Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
1.
Commun Biol ; 5(1): 40, 2022 01 11.
Article in English | MEDLINE | ID: mdl-35017678

ABSTRACT

T cells are critically important for host defense against infections. T cell activation is specific because signal initiation requires T cell receptor (TCR) recognition of foreign antigen peptides presented by major histocompatibility complexes (pMHC) on antigen presenting cells (APCs). Recent advances reveal that the TCR acts as a mechanoreceptor, but it remains unclear how pMHC/TCR engagement generates mechanical forces that are converted to intracellular signals. Here we propose a TCR Bending Mechanosignal (TBM) model, in which local bending of the T cell membrane on the nanometer scale allows sustained contact of relatively small pMHC/TCR complexes interspersed among large surface receptors and adhesion molecules on the opposing surfaces of T cells and APCs. Localized T cell membrane bending is suggested to increase accessibility of TCR signaling domains to phosphorylation, facilitate selective recognition of agonists that form catch bonds, and reduce noise signals associated with slip bonds.


Subject(s)
Biomechanical Phenomena/physiology , Cell Membrane , Mechanoreceptors , Receptors, Antigen, T-Cell , Signal Transduction/physiology , Antigen-Presenting Cells/chemistry , Cell Membrane/chemistry , Cell Membrane/metabolism , Cells, Cultured , Histocompatibility Antigens/chemistry , Histocompatibility Antigens/metabolism , Humans , Lymphocyte Activation/physiology , Mechanoreceptors/chemistry , Mechanoreceptors/metabolism , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/chemistry , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
2.
ACS Appl Mater Interfaces ; 13(48): 56719-56724, 2021 Dec 08.
Article in English | MEDLINE | ID: mdl-34797622

ABSTRACT

Immunoregulation represents a booming field of biomaterial design. The unique physical and chemical properties of biomaterials offer tremendous opportunities for development. Each of their parameters exerts immunogenic effects at the immune system, cellular, and molecular levels. Herein, the perspective summarizes the interaction of biomaterials with immune cells and the underlying mechanisms to control immunoregulation in a top-down manner, providing solid inspiration for biomedical applications of immunologically effective biomaterials.


Subject(s)
Antigen-Presenting Cells/immunology , Biomimetic Materials/chemistry , Antigen-Presenting Cells/chemistry , Biomimetic Materials/chemical synthesis , Homeostasis/immunology , Materials Testing
3.
Int J Mol Sci ; 22(7)2021 Mar 26.
Article in English | MEDLINE | ID: mdl-33810617

ABSTRACT

The foundation of precision immunotherapy in oncology is rooted in computational biology and patient-derived sample sequencing to enrich for and target immunogenic epitopes. Discovery of these tumor-specific epitopes through tumor sequencing has revolutionized patient outcomes in many types of cancers that were previously untreatable. However, these therapeutic successes are far from universal, especially with cancers that carry high intratumoral heterogeneity such as glioblastoma (GBM). Herein, we present the technical aspects of Mannan-BAM, TLR Ligands, Anti-CD40 Antibody (MBTA) vaccine immunotherapy, an investigational therapeutic that potentially circumvents the need for in silico tumor-neoantigen enrichment. We then review the most promising GBM vaccination strategies to contextualize the MBTA vaccine. By reviewing current evidence using translational tumor models supporting MBTA vaccination, we evaluate the underlying principles that validate its clinical applicability. Finally, we showcase the translational potential of MBTA vaccination as a potential immunotherapy in GBM, along with established surgical and immunologic cancer treatment paradigms.


Subject(s)
Brain Neoplasms/immunology , Brain Neoplasms/therapy , CD40 Antigens/immunology , Glioblastoma/immunology , Glioblastoma/therapy , Immunotherapy/methods , Animals , Antigen-Presenting Cells/chemistry , Cancer Vaccines , Computational Biology , Epitopes/chemistry , Humans , Immunophenotyping , Ligands , Medical Oncology/trends , Mice , Neoplasm Metastasis , Peptides/chemistry , Treatment Outcome
4.
PLoS Comput Biol ; 17(3): e1008825, 2021 03.
Article in English | MEDLINE | ID: mdl-33684103

