributes to proteinuric kidney disease. While active integrin can be targeted by antibodies and small molecules, endogenous inhibitors haven't been discovered yet. Here we report what we believe is a novel renoprotective role for the inducible costimulator ligand (ICOSL) in early kidney disease through its selective binding to podocyte αvß3 integrin. Contrary to ICOSL's immune-regulatory role, ICOSL in nonhematopoietic cells limited the activation of αvß3 integrin. Specifically, ICOSL contains the arginine-glycine-aspartate (RGD) motif, which allowed for a high-affinity and selective binding to αvß3 and modulation of podocyte adhesion. This binding was largely inhibited either by a synthetic RGD peptide or by a disrupted RGD sequence in ICOSL. ICOSL binding favored the active αvß3 rather than the inactive form and showed little affinity for other integrins. Consistent with the rapid induction of podocyte ICOSL by inflammatory stimuli, glomerular ICOSL expression was increased in biopsies of early-stage human proteinuric kidney diseases. Icosl deficiency in mice resulted in an increased susceptibility to proteinuria that was rescued by recombinant ICOSL. Our work identified a potentially novel role for ICOSL, which serves as an endogenous αvß3-selective antagonist to maintain glomerular filtration.


Inducible T-Cell Co-Stimulator Ligand , Integrin alphaVbeta3 , Kidney Failure, Chronic , Podocytes , Proteinuria , Amino Acid Motifs , Animals , Glomerular Filtration Rate/drug effects , Glomerular Filtration Rate/genetics , Glomerular Filtration Rate/immunology , Humans , Inducible T-Cell Co-Stimulator Ligand/genetics , Inducible T-Cell Co-Stimulator Ligand/immunology , Inducible T-Cell Co-Stimulator Ligand/pharmacology , Integrin alphaVbeta3/antagonists & inhibitors , Integrin alphaVbeta3/genetics , Integrin alphaVbeta3/immunology , Kidney Failure, Chronic/drug therapy , Kidney Failure, Chronic/genetics , Kidney Failure, Chronic/immunology , Kidney Failure, Chronic/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Podocytes/immunology , Podocytes/pathology , Proteinuria/drug therapy , Proteinuria/genetics , Proteinuria/immunology , Proteinuria/pathology
12.
Adv Healthc Mater ; 8(4): e1801234, 2019 02.
Article En | MEDLINE | ID: mdl-30537061

Macrophages are a predominant immune cell population that drive inflammatory responses and exhibit transitions in phenotype and function during tissue remodeling in disease and repair. Thus, engineering an immunomodulatory biomaterial has significant implications for resolving inflammation. Here, a biomimetic and photoresponsive hyaluronan (HA) hydrogel nanocomposite with tunable 3D extracellular matrix (ECM) adhesion sites for dynamic macrophage immunomodulation is engineered. Photodegradative alkoxylphenacyl-based polycarbonate (APP) nanocomposites are exploited to permit user-controlled Arg-Gly-Asp (RGD) adhesive peptide release and conjugation to a HA-based ECM for real-time integrin activation of macrophages encapsulated in 3D HA-APP nanocomposite hydrogels. It is demonstrated that photocontrolled 3D ECM-RGD peptide conjugation can activate αvß3 integrin of macrophages, and periodic αvß3 integrin activation can enhance anti-inflammatory M2 macrophage polarization. Altogether, an emerging use of biomimetic, photoresponsive, and bioactive HA-APP nanocomposite hydrogel is highlighted to command 3D cell-ECM interactions for modulating macrophage polarization, which may shed light on cell-ECM interactions in innate immunity and inspire new biomaterial-based immunomodulatory therapies.


Hyaluronic Acid , Hydrogels , Immunomodulation , Macrophages/immunology , Nanocomposites/chemistry , Ultraviolet Rays , Animals , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Hydrogels/chemistry , Hydrogels/pharmacology , Immunomodulation/drug effects , Immunomodulation/radiation effects , Integrin alphaVbeta3/immunology , Mice , Oligopeptides/chemistry , Oligopeptides/pharmacology , RAW 264.7 Cells
13.
J Thromb Haemost ; 16(9): 1843-1856, 2018 09.
Article En | MEDLINE | ID: mdl-29953749

