Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 685
1.
Front Immunol ; 15: 1345381, 2024.
Article En | MEDLINE | ID: mdl-38736890

Sjögren's syndrome (SS) is a chronic systemic autoimmune disease that typically presents with lymphocyte, dendritic cell, and macrophage infiltration of exocrine gland ducts and the formation of ectopic germinal centers. The interactions of lymphocyte homing receptors and addressins and chemokines and their receptors, such as α4ß7/MAdCAM-1, LFA-1/ICAM-1, CXCL13/CXCR5, CCL25/CCR9, CX3CL1/CX3CR1, play important roles in the migration of inflammatory cells to the focal glands and the promotion of ectopic germinal center formation in SS. A variety of molecules have been shown to be involved in lymphocyte homing, including tumor necrosis factor-α, interferon (IFN)-α, IFN-ß, and B cell activating factor. This process mainly involves the Janus kinase-signal transducer and activator of transcription signaling pathway, lymphotoxin-ß receptor pathway, and nuclear factor-κB signaling pathway. These findings have led to the development of antibodies to cell adhesion molecules, antagonists of chemokines and their receptors, compounds interfering with chemokine receptor signaling, and gene therapies targeting chemokines and their receptors, providing new targets for the treatment of SS in humans. The aim of this study was to explore the relationship between lymphocyte homing and the pathogenesis of SS, and to provide a review of recent studies addressing lymphocyte homing in targeted therapy for SS.


Chemokines , Sjogren's Syndrome , Sjogren's Syndrome/immunology , Sjogren's Syndrome/metabolism , Humans , Chemokines/metabolism , Chemokines/immunology , Signal Transduction , Animals , Receptors, Lymphocyte Homing/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Receptors, Chemokine/metabolism , Receptors, Chemokine/immunology
2.
Int J Mol Sci ; 24(11)2023 May 31.
Article En | MEDLINE | ID: mdl-37298541

Integrin receptors mediate cell-cell interactions via the recognition of cell-adhesion glycoproteins, as well as via the interactions of cells with proteins of the extracellular matrix, and upon activation they transduce signals bi-directionally across the cell membrane. In the case of injury, infection, or inflammation, integrins of ß2 and α4 families participate in the recruitment of leukocytes, a multi-step process initiated by the capturing of rolling leukocytes and terminated by their extravasation. In particular, α4ß1 integrin is deeply involved in leukocyte firm adhesion preceding extravasation. Besides its well-known role in inflammatory diseases, α4ß1 integrin is also involved in cancer, being expressed in various tumors and showing an important role in cancer formation and spreading. Hence, targeting this integrin represents an opportunity for the treatment of inflammatory disorders, some autoimmune diseases, and cancer. In this context, taking inspiration from the recognition motives of α4ß1 integrin with its natural ligands FN and VCAM-1, we designed minimalist α/ß hybrid peptide ligands, with our approach being associated with a retro strategy. These modifications are expected to improve the compounds' stability and bioavailability. As it turned out, some of the ligands were found to be antagonists, being able to inhibit the adhesion of integrin-expressing cells to plates coated with the natural ligands without inducing any conformational switch and any activation of intracellular signaling pathways. An original model structure of the receptor was generated using protein-protein docking to evaluate the bioactive conformations of the antagonists via molecular docking. Since the experimental structure of α4ß1 integrin is still unknown, the simulations might also shed light on the interactions between the receptor and its native protein ligands.


Neoplasms , Peptidomimetics , Humans , Integrin alpha4beta1/metabolism , Receptors, Lymphocyte Homing/metabolism , Molecular Docking Simulation , Peptidomimetics/pharmacology , Integrin beta1 , Ligands , Integrins/metabolism , Cell Adhesion , Vascular Cell Adhesion Molecule-1/metabolism
3.
J Nucl Med ; 64(4): 555-560, 2023 04.
Article En | MEDLINE | ID: mdl-36302655

Bexmarilimab is a new humanized monoclonal antibody against common lymphatic endothelial and vascular endothelial receptor-1 (CLEVER-1) and is in clinical trials for macrophage-guided cancer immunotherapy. In addition being associated with cancer, CLEVER-1 is also associated with fibrosis. To facilitate prospective human PET studies, we preclinically evaluated 89Zr-labeled bexmarilimab in rabbits. Methods: Bexmarilimab was conjugated with desferrioxamine (DFO) and radiolabeled with 89Zr. Retained immunoreactivity was confirmed by flow cytometry. The distribution kinetics of intravenously administered 89Zr-DFO-bexmarilimab (0.1 mg/kg) were determined for up to 7 d in a rabbit model of renal fibrosis mediated by unilateral ureteric obstruction. The in vivo stability of 89Zr-DFO-bexmarilimab was evaluated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis in combination with autoradiography. Additionally, we estimated the human radiation dose from data obtained in healthy rabbits. Results: 89Zr-DFO-bexmarilimab cleared rapidly from the blood circulation and distributed to the liver and spleen. At 24 h after injection, PET/CT, ex vivo γ-counting, and autoradiography demonstrated that there was significantly higher 89Zr-DFO-bexmarilimab uptake in unilateral ureteric obstruction-operated fibrotic renal cortex, characterized by abundant CLEVER-1-positive cells, than in contralateral or healthy kidneys. The estimated effective dose for a 70-kg human was 0.70 mSv/MBq. Conclusion: The characteristics of 89Zr-DFO-bexmarilimab support future human PET studies to, for example, stratify patients for bexmarilimab treatment, evaluate the efficacy of treatment, or monitor disease progression.