ABSTRACT

The activation and differentiation of T-cells are mainly directly by their co-regulatory receptors. T lymphocyte-associated protein-4 (CTLA-4) and programed cell death-1 (PD-1) are two of the most important co-regulatory receptors. Binding of PD-1 and CTLA-4 with their corresponding ligands programed cell death-ligand 1 (PD-L1) and B7 on the antigen presenting cells (APC) activates two central co-inhibitory signaling pathways to suppress T cell functions. Interestingly, recent experiments have identified a new cis-interaction between PD-L1 and B7, suggesting that a crosstalk exists between two co-inhibitory receptors and the two pairs of ligand-receptor complexes can undergo dynamic oligomerization. Inspired by these experimental evidences, we developed a coarse-grained model to characterize the assembling of an immune complex consisting of CLTA-4, B7, PD-L1 and PD-1. These four proteins and their interactions form a small network motif. The temporal dynamics and spatial pattern formation of this network was simulated by a diffusion-reaction algorithm. Our simulation method incorporates the membrane confinement of cell surface proteins and geometric arrangement of different binding interfaces between these proteins. A wide range of binding constants was tested for the interactions involved in the network. Interestingly, we show that the CTLA-4/B7 ligand-receptor complexes can first form linear oligomers, while these oligomers further align together into two-dimensional clusters. Similar phenomenon has also been observed in other systems of cell surface proteins. Our test results further indicate that both co-inhibitory signaling pathways activated by B7 and PD-L1 can be down-regulated by the new cis-interaction between these two ligands, consistent with previous experimental evidences. Finally, the simulations also suggest that the dynamic and the spatial properties of the immune complex assembly are highly determined by the energetics of molecular interactions in the network. Our study, therefore, brings new insights to the co-regulatory mechanisms of T cell activation.


Subject(s)
Antigen-Antibody Complex , Antigen-Presenting Cells , T-Lymphocytes , Antigen-Antibody Complex/chemistry , Antigen-Antibody Complex/metabolism , Antigen-Presenting Cells/chemistry , Antigen-Presenting Cells/metabolism , B7 Antigens/chemistry , B7 Antigens/metabolism , B7-H1 Antigen/chemistry , B7-H1 Antigen/metabolism , CTLA-4 Antigen/chemistry , CTLA-4 Antigen/metabolism , Computational Biology , Humans , Molecular Dynamics Simulation , Programmed Cell Death 1 Receptor/chemistry , Programmed Cell Death 1 Receptor/metabolism , Protein Binding , T-Lymphocytes/chemistry , T-Lymphocytes/metabolism
5.
Bioconjug Chem ; 31(11): 2585-2595, 2020 11 18.
Article in English | MEDLINE | ID: mdl-33151667

ABSTRACT

Immunotherapy using antigen-specific cytotoxic T lymphocytes (CTLs) has become one of the most attractive strategies for cancer treatment. For the induction of antigen-specific CTLs in vivo, the co-delivery of CpG-DNAs and antigens to the same antigen-presenting cells (APCs) is a promising strategy. In this study, we prepared conjugates consisting of 40mer of CpG-DNA (CpG40) and antigenic peptide (OVA257-264), which have the following distinctive features: (1) multiple CpG motifs in a molecule; (2) cleavage in the cytosol because of the disulfide bonding via cysteine residue between peptide and CpG-DNA; (3) conjugation designed to induce antigen presentation on MHC class I molecules. Immunization with the conjugate CpG40-C-OVA257-264 at the mouse tail base induced strong CTL activity at a very low peptide dose of 20 ng/head. It was found that the conjugates were internalized into C-type mannose receptor 1 (MRC1)-expressing cells in inguinal lymph nodes, indicating that the CpG portion in the conjugate acts as not only an adjuvant for the activation of TLR9 but also a carrier to APCs expressing MRC1. In a tumor-bearing mice model, mice immunized with CpG40-C-OVA257-264 conjugates exhibited long delays in tumor growth compared with those treated with PBS, OVA257-264 alone, or a mixture of CpG40 and OVA257-264. Therefore, CpG-C-peptide conjugates could be a new and effective platform for peptide vaccine for the treatment of cancers and infectious diseases.