Essentials The pathogenesis of immune thrombocytopenia (ITP) has not been fully clarified. We analyzed the role of anti-αvß3 autoantibody in the pathogenesis of ITP in patients. Anti-αvß3 autoantibody impeded megakaryocyte migration and adhesion to the vascular niche. Anti-αv ß3 autoantibody potentially contributes to the pathogenesis of refractory ITP. SUMMARY: Background The pathogenesis of immune thrombocytopenia (ITP) has not been fully clarified. Anti-αvß3 integrin autoantibody is detected in chronic ITP patients, but its contribution to ITP is still unclear. Objectives To clarify the potential role of anti-αvß3 integrin autoantibody in chronic ITP and the related mechanism. Methods Relationship between levels of anti-αvß3 autoantibody and platelets in chronic ITP patients was evaluated. The influence of anti-αvß3 antibody on megakaryocyte (MK) survival, differentiation, migration and adhesion was assessed, and the associated signal pathways were investigated. Platelet recovery and MKs' distribution were observed in an ITP mouse model pretreated with different antibodies. Result In this study, we showed that the anti-αvß3 autoantibody usually coexists with anti-αIIbß3 autoantibody in chronic ITP patients, and patients with both autoantibodies have lower platelets. In in vitro studies, we showed that the anti-αvß3 antibody had no significant effect on the survival and proliferation of MKs, whereas it decreased formations of proplatelet significantly. Anti-αvß3 antibody impeded stromal cell derived facor-1 alpha (SDF-1α)- mediated migration and inhibited the phosphorylation of protein kinase B. Anti-αvß3 antibody significantly inhibited MKs' adhesion to endothelial cells and Fibrogen. The phosphorylation of focal adhesion kinase and proto-oncogene tyrosine-protein kinase Src induced by adhesion was inhibited when MKs were pretreated with anti-αvß3 antibody. In in vivo studies, we showed that injection with anti-αv antibody delayed platelet recovery in a mouse model of ITP. Conclusions These findings demonstrate that the autoantibody against integrin αv ß3 may aggravate thrombocytopenia in ITP patients by impeding MK migration and adhesion to the vascular niche, which provides new insights into the pathogenesis of ITP.


Autoantibodies/immunology , Autoantigens/immunology , Integrin alphaVbeta3/immunology , Megakaryocytes/immunology , Purpura, Thrombocytopenic, Idiopathic/immunology , Adolescent , Adult , Aged , Animals , Cell Adhesion , Cell Movement , Cells, Cultured , Chemokine CXCL12/metabolism , Endothelial Cells/metabolism , Female , Fetal Blood/cytology , Humans , Male , Megakaryocytes/cytology , Mice , Mice, Inbred BALB C , Middle Aged , Phosphorylation , Platelet Count , Platelet Membrane Glycoprotein IIb/immunology , Protein Kinases/metabolism , Protein Processing, Post-Translational , Proto-Oncogene Mas , Proto-Oncogene Proteins c-akt/metabolism , Purpura, Thrombocytopenic, Idiopathic/blood , Stromal Cells/metabolism , Thrombopoiesis , Young Adult
14.
Front Immunol ; 9: 3118, 2018.
Article En | MEDLINE | ID: mdl-30687322

Intestinal macrophages are highly mobile cells with extraordinary plasticity and actively contribute to cytokine-mediated epithelial cell damage. The mechanisms triggering macrophage polarization into a proinflammatory phenotype are unknown. Here, we report that during inflammation macrophages enhance its intercellular adhesion properties in order to acquire a M1-phenotype. Using in vitro and in vivo models we demonstrate that intercellular adhesion is mediated by integrin-αVß3 and relies in the presence of the unconventional class I myosin 1F (Myo1F). Intercellular adhesion mediated by αVß3 stimulates M1-like phenotype in macrophages through hyperactivation of STAT1 and STAT3 downstream of ILK/Akt/mTOR signaling. Inhibition of integrin-αVß3, Akt/mTOR, or lack of Myo1F attenuated the commitment of macrophages into a pro-inflammatory phenotype. In a model of colitis, Myo1F deficiency strongly reduces the secretion of proinflammatory cytokines, decreases epithelial damage, ameliorates disease activity, and enhances tissue repair. Together our findings uncover an unknown role for Myo1F as part of the machinery that regulates intercellular adhesion and polarization in macrophages.