Kidney Diseases , Neoplasms , Animals , Humans , Rabbits , Antibodies, Monoclonal/therapeutic use , Cell Line, Tumor , Deferoxamine , Fibrosis , Positron Emission Tomography Computed Tomography/methods , Positron-Emission Tomography/methods , Prospective Studies , Radioisotopes/therapeutic use , Zirconium/therapeutic use , Cell Adhesion Molecules, Neuronal/metabolism , Receptors, Lymphocyte Homing/metabolism
4.
J Virol ; 96(15): e0080422, 2022 08 10.
Article En | MEDLINE | ID: mdl-35852355

CD4dim CD8bright T cells are a mature population of CD8+ T cells that upon activation upregulate CD4 dimly on their surface. Expression of CD4 on these cells suggests that they can be an additional source of HIV neuroinvasion and persistence in the brain. We used HIV-infected NOD/SCID/IL-2rcγ-/- (NSG) humanized mice to track CD4dim CD8bright T cell homing to the brain and define their role in HIV dissemination into the brain. We report here that CD4dim CD8bright T cells are found in the brain at a median frequency of 2.6% and in the spleen at median frequency of 7.6% of CD3+ T cells. In the brain, 10 to 20% of CD4dim CD8bright T cells contain integrated provirus, which is infectious as demonstrated by viral outgrowth assay. CD4dim CD8bright T cells in the brain exhibited significantly higher expression of the brain homing receptors CX3CR1 and CXCR3 in comparison to their single-positive CD8+ T cell counterpart. Blocking lymphocyte trafficking into the brain of humanized mice via anti-VLA4 and anti-LFA1 antibodies reduced CD4dim CD8bright T cell trafficking into the brain by 60% and diminished brain HIV proviral DNA by 72%. Collectively, our findings demonstrate that CD4dim CD8bright T cells can home to the brain and support productive HIV replication. These studies also reveal for the first time that CD4dim CD8bright T cells are capable of HIV neuroinvasion and are a reservoir for HIV. IMPORTANCE We report here a seminal finding of a novel population of T cells, termed CD4dim CD8bright T cells, that plays a role in HIV neuroinvasion and is a reservoir for HIV in the brain.


Brain , CD4 Antigens , CD8 Antigens , CD8-Positive T-Lymphocytes , Cell Movement , HIV Infections , HIV-1 , Viral Tropism , Animals , Brain/immunology , Brain/metabolism , Brain/virology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CX3C Chemokine Receptor 1/metabolism , HIV Infections/immunology , HIV Infections/virology , HIV-1/pathogenicity , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Mice , Mice, Inbred NOD , Mice, SCID , Proviruses/genetics , Proviruses/isolation & purification , Receptors, CXCR3/metabolism , Receptors, Lymphocyte Homing/metabolism
5.
Front Immunol ; 12: 721453, 2021.
Article En | MEDLINE | ID: mdl-34539657

Previous studies have evaluated the roles of T and B cells in the pathogenesis of Sjögren's syndrome (SS); however, their relationships with age-dependent and metabolic abnormalities remain unclear. We examined the impacts of changes associated with aging or metabolic abnormalities on populations of T and B cells and SS disease severity. We detected increased populations of IL-17-producing T and B cells, which regulate inflammation, in the salivary glands of NOD/ShiLtJ mice. Inflammation-induced human submandibular gland cell death, determined based on p-MLKL and RIPK3 expression levels, was significantly increased by IL-17 treatment. Among IL-17-expressing cells in the salivary gland, peripheral blood, and spleen, the α4ß7 (gut-homing integrin)-negative population was significantly increased in aged NOD/ShiLtJ mice. The α4ß7-positive population markedly increased in the intestines of aged NOD/ShiLtJ mice following retinoic acid (RA) treatment. A significant increase in α4ß7-negative IL-17-expressing cells in salivary glands may be involved in the onset and progression of SS. These results suggest the potential therapeutic utility of RA in SS treatment.