Subject(s)
Antigen-Presenting Cells/immunology , CpG Islands , DNA/chemistry , Neoplasms/therapy , Peptides/chemistry , Adjuvants, Immunologic/pharmacology , Animals , Antigen-Presenting Cells/chemistry , Antigen-Presenting Cells/drug effects , Immunotherapy/methods , Mice , Ovalbumin/immunology , T-Lymphocytes, Cytotoxic/immunology
6.
J Mater Chem B ; 8(26): 5620-5626, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32538414

ABSTRACT

Cellular uptake of antigens (Ags) by antigen-presenting cells (APCs) is vital for effective functioning of the immune system. Intramuscular or subcutaneous administration of vaccine Ags alone is not sufficient to elicit optimal immune responses. Thus, adjuvants are required to induce strong immunogenicity. Here, we developed nanoparticulate adjuvants that assemble into a bilayer spherical polymersome (PSome) to promote the cellular uptake of Ags by APCs. PSomes were synthesized by using a biodegradable and biocompatible block copolymer methoxy-poly(ethylene glycol)-b-poly(d,l-lactide) to encapsulate both hydrophilic and lipophilic biomacromolecules, such as ovalbumin (OVA) as a model Ag and monophosphoryl lipid A (MPLA) as an immunostimulant. After co-encapsulation of OVA and MPLA, the PSome synthetic vehicle exhibited the sustained release of OVA in cell environments and allowed efficient delivery of cargos into APCs. The administration of PSomes loaded with OVA and MPLA induced the production of interleukin-6 and tumor necrosis factor-alpha cytokines by macrophage activation in vitro and elicited effective Ag-specific antibody responses in vivo. These findings indicate that the nano-sized PSome may serve as a potent adjuvant for vaccine delivery systems to modulate enhanced immune responses.


Subject(s)
Antigen-Presenting Cells/chemistry , Lipid A/analogs & derivatives , Nanoparticles/chemistry , Ovalbumin/chemistry , Polymers/chemistry , Animals , Antigen-Antibody Reactions , Antigen-Presenting Cells/immunology , Female , Lipid A/chemistry , Lipid A/immunology , Mice , Mice, Inbred C57BL , Molecular Structure , Ovalbumin/immunology , Particle Size , Polymers/chemical synthesis , RAW 264.7 Cells , Surface Properties
7.
Chemosphere ; 219: 1052-1060, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30558807

ABSTRACT

Fluorotelomer alcohols (FTOHs, F(CF2)nCH2CH2OH) are members of per- and polyfluoroalkyl substances (PFASs) and are increasingly used in surfactant and polymer industries. FTOHs pose hepatotoxicity, nephrotoxicity and endocrine-disrupting risks. Nevertheless, there is limited research on the immunotoxic effects of FTOHs. In this study, we examined the immunotoxicity of 8:2 FTOH (n = 8) on murine macrophage cell line RAW 264.7. The results showed that 8:2 FTOH exposure reduced cell viability in dose- and time-dependent manners, inhibited cell proliferation and caused cell cycle arrest. Exposure to 8:2 FTOH downregulated the mRNA expression of some cell cycle-related genes, including Cdk4, Ccnd1, Ccne1, and p53, but also upregulated the mRNA expression of other cell cycle related genes, including Ccna2, p21, and p27. Additionally, exposure to 8:2 FTOH under unstimulated and LPS-stimulated conditions downregulated the mRNA expression of pro-inflammatory genes, including Il1b, Il6, Cxcl1, and Tnfa, and secreted levels of IL-6 and TNF-α. Treatment with 8:2 FTOH upregulated the mRNA expression of antigen-presenting-related genes, including H2-K1, H2-Ka, Cd80, and Cd86. The abovementioned immunotoxic effects caused by 8:2 FTOH in RAW 264.7 cells were partially or completely blocked by co-treatment with hydralazine hydrochloride (Hyd), a reactive carbonyl species (RCS) scavenger. However, exposure to 8:2 FTOH did not exhibit any effects on intracellular reactive oxygen species (ROS) level with or without LPS stimulation. Taken together, these results suggest that 8:2 FTOH may have immunotoxic effects on macrophages and RCS may underlie the responsible mechanism. The present study aids in understanding the health risks caused by FTOHs.