Colitis, Ulcerative/immunology , Integrin alphaVbeta3/metabolism , Macrophage Activation , Macrophages/immunology , Myosin Type I/metabolism , Animals , Cell Line, Tumor , Colitis, Ulcerative/chemically induced , Cytoskeleton/immunology , Cytoskeleton/metabolism , Dextran Sulfate/administration & dosage , Dextran Sulfate/toxicity , Disease Models, Animal , Humans , Integrin alphaVbeta3/immunology , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myosin Type I/genetics , Myosin Type I/immunology , Primary Cell Culture , RAW 264.7 Cells
15.
Structure ; 25(11): 1732-1739.e5, 2017 11 07.
Article En | MEDLINE | ID: mdl-29033288

The LM609 antibody specifically recognizes αVß3 integrin and inhibits angiogenesis, bone resorption, and viral infections in an arginine-glycine-aspartate-independent manner. LM609 entered phase II clinical trials for the treatment of several cancers and was also used for αVß3-targeted radioimmunotherapy. To elucidate the mechanisms of recognition and inhibition of αVß3 integrin, we solved the structure of the LM609 antigen-binding fragment by X-ray crystallography and determined its binding affinity for αVß3. Using single-particle electron microscopy, we show that LM609 binds at the interface between the ß-propeller domain of the αV chain and the ßI domain of the ß3 chain, near the RGD-binding site, of all observed integrin conformational states. Integrating these data with fluorescence size-exclusion chromatography, we demonstrate that LM609 sterically hinders access of large ligands to the RGD-binding pocket, without obstructing it. This work provides a structural framework to expedite future efforts utilizing LM609 as a diagnostic or therapeutic tool.


Antibodies, Monoclonal/chemistry , Antigens/chemistry , Immunoglobulin Fab Fragments/chemistry , Integrin alphaVbeta3/chemistry , Oligopeptides/chemistry , Amino Acid Motifs , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/immunology , Angiogenesis Inhibitors/metabolism , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antigens/genetics , Antigens/immunology , Antiviral Agents/chemistry , Antiviral Agents/immunology , Antiviral Agents/metabolism , Binding Sites , Bone Density Conservation Agents/chemistry , Bone Density Conservation Agents/immunology , Bone Density Conservation Agents/metabolism , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/immunology , Integrin alphaVbeta3/genetics , Integrin alphaVbeta3/immunology , Ligands , Models, Molecular , Oligopeptides/genetics , Oligopeptides/immunology , Protein Binding , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology
16.
J Immunol ; 199(3): 982-991, 2017 08 01.
Article En | MEDLINE | ID: mdl-28646039

In tuberculosis (TB), the innate inflammatory immune response drives tissue destruction, morbidity, and mortality. Monocytes secrete matrix metalloproteinases (MMPs), which have key roles in local tissue destruction and cavitation. We hypothesized that integrin signaling might regulate monocyte MMP secretion in pulmonary TB during cell adhesion to the extracellular matrix (ECM). Adhesion to type I collagen and fibronectin by Mycobacterium tuberculosis-stimulated monocytes increased MMP-1 gene expression by 2.6-fold and 4.3-fold respectively, and secretion by 60% (from 1208.1 ± 186 to 1934.4 ± 135 pg/ml; p < 0.0001) and 63% (1970.3 ± 95 pg/ml; p < 0.001). MMP-10 secretion increased by 90% with binding to type I collagen and 55% with fibronectin, whereas MMP-7 increased 57% with collagen. The ECM did not affect the secretion of tissue inhibitors of metalloproteinases-1 or -2. Integrin αVß3 surface expression was specifically upregulated in stimulated monocytes and was further increased after adhesion to type I collagen. Binding of either ß3 or αV integrin subunits increased MMP-1/10 secretion in M. tuberculosis-stimulated monocytes. In a cohort of TB patients, significantly increased integrin ß3 mRNA accumulation in induced sputum was detected, to our knowledge, for the first time, compared with control subjects (p < 0.05). Integrin αVß3 colocalized with areas of increased and functionally active MMP-1 on infected monocytes, and αVß3 blockade markedly decreased type I collagen breakdown, and impaired both monocyte adhesion and leukocyte migration in a transwell system (p < 0.0001). In summary, our data demonstrate that M. tuberculosis stimulation upregulates integrin αVß3 expression on monocytes, which upregulates secretion of MMP-1 and -10 on adhesion to the ECM. This leads to increased monocyte recruitment and collagenase activity, which will drive inflammatory tissue damage.