Interleukin-17/metabolism , Receptors, CCR/metabolism , Receptors, Lymphocyte Homing/metabolism , Sjogren's Syndrome/etiology , Sjogren's Syndrome/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Animals , Biomarkers , Blood Glucose , Cell Death , Cell Self Renewal , Disease Models, Animal , Disease Susceptibility , Interleukin-17/blood , Mice , Salivary Glands/immunology , Salivary Glands/metabolism , Salivary Glands/pathology , Sjogren's Syndrome/pathology , Stem Cells/cytology , Stem Cells/metabolism
6.
J Immunol ; 207(7): 1763-1775, 2021 10 01.
Article En | MEDLINE | ID: mdl-34470859

Regulatory T cells (Tregs) reside in nonlymphoid tissues where they carry out unique functions. The molecular mechanisms responsible for Treg accumulation and maintenance in these tissues are relatively unknown. Using an unbiased discovery approach, we identified LAYN (layilin), a C-type lectin-like receptor, to be preferentially and highly expressed on a subset of activated Tregs in healthy and diseased human skin. Expression of layilin on Tregs was induced by TCR-mediated activation in the presence of IL-2 or TGF-ß. Mice with a conditional deletion of layilin in Tregs had reduced accumulation of these cells in tumors. However, these animals somewhat paradoxically had enhanced immune regulation in the tumor microenvironment, resulting in increased tumor growth. Mechanistically, layilin expression on Tregs had a minimal effect on their activation and suppressive capacity in vitro. However, expression of this molecule resulted in a cumulative anchoring effect on Treg dynamic motility in vivo. Taken together, our results suggest a model whereby layilin facilitates Treg adhesion in skin and, in doing so, limits their suppressive capacity. These findings uncover a unique mechanism whereby reduced Treg motility acts to limit immune regulation in nonlymphoid organs and may help guide strategies to exploit this phenomenon for therapeutic benefit.


Carrier Proteins/metabolism , Membrane Glycoproteins/metabolism , Receptors, Lymphocyte Homing/metabolism , Skin/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Carrier Proteins/genetics , Cell Movement , Cells, Cultured , Humans , Immune Tolerance , Lymphocyte Activation , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Immunological , Organ Specificity , Receptors, Lymphocyte Homing/genetics , Transforming Growth Factor beta/metabolism
7.
Cell Mol Life Sci ; 78(21-22): 6963-6978, 2021 Nov.
Article En | MEDLINE | ID: mdl-34586443

The endogenous chemokines CCL19 and CCL21 signal via their common receptor CCR7. CCL21 is the main lymph node homing chemokine, but a weak chemo-attractant compared to CCL19. Here we show that the 41-amino acid positively charged peptide, released through C-terminal cleavage of CCL21, C21TP, boosts the immune cell recruiting activity of CCL21 by up to 25-fold and the signaling activity via CCR7 by ~ 100-fold. Such boosting is unprecedented. Despite the presence of multiple basic glycosaminoglycan (GAG) binding motifs, C21TP boosting of CCL21 signaling does not involve interference with GAG mediated cell-surface retention. Instead, boosting is directly dependent on O-glycosylations in the CCR7 N-terminus. As dictated by the two-step binding model, the initial chemokine binding involves interaction of the chemokine fold with the receptor N-terminus, followed by insertion of the chemokine N-terminus deep into the receptor binding pocket. Our data suggest that apart from a role in initial chemokine binding, the receptor N-terminus also partakes in a gating mechanism, which could give rise to a reduced ligand activity, presumably through affecting the ligand positioning. Based on experiments that support a direct interaction of C21TP with the glycosylated CCR7 N-terminus, we propose that electrostatic interactions between the positively charged peptide and sialylated O-glycans in CCR7 N-terminus may create a more accessible version of the receptor and thus guide chemokine docking to generate a more favorable chemokine-receptor interaction, giving rise to the peptide boosting effect.


Chemokine CCL21/metabolism , Dendritic Cells/metabolism , Lymph Nodes/metabolism , Receptors, CCR7/metabolism , Receptors, Lymphocyte Homing/metabolism , Signal Transduction/physiology , Animals , CHO Cells , Cells, Cultured , Cricetulus , Glycosylation , Humans , Ligands , Peptides/metabolism , Protein Binding/physiology , Static Electricity
8.
J Immunol ; 207(9): 2245-2254, 2021 11 01.
Article En | MEDLINE | ID: mdl-34561227