Subject(s)
Antigen-Presenting Cells/chemistry , Cytokines/chemistry , Ethanol/chemistry , Macrophages/metabolism , Animals , Cell Proliferation , Mice
8.
Front Immunol ; 9: 2085, 2018.
Article in English | MEDLINE | ID: mdl-30279692

ABSTRACT

We created APC-mimetic synthetic substrates to study the impact of ligand clustering on T cell activation and spreading. The substrates exhibit antibodies directed against the TCR-complex in the form of a patterned array of sub micrometric dots surrounded by a fluid supported lipid bilayer (SLB) which may itself be functionalized with another bio-molecule. We show that for T cell adhesion mediated by T cell receptor (TCR) alone, in the patterned, but not in the corresponding homogeneous controls, the TCR, ZAP-70 and actin are present in the form of clusters or patches that co-localize with the ligand-dots. However, global cell scale parameters like cell area and actin distribution are only weakly impacted by ligand clustering. In presence of ICAM-1 - the ligand of the T cell integrin LFA-1 - on the SLB, the TCR is still clustered due to the patterning of its ligands, but now global parameters are also impacted. The actin organization changes to a peripheral ring, resembling the classical actin distribution seen on homogeneous substrates, the patterned membrane topography disappears and the membrane is flat, whereas the cell area increases significantly. These observations taken together point to a possible pivotal role for LFA-1 in amplifying the effect of TCR-clustering. No such effect is evident for co-engagement of CD28, affected via its ligand B7.2. Unlike on ICAM-1, on B7.2 cell spreading and actin organization are similar for homogeneous and patterned substrates. However, TCR and ZAP-70 clusters are still formed in the patterned case. These results indicate complementary role for LFA-1 and CD28 in the regulation and putative coupling of TCR micro-clusters to actin. The engineered substrates presented here clearly have the potential to act as platform for fundamental research in immune cell biology, as well as translational analyses in immunotherapy, for example to screen molecules for their role in T cell adhesion/activation.


Subject(s)
Antigen-Presenting Cells/immunology , Biomimetics/methods , Lymphocyte Function-Associated Antigen-1/metabolism , Multiprotein Complexes/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Actins/metabolism , Antigen-Presenting Cells/chemistry , CD28 Antigens/metabolism , Humans , Intercellular Adhesion Molecule-1/metabolism , Jurkat Cells , Lipid Bilayers/chemistry , Lymphocyte Activation , Receptor Aggregation , Receptor Cross-Talk , Signal Transduction , ZAP-70 Protein-Tyrosine Kinase/metabolism
9.
ACS Nano ; 12(10): 9702-9713, 2018 10 23.
Article in English | MEDLINE | ID: mdl-30141896

ABSTRACT

Efficient delivery of tumor antigens and immunostimulatory adjuvants into lymph nodes is crucial for the maturation and activation of antigen-presenting cells (APCs), which subsequently induce adaptive antitumor immunity. A dissolving microneedle (MN) has been considered as an attractive method for transcutaneous immunization due to its superior ability to deliver vaccines through the stratum corneum in a minimally invasive manner. However, because dissolving MNs are mostly prepared using water-soluble sugars or polymers for their rapid dissolution in intradermal fluid after administration, they are often difficult to formulate with poorly water-soluble vaccine components. Here, we develop amphiphilic triblock copolymer-based dissolving MNs in situ that generate nanomicelles (NMCs) upon their dissolution after cutaneous application, which facilitate the efficient encapsulation of poorly water-soluble Toll-like receptor 7/8 agonist (R848) and the delivery of hydrophilic antigens. The sizes of NMCs range from 30 to 40 nm, which is suitable for the efficient delivery of R848 and antigens to lymph nodes and promotion of cellular uptake by APCs, minimizing systemic exposure of the R848. Application of MNs containing tumor model antigen (OVA) and R848 to the skin of EG7-OVA tumor-bearing mice induced a significant level of antigen-specific humoral and cellular immunity, resulting in significant antitumor activity.