Cell Adhesion , Cell Movement , Extracellular Matrix/metabolism , Integrin alphaVbeta3/genetics , Monocytes/immunology , Mycobacterium tuberculosis/immunology , Collagen Type I/metabolism , Collagenases/metabolism , Extracellular Matrix/drug effects , Fibronectins/metabolism , Gene Expression Regulation , Humans , Integrin alphaVbeta3/antagonists & inhibitors , Integrin alphaVbeta3/immunology , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 10/immunology , Matrix Metalloproteinase 10/metabolism , Matrix Metalloproteinase 7/immunology , Matrix Metalloproteinase 7/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Monocytes/microbiology , Monocytes/physiology , Signal Transduction , Sputum/chemistry , Up-Regulation
17.
Atherosclerosis ; 258: 40-50, 2017 03.
Article En | MEDLINE | ID: mdl-28189040

BACKGROUND AND AIMS: Diabetes is a major risk factor for the development of atherosclerosis. Hyperglycemia stimulates vascular smooth muscle cells (VSMC) to secrete ligands that bind to the αVß3 integrin, a receptor that regulates VSMC proliferation and migration. This study determined whether an antibody that had previously been shown to block αVß3 activation and to inhibit VSMC proliferation and migration in vitro, inhibited the development of atherosclerosis in diabetic pigs. METHODS: Twenty diabetic pigs were maintained on a high fat diet for 22 weeks. Ten received injections of anti-ß3 F(ab)2 and ten received control F(ab)2 for 18 weeks. RESULTS: The active antibody group showed reduction of atherosclerosis of 91 ± 9% in the left main, 71 ± 11%, in left anterior descending, 80 ± 10.2% in circumflex, and 76 ± 25% in right coronary artery, (p < 0.01 compared to lesions areas from corresponding control treated arteries). There were significant reductions in both cell number and extracellular matrix. Histologic analysis showed neointimal hyperplasia with macrophage infiltration, calcifications and cholesterol clefts. Antibody treatment significantly reduced number of macrophages contained within lesions, suggesting that this change contributed to the decrease in lesion cellularity. Analysis of the biochemical changes within the femoral arteries that received the active antibody showed a 46 ± 12% (p < 0.05) reduction in the tyrosine phosphorylation of the ß3 subunit of αVß3 and a 40 ± 14% (p < 0.05) reduction in MAP kinase activation. CONCLUSIONS: Blocking ligand binding to the αVß3 integrin inhibits its activation and attenuates increased VSMC proliferation that is induced by chronic hyperglycemia. These changes result in significant decreases in atherosclerotic lesion size in the coronary arteries. The results suggest that this approach may have efficacy in treating the proliferative phase of atherosclerosis in patients with diabetes.


Coronary Artery Disease/prevention & control , Diabetes Mellitus, Experimental/drug therapy , Diabetic Angiopathies/prevention & control , Immunoglobulin Fab Fragments/pharmacology , Integrin alphaVbeta3/antagonists & inhibitors , Muscle, Smooth, Vascular/drug effects , Animals , Cell Proliferation/drug effects , Coronary Artery Disease/etiology , Coronary Artery Disease/metabolism , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Coronary Vessels/pathology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Diabetic Angiopathies/etiology , Diabetic Angiopathies/metabolism , Femoral Artery/drug effects , Femoral Artery/metabolism , Femoral Artery/pathology , Immunoglobulin Fab Fragments/administration & dosage , Injections, Subcutaneous , Integrin alphaVbeta3/immunology , Integrin alphaVbeta3/metabolism , Ligands , Macrophages/drug effects , Macrophages/metabolism , Male , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Neointima , Phosphorylation , Plaque, Atherosclerotic , Protein Binding , Sus scrofa
18.
Biopolymers ; 108(1)2017 Jan.
Article En | MEDLINE | ID: mdl-27603160

Peptide-conjugated polysaccharide matrices using bioactive laminin-derived peptides are useful biomaterials for tissue and cell engineering. Here, we demonstrate an easy handling preparation method for peptide-polysaccharide matrices using polyion complex with both alginate and chitosan. First, aldehyde-alginate was synthesized by oxidization of alginate using NaIO4 , and then, reacted with Cys-peptides. Next, the peptide-alginate solution was added to a chitosan-coated plate, and the peptide-polyion complex matrices (peptide-PCMs) were prepared. The peptide-PCMs using an integrin αvß3-binding peptide (A99a: ALRGDN, mouse laminin α1 chain 1145-1150) and an integrin α2ß1-binding peptide (EF1XmR: RLQLQEGRLHFXFD, X = Nle, mouse laminin α1 chain 2751-2763) showed strong cell attachment activity in a dose-dependent manner. When we examined the effect of various spacers on the biological activity of A99a-PCM, hydrophobic and long spacers enhanced the cell attachment activity. Further, the A99a-PCM with the spacers strongly promoted neurite outgrowth. The polyion complex method is an easy way to obtain insolubilized matrix and is widely applicable for various polysaccharides. The peptide-PCM is useful as a biomaterial for cell and tissue engineering.