Targeting interactions between α4ß7 integrin and endothelial adhesion molecule MAdCAM-1 to inhibit lymphocyte migration to the gastrointestinal tract is an effective therapy in inflammatory bowel disease (IBD). Following lymphocyte entry into the mucosa, a subset of these cells expresses αEß7 integrin, which is expressed on proinflammatory lymphocytes, to increase cell retention. The factors governing lymphocyte migration into the intestinal mucosa and αE integrin expression in healthy subjects and IBD patients remain incompletely understood. We evaluated changes in factors involved in lymphocyte migration and differentiation within tissues. Both ileal and colonic tissue from active IBD patients showed upregulation of ICAM-1, VCAM-1, and MAdCAM-1 at the gene and protein levels compared with healthy subjects and/or inactive IBD patients. ß1 and ß7 integrin expression on circulating lymphocytes was similar across groups. TGF-ß1 treatment induced expression of αE on both ß7+ and ß7- T cells, suggesting that cells entering the mucosa independently of MAdCAM-1/α4ß7 can become αEß7+ ITGAE gene polymorphisms did not alter protein induction following TGF-ß1 stimulation. Increased phospho-SMAD3, which is directly downstream of TGF-ß, and increased TGF-ß-responsive gene expression were observed in the colonic mucosa of IBD patients. Finally, in vitro stimulation experiments showed that baseline ß7 expression had little effect on cytokine, chemokine, transcription factor, and effector molecule gene expression in αE+ and αE- T cells. These findings suggest cell migration to the gut mucosa may be altered in IBD and α4ß7-, and α4ß7+ T cells may upregulate αEß7 in response to TGF-ß once within the gut mucosa.


Antigens, CD/metabolism , Inflammatory Bowel Diseases/immunology , Integrin alpha Chains/metabolism , Integrin beta Chains/metabolism , Intestinal Mucosa/immunology , Receptors, Lymphocyte Homing/metabolism , T-Lymphocytes/immunology , Adult , Aged , Cell Movement , Female , Humans , Integrin beta Chains/genetics , Male , Middle Aged , Signal Transduction , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism
9.
Front Immunol ; 12: 760577, 2021.
Article En | MEDLINE | ID: mdl-34975851

Macrophages are key innate immune cells that mediate implant acceptance or rejection. Titanium implants degrade over time inside the body, which results in the release of implant wear-off particles. Titanium nanoparticles (TiNPs) favor pro-inflammatory macrophage polarization (M1) and lower tolerogenic activation (M2). GDF-15 regulates immune tolerance and fibrosis and is endocytosed by stabilin-1. How TiNPs affect the healing activities of macrophages and their release of circulating cytokines is an open question in regenerative medicine. In this study for the first time, we identified the transcriptional program induced and suppressed by TiNPs in human pro-inflammatory and healing macrophages. Microarray analysis revealed that TiNPs altered the expression of 5098 genes in M1 (IFN-γ-stimulated) and 4380 genes in M2 (IL-4-stimulated) macrophages. 1980 genes were differentially regulated in both M1 and M2. Affymetrix analysis, confirmed by RT-PCR, demonstrated that TiNPs upregulate expression of GDF-15 and suppress stabilin-1, scavenger receptor of GDF-15. TiNPs also significantly stimulated GDF-15 protein secretion in inflammatory and healing macrophages. Flow cytometry demonstrated, that scavenging activity of stabilin-1 was significantly suppressed by TiNPs. Confocal microscopy analysis showed that TiNPs impair internalization of stabilin-1 ligand acLDL and its transport to the endocytic pathway. Our data demonstrate that TiNPs have a dual effect on the GDF-15/stabilin-1 interaction in macrophage system, by increasing the production of GDF-15 and suppressing stabilin-1-mediated clearance function. In summary, this process can result in a significant increase of GDF-15 in the extracellular space and in circulation leading to unbalanced pro-fibrotic reactions and implant complications.


Cell Adhesion Molecules, Neuronal/metabolism , Growth Differentiation Factor 15/metabolism , Macrophages/drug effects , Nanoparticles/administration & dosage , Receptors, Lymphocyte Homing/metabolism , Titanium/administration & dosage , Cell Adhesion Molecules, Neuronal/genetics , Cells, Cultured , Endocytosis , Gene Expression/drug effects , Growth Differentiation Factor 15/genetics , Humans , Macrophages/metabolism , Prostheses and Implants , Receptors, Lymphocyte Homing/genetics
10.
Mucosal Immunol ; 14(3): 652-666, 2021 05.
Article En | MEDLINE | ID: mdl-33184477

Evidence from inflammatory bowel diseases (IBD) patients and animal models has indicated that gut inflammation is driven by effector CD4+ T-cell, including Th1 and Th17. Conversely, Treg seem to be dysfunctional in IBD. Importantly, dopamine, which is abundant in the gut mucosa under homoeostasis, undergoes a sharp reduction upon intestinal inflammation. Here we analysed the role of the high-affinity dopamine receptor D3 (DRD3) in gut inflammation. Our results show that Drd3 deficiency confers a stronger immunosuppressive potency to Treg, attenuating inflammatory colitis manifestation in mice. Mechanistic analyses indicated that DRD3-signalling attenuates IL-10 production and limits the acquisition of gut-tropism. Accordingly, the ex vivo transduction of wild-type Treg with a siRNA for Drd3 induced a potent therapeutic effect abolishing gut inflammation. Thus, our findings show DRD3-signalling as a major regulator of Treg upon gut inflammation.