Subject(s)
Cancer Vaccines/immunology , Nanoparticles/chemistry , Needles , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/therapeutic use , Animals , Antigen-Presenting Cells/chemistry , Antigen-Presenting Cells/immunology , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/immunology , Cancer Vaccines/chemistry , Drug Delivery Systems , Female , HCT116 Cells , Humans , Imidazoles/administration & dosage , Imidazoles/pharmacology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Micelles , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Polymers/chemistry , RAW 264.7 Cells , Surface-Active Agents/chemistry , Vaccination
10.
Nano Lett ; 17(11): 7045-7054, 2017 11 08.
Article in English | MEDLINE | ID: mdl-28994285

ABSTRACT

Particles engineered to engage and interact with cell surface ligands and to modulate cells can be harnessed to explore basic biological questions as well as to devise cellular therapies. Biology has inspired the design of these particles, such as artificial antigen-presenting cells (aAPCs) for use in immunotherapy. While much has been learned about mimicking antigen presenting cell biology, as we decrease the size of aAPCs to the nanometer scale, we need to extend biomimetic design to include considerations of T cell biology-including T-cell receptor (TCR) organization. Here we describe the first quantitative analysis of particle size effect on aAPCs with both Signals 1 and 2 based on T cell biology. We show that aAPCs, larger than 300 nm, activate T cells more efficiently than smaller aAPCs, 50 nm. The 50 nm aAPCs require saturating doses or require artificial magnetic clustering to activate T cells. Increasing ligand density alone on the 50 nm aAPCs did not increase their ability to stimulate CD8+ T cells, confirming the size-dependent phenomenon. These data support the need for multireceptor ligation and activation of T-cell receptor (TCR) nanoclusters of similar sizes to 300 nm aAPCs. Quantitative analysis and modeling of a nanoparticle system provides insight into engineering constraints of aAPCs for T cell immunotherapy applications and offers a case study for other cell-modulating particles.


Subject(s)
Antigen-Presenting Cells/chemistry , Artificial Cells/chemistry , Immunomodulation , Lymphocyte Activation , Nanoparticles/chemistry , Artificial Cells/immunology , Artificial Cells/ultrastructure , Biomimetic Materials/chemistry , Biomimetic Materials/therapeutic use , Biomimetics/methods , CD28 Antigens/immunology , CD8 Antigens/immunology , Humans , Immunotherapy , Ligands , Major Histocompatibility Complex , Nanoparticles/therapeutic use , Nanoparticles/ultrastructure , Neoplasms/therapy , Particle Size , Receptors, Antigen, T-Cell/immunology
11.
J Infect Dis ; 215(10): 1569-1579, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28368498

ABSTRACT

During Plasmodium falciparum infections, erythrocyte-stage parasites inhibit dendritic cell maturation and function, compromising effective antimalarial adaptive immunity. Human Vγ9Vδ2 T cells can act in vitro as antigen-presenting cells (APCs) and induce αß T-cell activation. However, the relevance of this activity in vivo has remained elusive. Because Vγ9Vδ2 T cells are activated during the early immune response against P. falciparum infection, we investigated whether they could contribute to the instruction of adaptive immune responses toward malaria parasites. In P. falciparum-infected patients, Vγ9Vδ2 T cells presented increased surface expression of APC-associated markers HLA-DR and CD86. In response to infected red blood cells in vitro, Vγ9Vδ2 T cells upregulated surface expression of HLA-DR, HLA-ABC, CD40, CD80, CD83, and CD86, induced naive αß T-cell responses, and cross- presented soluble prototypical protein to antigen-specific CD8+ T cells. Our findings qualify Vγ9Vδ2 T cells as alternative APCs, which could be harnessed for therapeutic interventions and vaccine design.


Subject(s)
Antigen-Presenting Cells/immunology , Lymphocyte Activation/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , T-Lymphocytes/immunology , Antigen Presentation/immunology , Antigen-Presenting Cells/chemistry , Humans , Phenotype , T-Lymphocytes/chemistry
12.
Indian J Pathol Microbiol ; 59(4): 469-473, 2016.
Article in English | MEDLINE | ID: mdl-27721276