Alginates/chemistry , Chitosan/chemistry , Peptides/chemistry , Aldehydes/chemistry , Amino Acid Sequence , Animals , Antibodies/chemistry , Antibodies/immunology , Cell Adhesion/drug effects , Cell Line , Edetic Acid/chemistry , Heparin/chemistry , Humans , Integrin alpha1beta1/chemistry , Integrin alpha1beta1/immunology , Integrin alphaVbeta3/chemistry , Integrin alphaVbeta3/immunology , Laminin/chemistry , Mice , Microscopy, Fluorescence , Neurites/metabolism , Oxidation-Reduction , Peptides/chemical synthesis , Peptides/metabolism , Peptides/pharmacology
19.
Adv Exp Med Biol ; 925: 103-115, 2017.
Article En | MEDLINE | ID: mdl-27864802

Secreted phospholipase A2 type IIA (sPLA2-IIA) is a well-established pro-inflammatory protein and has been a major target for drug discovery. However, the mechanism of its signaling action has not been fully understood. We previously found that sPLA2-IIA binds to integrins αvß3 and α4ß1 in human and that this interaction plays a role in sPLA2-IIA's signaling action. Our recent studies found that sPLA2-IIA activates integrins in an allosteric manner through direct binding to a newly identified binding site of integrins (site 2), which is distinct from the classical RGD-binding site (site 1). The sPLA2-IIA-induced integrin activation may be related to the signaling action of sPLA2-IIA. Since sPLA2-IIA is present in normal human tears in addition to rheumatoid synovial fluid at high concentrations the sPLA2-IIA-mediated integrin activation on leukocytes may be involved in immune responses in normal and pathological conditions.


Group II Phospholipases A2/chemistry , Integrin alpha4beta1/chemistry , Integrin alphaVbeta3/chemistry , Signal Transduction/immunology , Allosteric Regulation , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Binding Sites , Gene Expression Regulation , Group II Phospholipases A2/genetics , Group II Phospholipases A2/immunology , Humans , Integrin alpha4beta1/genetics , Integrin alpha4beta1/immunology , Integrin alphaVbeta3/genetics , Integrin alphaVbeta3/immunology , Molecular Docking Simulation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Synovial Fluid/chemistry , Synovial Fluid/immunology , Tears/chemistry , Tears/immunology
20.
Biochem J ; 474(4): 589-596, 2017 02 15.
Article En | MEDLINE | ID: mdl-27993971

Tetraspanins play important roles in normal (e.g. cell adhesion, motility, activation, and proliferation) and pathological conditions (e.g. metastasis and viral infection). Tetraspanins interact with integrins and regulate integrin functions, but the specifics of tetraspanin-integrin interactions are unclear. Using co-immunoprecipitation with integrins as a sole method to detect interaction between integrins and full-length tetraspanins, it has been proposed that the variable region (helices D and E) of the extracellular-2 (EC2) domain of tetraspanins laterally associates with a non-ligand-binding site of integrins. We describe that, using adhesion assays, the EC2 domain of CD81, CD9, and CD151 bound to integrin αvß3, and this binding was suppressed by cRGDfV, a specific inhibitor of αvß3, and antibody 7E3, which is mapped to the ligand-binding site of ß3. We also present evidence that the specificity loop of ß3 directly bound to the EC2 domains. This suggests that the EC2 domains specifically bind to the classical ligand-binding site of αvß3. αvß3 was a more effective receptor for the EC2 domains than the previously known tetraspanin receptors α3ß1, α4ß1, and α6ß1. Docking simulation predicted that the helices A and B of CD81 EC2 bind to the RGD-binding site of αvß3. Substituting Lys residues at positions 116 and 144/148 of CD81 EC2 in the predicted integrin-binding interface reduced the binding of CD81 EC2 to αvß3, consistent with the docking model. These findings suggest that, in contrast with previous models, the ligand-binding site of integrin αvß3, a new tetraspanin receptor, binds to the constant region (helices A and B) of the EC2 domain.


Integrin alphaVbeta3/chemistry , Tetraspanin 24/chemistry , Tetraspanin 28/chemistry , Tetraspanin 29/chemistry , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Binding Sites , CHO Cells , Cloning, Molecular , Cricetulus , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Integrin alphaVbeta3/genetics , Integrin alphaVbeta3/immunology , Kinetics , Molecular Docking Simulation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Tetraspanin 24/genetics , Tetraspanin 24/immunology , Tetraspanin 28/genetics , Tetraspanin 28/immunology , Tetraspanin 29/genetics , Tetraspanin 29/immunology
...