Colitis/immunology , Dopaminergic Neurons/immunology , Inflammation/immunology , Inflammatory Bowel Diseases/immunology , Intestines/immunology , Receptors, Dopamine D3/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Cells, Cultured , Disease Models, Animal , Humans , Immunosuppression Therapy , Interleukin-10/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroimmunomodulation , RNA, Small Interfering/genetics , Receptors, Dopamine D3/genetics , Receptors, Lymphocyte Homing/metabolism , Signal Transduction
11.
Nature ; 587(7835): 619-625, 2020 11.
Article En | MEDLINE | ID: mdl-33208946

Although single-cell RNA sequencing studies have begun to provide compendia of cell expression profiles1-9, it has been difficult to systematically identify and localize all molecular cell types in individual organs to create a full molecular cell atlas. Here, using droplet- and plate-based single-cell RNA sequencing of approximately 75,000 human cells across all lung tissue compartments and circulating blood, combined with a multi-pronged cell annotation approach, we create an extensive cell atlas of the human lung. We define the gene expression profiles and anatomical locations of 58 cell populations in the human lung, including 41 out of 45 previously known cell types and 14 previously unknown ones. This comprehensive molecular atlas identifies the biochemical functions of lung cells and the transcription factors and markers for making and monitoring them; defines the cell targets of circulating hormones and predicts local signalling interactions and immune cell homing; and identifies cell types that are directly affected by lung disease genes and respiratory viruses. By comparing human and mouse data, we identified 17 molecular cell types that have been gained or lost during lung evolution and others with substantially altered expression profiles, revealing extensive plasticity of cell types and cell-type-specific gene expression during organ evolution including expression switches between cell types. This atlas provides the molecular foundation for investigating how lung cell identities, functions and interactions are achieved in development and tissue engineering and altered in disease and evolution.


Cells/classification , Cells/metabolism , Immunity , Lung/cytology , Sequence Analysis, RNA , Single-Cell Analysis , Transcriptome/genetics , Aged , Animals , Atlases as Topic , Biomarkers , Cell Communication , Cells/immunology , Chemokines/metabolism , Endothelial Cells/metabolism , Epithelial Cells/metabolism , Female , Humans , Lung/immunology , Male , Mice , Middle Aged , Receptors, Lymphocyte Homing/metabolism , Signal Transduction , Stromal Cells/metabolism
12.
Front Immunol ; 11: 572475, 2020.
Article En | MEDLINE | ID: mdl-33042152

Memory B cells have known to play an important role in the pathogenesis of rheumatoid arthritis (RA). With the emergence of B cell-targeted therapies, the modulation of memory B cells appears to be a key therapeutic target. Human peripheral memory B cells can be distinguished based on the phenotypic expression of CD27 and IgD, characterizing the three major B cell subpopulations: CD27+IgD+ pre-switch, CD27+IgD- post-switch, and CD27-IgD- double-negative memory B cells. We evaluated different memory cell populations for activation markers (CD95 and Ki-67) and chemokine receptors (CXCR3 and 4) expressing B cells in active RA, as well as under IL6-R blockade by tocilizumab (TCZ) and TNF-α blockade by adalimumab (ADA). Memory B cells were phenotypically analyzed from RA patients at baseline, week 12, and week 24 under TCZ or ADA treatment, respectively. Using flow cytometry, surface expression of CD95, intracellular Ki-67, and surface expressions of CXCR3 and CXCR4 were determined. Compared with healthy donors (n = 40), the phenotypic analysis of RA patients (n = 80) demonstrated that all three types of memory B cells were activated in RA patients. Surface and intracellular staining of B cells showed a significantly higher percentage of CD95+ (p < 0.0001) and Ki-67+ (p < 0.0001) cells, with numerically altered CXCR3+ and CXCR4+ cells in RA. CD95 and Ki-67 expressions were highest in post-switch memory B cells, whereas CD19+CXCR3+ and CD19+CXCR4+ expressing cells were substantially higher in the pre-switch compartment. In all subsets of the memory B cells, in vivo IL-6R, and TNF-α blockade significantly reduced the enhanced expressions of CD95 and Ki-67. Based on our findings, we conclude that the three major peripheral memory B cell populations, pre-, post-switch, and double-negative B cells, are activated in RA, demonstrating enhanced CD95 and Ki-67 expressions, and varied expression of CXCR3 and CXCR4 chemokine receptors when compared with healthy individuals. This activation can be efficaciously modulated under cytokine inhibition in vivo.


Adalimumab/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Arthritis, Rheumatoid/immunology , B-Lymphocyte Subsets/immunology , B-Lymphocytes/immunology , Immunotherapy/methods , Adult , Aged , Arthritis, Rheumatoid/drug therapy , Cells, Cultured , Female , Humans , Immunoglobulin D/metabolism , Immunologic Memory , Immunomodulation , Interleukin-6/antagonists & inhibitors , Male , Middle Aged , Receptors, Interleukin-6/antagonists & inhibitors , Receptors, Lymphocyte Homing/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
13.
Nat Commun ; 11(1): 4767, 2020 09 21.
Article En | MEDLINE | ID: mdl-32958743

Psoriatic arthritis (PsA) is a debilitating immune-mediated inflammatory arthritis of unknown pathogenesis commonly affecting patients with skin psoriasis. Here we use complementary single-cell approaches to study leukocytes from PsA joints. Mass cytometry demonstrates a 3-fold expansion of memory CD8 T cells in the joints of PsA patients compared to peripheral blood. Meanwhile, droplet-based and plate-based single-cell RNA sequencing of paired T cell receptor alpha and beta chain sequences show pronounced CD8 T cell clonal expansions within the joints. Transcriptome analyses find these expanded synovial CD8 T cells to express cycling, activation, tissue-homing and tissue residency markers. T cell receptor sequence comparison between patients identifies clonal convergence. Finally, chemokine receptor CXCR3 is upregulated in the expanded synovial CD8 T cells, while two CXCR3 ligands, CXCL9 and CXCL10, are elevated in PsA synovial fluid. Our data thus provide a quantitative molecular insight into the cellular immune landscape of psoriatic arthritis.


Arthritis, Psoriatic/immunology , CD8-Positive T-Lymphocytes/immunology , Clonal Selection, Antigen-Mediated , Receptors, Lymphocyte Homing/metabolism , Synovial Fluid/immunology , Arthritis, Psoriatic/blood , CD8-Positive T-Lymphocytes/metabolism , Gene Expression Profiling , Humans , Immunologic Memory , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Chemokine/metabolism , Receptors, Lymphocyte Homing/genetics , Single-Cell Analysis , Synovial Membrane/immunology
14.
Br J Cancer ; 123(4): 501-509, 2020 08.
Article En | MEDLINE | ID: mdl-32595212

Clever-1 (also known as Stabilin-1 and FEEL-1) is a scavenger receptor expressed on lymphatic endothelial cells, sinusoidal endothelial cells and immunosuppressive monocytes and macrophages. Its role in cancer growth and spread first became evident in Stab1-/- knockout mice, which have smaller primary tumours and metastases. Subsequent studies in mice and humans have shown that immunotherapeutic blockade of Clever-1 can activate T-cell responses, and that this response is mainly mediated by a phenotypic change in macrophages and monocytes from immunosuppressive to pro-inflammatory following Clever-1 inhibition. Analyses of human cancer cohorts have revealed marked associations between the number of Clever-1-positive macrophages and patient outcome. As hardly any reports to date have addressed the role of Clever-1 in immunotherapy resistance and T-cell dysfunction, we performed data mining using several published cancer cohorts, and observed a remarkable correlation between Clever-1 positivity and resistance to immune checkpoint therapies. This result provides impetus and potential for the ongoing clinical trial targeting Clever-1 in solid tumours, which has so far shown a shift towards immune activation when a particular epitope of Clever-1 is blocked.


Cell Adhesion Molecules, Neuronal/genetics , Drug Resistance, Neoplasm , Neoplasms/genetics , Receptors, Lymphocyte Homing/genetics , Animals , Cell Adhesion Molecules, Neuronal/metabolism , Disease Progression , Humans , Immunotherapy , Macrophages/metabolism , Mice , Mice, Knockout , Neoplasms/drug therapy , Neoplasms/immunology , Receptors, Lymphocyte Homing/metabolism , T-Lymphocytes/metabolism
15.
Front Immunol ; 11: 529, 2020.
Article En | MEDLINE | ID: mdl-32318062

The chemokine receptor CCR4 has emerged as a skin-homing molecule important for the migration of T cells from the blood to the dermis. From our previous data on psoriasis patients, CCR4+ memory T cells emerged as a putative recirculating population between skin and blood. Here we focused our attention on the expression of CCR4 and skin-tropic molecules in the different stages of memory T cell differentiation. We analyzed the chemokine receptor profile in CD8+ and CD4+ CD45RA-CCR7+ (TCM) and CD45RA-CCR7- (TEM) cells. Subpopulations were further divided on the basis of CD62L expression, and the distribution among the subsets of the skin-homing molecule CLA (Cutaneous Lymphocyte Antigen) was evaluated. The characterization was performed on peripheral blood mononuclear cells isolated from 21 healthy subjects and 24 psoriasis patients. The results indicate that (i) the skin-homing CCR4 marker is mainly expressed in TCM cells, (ii) CCR4+ TCM cells also express high level of CLA and that (iii) the more differentiated phenotype TEM expresses CXCR3 and CCR5 but lower level of CCR4 and CLA. This indicates that progressive stages of memory T cell differentiation have profoundly different chemokine receptor patterns, with CD8+ TCM displaying a marked skin-tropic phenotype CLA+CCR4+. Differential skin-tropic phenotype between TCM and TEM cells was observed in both healthy subjects and psoriasis patients. However, patients showed an expanded circulating population of CD8+ TCM cells with phenotype CCR4+CXCR3+ that could play a role in the pathophysiology of psoriasis and possibly in disease recurrence.


CD8-Positive T-Lymphocytes/immunology , Psoriasis/immunology , Receptors, CCR4/metabolism , Receptors, Lymphocyte Homing/metabolism , Skin/immunology , Adult , Cell Differentiation , Female , Humans , Immunologic Memory , Immunophenotyping , Male , Middle Aged , Oligosaccharides , Organ Specificity , Sialyl Lewis X Antigen/analogs & derivatives
16.
Nat Commun ; 11(1): 1114, 2020 02 28.
Article En | MEDLINE | ID: mdl-32111837

Little is known regarding lymph node (LN)-homing of immune cells via afferent lymphatics. Here, we show, using a photo-convertible Dendra-2 reporter, that recently activated CD4 T cells enter downstream LNs via afferent lymphatics at high frequencies. Intra-lymphatic immune cell transfer and live imaging data further show that activated T cells come to an instantaneous arrest mediated passively by the mechanical 3D-sieve barrier of the LN subcapsular sinus (SCS). Arrested T cells subsequently migrate randomly on the sinus floor independent of both chemokines and integrins. However, chemokine receptors are imperative for guiding cells out of the SCS, and for their subsequent directional translocation towards the T cell zone. By contrast, integrins are dispensable for LN homing, yet still contribute by increasing the dwell time within the SCS and by potentially enhancing T cell sensing of chemokine gradients. Together, these findings provide fundamental insights into mechanisms that control homing of lymph-derived immune cells.


CD4-Positive T-Lymphocytes/physiology , Cell Movement/immunology , Chemokines/metabolism , Integrins/metabolism , Lymph Nodes/physiology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Endothelium, Lymphatic/physiology , Integrins/genetics , Lymph/cytology , Lymph Nodes/cytology , Lymphocyte Activation , Mice , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Receptors, Lymphocyte Homing/metabolism
17.
Immunohorizons ; 4(2): 57-71, 2020 02 07.
Article En | MEDLINE | ID: mdl-32034085

The Tec kinase IL-2-inducible T cell kinase (ITK) regulates the expression of TCR-induced genes. Itk-/- T cell responses are impaired but not absent. ITK inhibition prevented colitis disease progression and impaired T cell migration to the colon in mice. To examine the function of ITK in T cell migration to the intestine, we examined the number of gut T cells in Itk-/- mice and then evaluated their expression of gut-homing receptors. Combined with in vitro murine T cell stimulation and in vivo migration assay using congenic B6 mice, we demonstrated an essential role for ITK in T cell migration to the intestine in mice. Reconstitution of Itk-/- mouse CD8+ T cells with IFN regulatory factor 4 restored gut-homing properties, providing mechanistic insight into the function of ITK-mediated signaling in CD8+ T cell migration to the intestinal mucosa in mice.


CD8-Positive T-Lymphocytes/physiology , Chemotaxis, Leukocyte , Intestines/immunology , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/transplantation , Cells, Cultured , Interferon Regulatory Factors/metabolism , Intestines/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein-Tyrosine Kinases/deficiency , Receptors, Lymphocyte Homing/metabolism , Rhadinovirus/physiology , Virus Replication
18.
FEBS Open Bio ; 10(4): 674-688, 2020 04.
Article En | MEDLINE | ID: mdl-32027093

Prostate adenocarcinoma (PCa) is the most common cause of death due to malignancy among men, and bone metastasis is the leading cause of mortality in patients with PCa. Therefore, identifying the causes and molecular mechanism of bone metastasis is important for early detection, diagnosis and personalized therapy. In this study, we systematically analyzed molecular correlates of bone metastasis by bioinformatics analysis. A total of 12 differentially expressed microRNAs (miRNAs) and 102 differentially expressed genes were identified. Five miRNAs had prognostic significance in biochemical recurrence-free survival (miR-636, miR-491-5p, miR-199b-5p, miR-199b-3p, miR-28-3p). The differentially expressed genes were significantly enriched in extracellular matrix, cell-substrate adhesion, collagen and integrin. Seven hub genes (VCAN, COL3A1, COL1A1, APOE, COL1A2, SDC1, THY1) with worse biochemical recurrence-free survival and one hub gene (MMP9) with worse overall survival were detected. miR-636, a novel oncogene, was found to be up-regulated in bone metastatic PCa tissues and also predominately up-regulated in human PCa cell lines. miR-636 promoted cellular invasion and migration, and may promote bone metastasis via targeting MBNL2, TNS1 and STAB1. In conclusion, we have successfully defined molecular signatures of bone metastasis in PCa.


Bone Neoplasms/genetics , Bone Neoplasms/secondary , Computational Biology/methods , MicroRNAs/genetics , Oncogenes , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Bone Neoplasms/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Cell Movement/genetics , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Male , MicroRNAs/metabolism , Neoplasm Invasiveness/genetics , PC-3 Cells , Prostatic Neoplasms/metabolism , Protein Interaction Maps/genetics , RNA-Binding Proteins/metabolism , Receptors, Lymphocyte Homing/metabolism , Signal Transduction/genetics , Tensins/metabolism , Transcriptome , Transfection , Up-Regulation/genetics
19.
World J Urol ; 38(3): 709-716, 2020 Mar.
Article En | MEDLINE | ID: mdl-31302753

PURPOSE: Emerging evidence has shown that macrophages (Mφs) at different tumor sites have diverse clinical attributes. Stabilin-1 is a multi-functional scavenger marker for specialized tumor-associated Mφs. This study investigates the relationship between the density and microlocalization of stabilin-1+ Mφs within tumors and the clinical outcomes of patients with urothelial carcinoma of the bladder (UCB). METHODS: In this retrospective study, 283 UCB patients who received radical cystectomy or transurethral resection were examined. Immunohistochemistry and immunofluorescence analyses were used to colocalize the expression of stabilin-1 with other markers for Mφs (CD14, CD68, CD163, and CD206). Kaplan-Meier analysis and Cox proportional hazards regression models were applied to estimate overall survival (OS) and recurrence-free survival (RFS). RESULTS: In UCB tissues, stabilin-1 was primarily expressed on Mφs, as evident from triple immunofluorescence staining for stabilin-1 and Mφ markers. Stabilin-1+ Mφs were often more prominent in stromal regions rather than intratumoral regions in UCB tissues (P < 0.0001). After dichotomization at the median cell density for stabilin-1+ Mφs, only intratumoral stabilin-1+ Mφ density was a predictor of poor OS (P < 0.001) and RFS (P = 0.026). Moreover, intratumoral stabilin-1+ Mφ density was positively associated with tumor stage (P < 0.01) and histological grade (P < 0.01), and emerged as an independent prognostic factor for OS (HR 2.371; P < 0.0001), but not for RFS (HR 1.491; P = 0.061). CONCLUSIONS: Our findings indicate that intratumoral stabilin-1+ Mφs could potentially be used as a pro-tumoral prognostic marker for UCB patients.


Carcinoma, Transitional Cell/pathology , Tumor-Associated Macrophages/pathology , Urinary Bladder Neoplasms/pathology , Aged , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Carcinoma, Transitional Cell/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Disease-Free Survival , Female , Humans , Lectins, C-Type/metabolism , Lipopolysaccharide Receptors/metabolism , Male , Mannose Receptor , Mannose-Binding Lectins/metabolism , Middle Aged , Neoplasm Grading , Neoplasm Staging , Receptors, Cell Surface/metabolism , Receptors, Lymphocyte Homing/metabolism , Retrospective Studies , Risk Factors , Survival Rate , Tumor Microenvironment , Tumor-Associated Macrophages/cytology , Tumor-Associated Macrophages/metabolism , Urinary Bladder Neoplasms/metabolism
20.
Viruses ; 11(9)2019 08 29.
Article En | MEDLINE | ID: mdl-31470568

Porcine reproductive and respiratory syndrome virus (PRRSV) continues to cause severe reproductive and respiratory pathologies resulting in immense monetary and welfare costs for the swine industry. The vaccines against PRRSV are available; but they struggle with providing protection against the plethora of heterologous PRRSV strains. To improve PRRSV vaccine development, the aim of this study was to provide an in-depth analysis of the crucial heterologous T-cell response to type-2 PRRSV. Following PRRSV modified live virus (MLV) vaccination or infection using one high- or one low-pathogenic PRRSV-strain, this nine-week study evaluated the T-cell response to different PRRSV strains. Our results demonstrate an important role for T cells in this homo- and heterologous response. Specifically, the T-helper cells were the main responders during viremia. Their peak response at 28 dpi correlated with a reduction in viremia, and their homing receptor expression indicated the additional importance for the anti-PRRSV response in the lymphatic and lung tissue. The cytocoxic T lymphocyte (CTL) response was the strongest at the site of infection-the lung and bronchoalveolar lavage. The TCR-γδ T cells were the main responders post viremia and PRRSV induced their expression of the lymph node homing the chemokine receptor, CCR7: This indicates a crucial role for TCR-γδ T cells in the anti-PRRSV response in the lymphatic system.


Porcine Reproductive and Respiratory Syndrome/immunology , Porcine respiratory and reproductive syndrome virus/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Adaptive Immunity , Animals , Kinetics , Lung/immunology , Lung/virology , Lymph Nodes/immunology , Lymph Nodes/virology , Porcine Reproductive and Respiratory Syndrome/pathology , Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus/growth & development , Porcine respiratory and reproductive syndrome virus/pathogenicity , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Lymphocyte Homing/metabolism , Swine , T-Lymphocyte Subsets/virology , T-Lymphocytes/virology , Vaccines, Attenuated , Viral Vaccines/immunology , Viremia/immunology , Viremia/virology
...