ABSTRACT

BACKGROUND: Bronchiectasis is a chronic disease characterized by permanent dilatation of the conducting airways accompanied by sustained inflammation. AIMS: To assess whether chronic inflammation of lungs in bronchiectasis is associated with alterations in the numbers of infiltrating antigen presenting cell (APC). SETTING AND DESIGN: Lobectomy specimens from 12 nonsmoker, nonasthmatic patients with acquired (noncongenital) bronchiectasis and six control patients were included in the study. Histopathology slides were reviewed, and immunohistochemical markers for dendritic cells (DCs) macrophages and Langerhans cells have been applied and analyzed. MATERIALS AND METHODS: Tissue specimens were stained by immunohistochemistry using markers for DCs (CD83 and CD23), macrophages (CD68 and CD163), and Langerhans cells (CD1A and S-100 protein). The mean cell counts of stained cells in five high power microscopic fields were recorded. STATISTICAL ANALYSIS USED: Descriptive statistics, mean, standard deviation, median, and interquartile range were used. A nonparametric Mann-Whitney U-test was used to compare cell counts between bronchiectasis and control patients. P <0.05 was considered significant. RESULTS: The mean age of patients with bronchiectasis and controls was 36.7 ± 16.6 and 31.8 ± 22.6 years, respectively. The predominant cell type among the patients was macrophage (median 50.5) followed by DCs (median 44.85), histiocytes (median 32), and Langerhans cells (median 5%). Compared to the controls a significantly higher number of macrophages (P = 0.01), DCs (P = 0.001), and Langerhans cells (P = 0.014) were present. CONCLUSION: Chronic inflammatory response in acquired (noncongenital) bronchiectasis is most probably mediated by increased infiltration of APCs in lung tissues.


Subject(s)
Antigen-Presenting Cells/immunology , Bronchiectasis/pathology , Inflammation/pathology , Lung/pathology , Adult , Antigen-Presenting Cells/chemistry , Antigens, CD/analysis , Female , Histocytochemistry , Humans , Immunohistochemistry , Leukocyte Count , Male , Microscopy , Middle Aged , S100 Proteins/analysis , Young Adult
13.
Sci Rep ; 6: 23820, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27068235

ABSTRACT

Diet and microbiome derived indole derivatives are known to activate the ligand induced transcription factor, the Aryl hydrocarbon Receptor (AhR). While the current understanding of AhR biology has confirmed its role in mucosal lymphocytes, its function in intestinal antigen presenting cells (APCs) is poorly understood. Here, we report that Cre-mediated deletion of AhR in CD11c-expressing cells in C57/BL6 mice is associated with altered intestinal epithelial morphogenesis in vivo. Moreover, when co-cultured with AhR-deficient DCs ex vivo, intestinal organoids showed reduced SRY (sex determining region Y)-box 9 and increased Mucin 2 expression, which correlates with reduced Paneth cells and increased goblet cell differentiation, similar to the data obtained in vivo. Further, characterization of intestinal APC subsets, devoid of AhR, revealed an expression pattern associated with aberrant intrinsic Wnt pathway regulation. At a functional level, the loss of AhR in APCs resulted in a dysfunctional epithelial barrier, associated with a more aggressive chemically induced colitis compared to wild type animals. Our results are consistent with a model whereby the AhR signalling pathway may participate in the regulation of innate immunity through intestinal epithelium development and mucosal immunity.


Subject(s)
Antigen-Presenting Cells/physiology , Basic Helix-Loop-Helix Transcription Factors/metabolism , CD11c Antigen/analysis , Colitis/pathology , Intestinal Mucosa/growth & development , Intestinal Mucosa/immunology , Receptors, Aryl Hydrocarbon/metabolism , Animals , Antigen-Presenting Cells/chemistry , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Coculture Techniques , Gene Deletion , Gene Expression Regulation , Immunity, Innate , Mice, Inbred C57BL , Organoids , Receptors, Aryl Hydrocarbon/deficiency , Receptors, Aryl Hydrocarbon/genetics , Wnt Signaling Pathway
14.
EMBO J ; 35(1): 89-101, 2016 Jan 04.
Article in English | MEDLINE | ID: mdl-26612827

ABSTRACT

Multiple sclerosis is the most frequent chronic inflammatory disease of the CNS. The entry and survival of pathogenic T cells in the CNS are crucial for the initiation and persistence of autoimmune neuroinflammation. In this respect, contradictory evidence exists on the role of the most potent type of antigen-presenting cells, dendritic cells. Applying intravital two-photon microscopy, we demonstrate the gatekeeper function of CNS professional antigen-presenting CD11c(+) cells, which preferentially interact with Th17 cells. IL-17 expression correlates with expression of GM-CSF by T cells and with accumulation of CNS CD11c(+) cells. These CD11c(+) cells are organized in perivascular clusters, targeted by T cells, and strongly express the inflammatory chemokines Ccl5, Cxcl9, and Cxcl10. Our findings demonstrate a fundamental role of CNS CD11c(+) cells in the attraction of pathogenic T cells into and their survival within the CNS. Depletion of CD11c(+) cells markedly reduced disease severity due to impaired enrichment of pathogenic T cells within the CNS.


Subject(s)
Antigen-Presenting Cells/physiology , Brain/pathology , CD11c Antigen/analysis , Dendritic Cells/physiology , Encephalomyelitis, Autoimmune, Experimental/pathology , T-Lymphocytes/immunology , Animals , Antigen-Presenting Cells/chemistry , Brain/immunology , Cell Movement , Dendritic Cells/chemistry , Encephalomyelitis, Autoimmune, Experimental/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Interleukin-17/metabolism , Mice, Inbred C57BL , T-Lymphocytes/physiology , Th17 Cells/physiology
15.
PLoS Comput Biol ; 11(12): e1004481, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26699430

ABSTRACT

We propose a minimal mathematical model for the physical basis of membrane protein patterning in the immunological synapse (IS), which encompass membrane mechanics, protein binding kinetics and motion, and fluid flow in the synaptic cleft. Our theory leads to simple predictions for the spatial and temporal scales of protein cluster formation, growth and arrest as a function of membrane stiffness, rigidity and kinetics of the adhesive proteins, and the fluid flow in the synaptic cleft. Numerical simulations complement these scaling laws by quantifying the nucleation, growth and stabilization of proteins domains on the size of the cell. Direct comparison with experiment shows that passive elastohydrodynamics and kinetics of protein binding in the synaptic cleft can describe the short-time formation and organization of protein clusters, without evoking any active processes in the cytoskeleton. Despite the apparent complexity of the process, our analysis shows that just two dimensionless parameters characterize the spatial and temporal evolution of the protein pattern: a ratio of membrane elasticity to protein stiffness, and the ratio of a hydrodynamic time scale for fluid flow relative to the protein binding rate. A simple phase diagram encompasses the variety of patterns that can arise.


Subject(s)
Antigen-Presenting Cells/chemistry , Lipid Bilayers/chemistry , Membrane Proteins/chemistry , Models, Chemical , Models, Immunological , T-Lymphocytes/chemistry , Antigen-Presenting Cells/immunology , Computer Simulation , Elastic Modulus , Humans , Hydrodynamics , Immunological Synapses , Lipid Bilayers/immunology , Mechanotransduction, Cellular/immunology , Membrane Fluidity/immunology , Membrane Proteins/immunology , Models, Molecular , Protein Binding , T-Lymphocytes/immunology , Tensile Strength
16.
Am J Dermatopathol ; 37(4): 269-73, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25365500

ABSTRACT

Indeterminate leprosy (IL) is the early phase of Hansen disease and reword (APCs). Langerhans cells and dermal dendrocytes FXIIIa positive (DDFXIIIa) are the major APCs in the skin and can be identified by the expression of CD1a and FXIIIa, respectively, by immunohistochemical techniques. Plasmacytoid dendritic cells (PDCs) are another type of dermal dendrocytes with a questionable antigen-presenting function and can be highlighted by anti-CD123 expression. To our knowledge, there are no studies evaluating DDFXIIIa and PDC in IL. The purpose was to investigate the involvement of these cells in the pathogenesis of IL. The authors performed a retrospective study on 18 cases of IL (10 confirmed and 8 suspected) to investigate expression of FXIIIa, CD1a, and CD123. The results were compared with normal skin (for CD1a and FXIIIa only). A higher amount of FXIIIa-positive cells (P , 0.05) in confirmed and suspected IL cases was noted when comparing with normal skin. However, CD1a showed no quantitative differences in the epidermis of IL lesions when comparing with normal skin and CD123 expression was negligible. Based on these findings, the authors postulate that Langerhans cells and PDCs do not have a major role in IL and that DDFXIIIa may be the main APCs in IL. Further study is required to establish this.


Subject(s)
Antigen-Presenting Cells/chemistry , Dermis/chemistry , Factor XIIIa/analysis , Leprosy/metabolism , Adolescent , Adult , Antigen-Presenting Cells/classification , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/pathology , Antigens, CD1/analysis , Biomarkers/analysis , Biopsy , Dermis/immunology , Dermis/pathology , Female , Humans , Immunohistochemistry , Interleukin-3 Receptor alpha Subunit/analysis , Leprosy/immunology , Leprosy/pathology , Male , Middle Aged , Phenotype , Retrospective Studies , Young Adult
17.
ACS Appl Mater Interfaces ; 6(21): 18435-9, 2014.
Article in English | MEDLINE | ID: mdl-25343426

ABSTRACT

Here we show that the multifunctionality of Janus particles can be exploited for in vitro T cell activation. We engineer bifunctional Janus particles on which the spatial distribution of two ligands, anti-CD3 and fibronectin, mimics the "bull's eye" protein pattern formed in the membrane junction between a T cell and an antigen-presenting cell. Different levels of T cell activation can be achieved by simply switching the spatial distribution of the two ligands on the surfaces of the "bull's eye" particles. We find that the ligand pattern also affects clustering of intracellular proteins. This study demonstrates that anisotropic particles, such as Janus particles, can be developed as artificial antigen-presenting cells for modulating T cell activation.


Subject(s)
Antigen-Presenting Cells/immunology , Artificial Cells/immunology , Biocompatible Materials/pharmacology , Lymphocyte Activation/drug effects , Models, Immunological , T-Lymphocytes/drug effects , Antigen-Presenting Cells/chemistry , Artificial Cells/chemistry , Biocompatible Materials/chemistry , Biotechnology , Calcium/analysis , Calcium/metabolism , Humans , Immunological Synapses , Intracellular Space/chemistry , Intracellular Space/metabolism , Jurkat Cells , T-Lymphocytes/chemistry , T-Lymphocytes/metabolism
18.
Hum Immunol ; 75(6): 514-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24755351

ABSTRACT

Despite tremendous potential utility in clinical medicine and research, the discovery and characterization of T-cell antigens has lagged behind most other areas of health research in joining the high-throughput '-omics' revolution. Partially responsible for this is the complex nature of the interactions between effector T cells and antigen-presenting cells. Further contributing to the challenge is the vastness of both the T-cell repertoire and the large number of potential T-cell epitopes. In this review, we trace the development of various discovery strategies, the technical platforms used to carry them out, and we assess the level of success achieved in the field today.


Subject(s)
Antigen-Presenting Cells/chemistry , Epitopes, T-Lymphocyte/chemistry , Peptides/chemistry , Receptors, Antigen, T-Cell/chemistry , T-Lymphocytes/chemistry , Antigen Presentation , Antigen-Presenting Cells/immunology , Epitopes, T-Lymphocyte/immunology , Gene Library , High-Throughput Screening Assays , Humans , Major Histocompatibility Complex/immunology , Microarray Analysis , Peptides/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology
19.
Nano Lett ; 13(11): 5619-26, 2013.
Article in English | MEDLINE | ID: mdl-24117051

ABSTRACT

Antigen recognition is a key event during T cell activation. Here, we introduce nanopatterned antigen arrays that mimic the antigen presenting cell surface during T cell activation. The assessment of activation related events revealed the requirement of a minimal density of 90-140 stimulating major histocompatibility complex class II proteins (pMHC) molecules per µm(2). We demonstrate that these substrates induce T cell responses in a pMHC dose-dependent manner and that the number of presented pMHCs dominates over local pMHC density.


Subject(s)
Antigen-Presenting Cells/immunology , Lymphocyte Activation/immunology , Nanoparticles/chemistry , T-Lymphocytes/immunology , Antigen-Presenting Cells/chemistry , Humans , Major Histocompatibility Complex/immunology , Membrane Proteins/immunology , Synapses/immunology , T-Lymphocytes/chemistry
20.
Nanomedicine (Lond) ; 8(7): 1173-89, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23837856

ABSTRACT

Artificial antigen-presenting cells (aAPCs) have shown great initial promise for ex vivo activation of cytotoxic T cells. The development of aAPCs has focused mainly on the choice of proteins to use for surface presentation to T cells when conjugated to various spherical, microscale particles. We review here biomimetic nanoengineering approaches that have been applied to the development of aAPCs that move beyond initial concepts about aAPC development. This article also discusses key technologies that may be enabling for the development of nano- and micro-scale aAPCs with nanoscale features, and suggests several future directions for the field.


Subject(s)
Antigen-Presenting Cells/immunology , Nanotechnology/methods , Animals , Antigen-Presenting Cells/chemistry , Biomedical Engineering/methods , Humans , Models, Biological , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL