Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Int J Radiat Oncol Biol Phys ; 119(1): 234-250, 2024 May 01.
Article in English | MEDLINE | ID: mdl-37981041

ABSTRACT

PURPOSE: Radiation therapy (RT) has been shown to effectively induce the expression of intercellular adhesion molecule-1 (ICAM-1), which is recognized by lymphocyte function-associated antigen 1 (LFA-1) expressed on natural killer (NK) cells. However, the potential synergistic antitumor immune response of tumor irradiation and administered NK cells has not been explored in intractable human liver cancers. Furthermore, NK cell targeting against both parental and cancer stemness has never been investigated. METHODS AND MATERIALS: Highly activated ex vivo NK cells were administered into the human liver tumor-bearing mice. Tumor direct RT was optimized according to tumor bearing site. HepG2 and Hep3B ICAM-1 knockout cells were generated using CRISPR/CAS9. Stemness tumor spheres were generated. NK cell cytolysis against parental and tumor sphere was evaluated using flow cytometry and real-time cytotoxicity assay. RESULTS: A combination of adoptive NK cell therapy with RT significantly improved therapeutic efficacy over monotherapies against subcutaneous, orthotopic, and metastatic human liver tumor models. Direct tumor irradiation potentiated NK cell recognition and conjugation against liver cancer through the LFA-1/ICAM-1 axis. Suppression of immune synapse formation on NK cells using high-affinity LFA-1 inhibitors or ICAM-1 knockout liver cancer induced "outside-in" signal blocking in NK cells, resulting in failure to eliminate liver tumor despite the combination therapy. NK cells effectively recognized and targeted triple-high epithelial cell adhesion molecule+CD133+CD24+ liver cancer expressing upregulated ICAM-1 in the irradiated tumor microenvironment, which led to prevention of the initiation of metastasis, improving survival in a metastatic model. In addition, the LFA-1/ICAM-1 axis interruption between NK cells and stemness liver tumor spheres significantly diminished NK cell cytolysis. Consistent with our preclinical data, the LFA-1/ICAM-1 axis correlated with survival outcomes in patients with metastatic cancer from the The Cancer Genome Atlas databases. CONCLUSIONS: NK cells in combination with tumor irradiation can provide synergistic therapeutic effects for NK cell recognition and elimination against both parental and stemlike liver cancer through LFA-1/ICAM-1.


Subject(s)
Intercellular Adhesion Molecule-1 , Liver Neoplasms , Humans , Mice , Animals , Lymphocyte Function-Associated Antigen-1/metabolism , Lymphocyte Function-Associated Antigen-1/pharmacology , Cytotoxicity, Immunologic , Killer Cells, Natural , Liver Neoplasms/metabolism , Parents , Tumor Microenvironment
2.
Nat Med ; 7(6): 687-92, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11385505

ABSTRACT

The beta2 integrin leukocyte function antigen-1 (LFA-1) has an important role in the pathophysiology of inflammatory and autoimmune diseases. Here we report that statin compounds commonly used for the treatment of hypercholesterolemia selectively blocked LFA-1-mediated adhesion and costimulation of lymphocytes. This effect was unrelated to the statins' inhibition of 3-hydroxy-3-methylglutaryl coenzyme-A reductase; instead it occurred via binding to a novel allosteric site within LFA-1. Subsequent optimization of the statins for LFA-1 binding resulted in potent, selective and orally active LFA-1 inhibitors that suppress the inflammatory response in a murine model of peritonitis. Targeting of the statin-binding site of LFA-1 could be used to treat diseases such as psoriasis, rheumatoid arthritis, ischemia/reperfusion injury and transplant rejection.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cell Adhesion/drug effects , Lovastatin/pharmacology , Lymphocyte Function-Associated Antigen-1/metabolism , Naphthalenes/pharmacology , Pravastatin/pharmacology , T-Lymphocytes/drug effects , Allosteric Site , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Disease Models, Animal , Drug Design , Humans , Integrin alpha4beta1 , Integrins/metabolism , Intercellular Adhesion Molecule-1/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Function-Associated Antigen-1/chemistry , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/pharmacology , Macrophage-1 Antigen/metabolism , Mice , Models, Molecular , Molecular Structure , Mutagenesis, Site-Directed , Naphthalenes/toxicity , Peritonitis/drug therapy , Protein Binding/drug effects , Receptors, Lymphocyte Homing/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/physiology
3.
Front Immunol ; 12: 680071, 2021.
Article in English | MEDLINE | ID: mdl-34975828

ABSTRACT

The trafficking of T-cells through peripheral tissues and into afferent lymphatic vessels is essential for immune surveillance and an adaptive immune response. Glycogen synthase kinase 3Ɵ (GSK3Ɵ) is a serine/threonine kinase and regulates numerous cell/tissue-specific functions, including cell survival, metabolism, and differentiation. Here, we report a crucial involvement of GSK3Ɵ in T-cell motility. Inhibition of GSK3Ɵ by CHIR-99021 or siRNA-mediated knockdown augmented the migratory behavior of human T-lymphocytes stimulated via an engagement of the T-cell integrin LFA-1 with its ligand ICAM-1. Proteomics and protein network analysis revealed ongoing interactions among GSK3Ɵ, the surface receptor Notch1 and the cytoskeletal regulator CRMP2. LFA-1 stimulation in T-cells reduced Notch1-dependent GSK3Ɵ activity by inducing phosphorylation at Ser9 and its nuclear translocation accompanied by the cleaved Notch1 intracellular domain and decreased GSK3Ɵ-CRMP2 association. LFA-1-induced or pharmacologic inhibition of GSK3Ɵ in T-cells diminished CRMP2 phosphorylation at Thr514. Although substantial amounts of CRMP2 were localized to the microtubule-organizing center in resting T-cells, this colocalization of CRMP2 was lost following LFA-1 stimulation. Moreover, the migratory advantage conferred by GSK3Ɵ inhibition in T-cells by CHIR-99021 was lost when CRMP2 expression was knocked-down by siRNA-induced gene silencing. We therefore conclude that GSK3Ɵ controls T-cell motility through interactions with CRMP2 and Notch1, which has important implications in adaptive immunity, T-cell mediated diseases and LFA-1-targeted therapies.


Subject(s)
Glycogen Synthase Kinase 3 beta/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Receptor, Notch1/metabolism , T-Lymphocytes/cytology , Adaptive Immunity , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Humans , Intercellular Adhesion Molecule-1/pharmacology , Lymphocyte Function-Associated Antigen-1/pharmacology , Phosphorylation , Protein Interaction Mapping , Protein Processing, Post-Translational , Pyridines/pharmacology , Pyrimidines/pharmacology , Recombinant Proteins/pharmacology , T-Lymphocytes/drug effects
4.
J Exp Med ; 174(1): 253-67, 1991 Jul 01.
Article in English | MEDLINE | ID: mdl-1676048

ABSTRACT

In an endeavor to further characterize human intercellular adhesion molecule-2 (ICAM-2), two murine monoclonal antibodies (mAb) were generated to ICAM-2 transfected COS cells, and designated CBR-IC2/1 and CBR-IC2/2. Immunoprecipitated, reduced ICAM-2 migrated as a broad band of Mr 60,000 in sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Treatment with N-glycanase revealed a peptide backbone of Mr 31,000, consistent with the size predicted from the cDNA. ICAM-2 had a broad distribution on hematopoietic cell lines and little expression on other cell lines, the sole exception being cultured endothelial cells which possess high levels of ICAM-2. Resting lymphocytes and monocytes expressed ICAM-2, while neutrophils did not. Staining of tissue sections with anti-ICAM-2 mAb confirmed their strong reactivity to vascular endothelium, but demonstrated a lack of ICAM-2 expression on other tissues. Small clusters of ICAM-2 positive cells were, however, seen in germinal centers. In contrast to ICAM-1 there was little or no induction of ICAM-2 expression on lymphocytes or cultured endothelium upon stimulation with inflammatory mediators. One of the two mAb, CBR-IC2/2, was found to totally inhibit binding of ICAM-2+ COS cells to purified lymphocyte function-associated antigen-1 (LFA-1). Using this mAb, LFA-1-dependent binding to both stimulated and unstimulated endothelium was found to be totally accounted for by ICAM-1 and ICAM-2. Homotypic aggregation of an Epstein-Barr virus-transformed B cell line, JY, was found to be solely ICAM-1 and ICAM-2-dependent, while in the case of the T cell lymphoma cell line, SKW3, anti- ICAM-2 mAb in conjunction with anti-ICAM-1 mAb could not inhibit the LFA-1-dependent aggregation. This suggests an additional LFA-1 ligand exists. Using a cell binding assay to purified LFA-1 in conjunction with anti-ICAM-1 and anti-ICAM-2 mAb, we have demonstrated that this putative third ligand for LFA-1 exists on SKW3 and other cell lines.


Subject(s)
Antigens, CD , Cell Adhesion Molecules/analysis , Lymphocyte Function-Associated Antigen-1/analysis , Animals , Antibodies, Monoclonal , Cell Adhesion , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Cell Aggregation/drug effects , Cell Line , Cells, Cultured , Endothelium, Vascular/cytology , Flow Cytometry , Humans , Intercellular Adhesion Molecule-1 , Lymphocyte Function-Associated Antigen-1/pharmacology , Mice , Monocytes/cytology , Organ Specificity , Tetradecanoylphorbol Acetate/pharmacology , Transfection
5.
ACS Infect Dis ; 4(7): 1073-1081, 2018 07 13.
Article in English | MEDLINE | ID: mdl-29742342

ABSTRACT

The Gram-negative bacterium Aggregatibacter actinomycetemcomitans, commonly associated with localized aggressive periodontitis (LAP), secretes an RTX (repeats-in-toxin) protein leukotoxin (LtxA) that targets human white blood cells, an interaction that is driven by its recognition of the lymphocyte function-associated antigen-1 (LFA-1) integrin. In this study, we report on the inhibition of LtxA-LFA-1 binding as an antivirulence strategy to inhibit LtxA-mediated cytotoxicity. Specifically, we designed and synthesized peptides corresponding to the reported LtxA binding domain on LFA-1 and characterized their capability to inhibit LtxA binding to LFA-1 and subsequent cytotoxic activity in human immune cells. We found that several of these peptides, corresponding to sequential Ɵ-strands in the LtxA-binding domain of LFA-1, inhibit LtxA activity, demonstrating the effectiveness of this approach. Further investigations into the mechanism by which these peptides inhibit LtxA binding to LFA-1 reveal a correlation between toxin-peptide affinity and LtxA-mediated cytotoxicity, leading to a diminished association between LtxA and LFA-1 on the cell membrane. Our results demonstrate the possibility of using target-based peptides to inhibit LtxA activity, and we expect that a similar approach could be used to hinder the activity of other RTX toxins.


Subject(s)
Anti-Bacterial Agents/pharmacology , Exotoxins/antagonists & inhibitors , Lymphocyte Function-Associated Antigen-1/chemistry , Peptides/pharmacology , Amino Acid Sequence , Anti-Bacterial Agents/chemistry , Exotoxins/chemistry , Exotoxins/toxicity , Humans , Lymphocyte Function-Associated Antigen-1/pharmacology , Models, Biological , Peptides/chemistry , Protein Binding , Structure-Activity Relationship , THP-1 Cells , Virulence Factors/antagonists & inhibitors , Virulence Factors/chemistry
6.
Methods Mol Biol ; 1584: 171-182, 2017.
Article in English | MEDLINE | ID: mdl-28255703

ABSTRACT

During antigen recognition by T cells, a specific spatial structure is formed at the contact face to an antigen-presenting cell (APC), called an immunological synapse (IS). The IS supports bidirectional signaling and release of effector molecules and is widely studied both biologically and numerically, in order to understand the process of T cell activation and signaling. This specialized structure harbors a central area (central supramolecular activation cluster, cSMAC) populated by T cell receptor-peptide-major histocompatibility complex (TCR-pMHC ) interactions, hedged by a peripheral ring (peripheral supramolecular activation cluster, pSMAC) of integrin lymphocyte function associated-1 interactions with its immunoglobulin superfamily ligand intercellular adhesion molecule-1 (LFA-1-ICAM-1). These two regions form the "bull's eye" pattern characteristic of the mature IS.In theoretical studies, different modeling architectures, including partial differential equations (PDE) and agent-based models , have been developed with the purpose to answer mechanistic questions about the IS dynamics. In this chapter, we explain possible physiological mechanisms that lead to the formation of ISs and technical issues that may occur in the course of development of agent-based models.


Subject(s)
Immunological Synapses/immunology , Intercellular Adhesion Molecule-1/immunology , Lymphocyte Function-Associated Antigen-1/pharmacology , Models, Immunological , Receptors, Antigen, T-Cell/immunology , Animals , Humans
7.
J Leukoc Biol ; 69(3): 458-66, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11261794

ABSTRACT

Lymphocyte infiltration in inflammation is induced by the dual actions of chemokines and cell adhesion molecules. The role of LFA-1 and VLA-4 in chemokine-induced T cell transendothelial migration (TEM) across cytokine-activated endothelium has not been examined. LFA-1, but not VLA-4, mediated blood T cell TEM to RANTES, macrophage inflammatory protein-1alpha (MIP-1alpha), and stromal cell-derived factor-1 (SDF-1), and across tumor necrosis factor alpha (TNF-alpha) or interferon-gamma (IFN-gamma) -stimulated endothelial cells (EC). Chemokine stimulation in combination with TNF-alpha activation of EC induced TEM, which was partially mediated by VLA-4. SDF-1 increased a beta1-integrin activation epitope on T cells and enhanced VLA-4-mediated adhesion. Thus, LFA-1 mediates TEM under most conditions, but VLA-4 can also mediate TEM, although, in contrast to LFA-1, this requires exogenous chemokines and EC activation. In addition, an LFA-1- and VLA-4-independent pathway of lymphocyte TEM can also be induced by SDF-1.


Subject(s)
Cell Movement/physiology , Cytokines/pharmacology , Endothelium, Vascular/cytology , Integrins/physiology , Lymphocyte Function-Associated Antigen-1/physiology , Receptors, Lymphocyte Homing/physiology , T-Lymphocytes/cytology , Cell Movement/immunology , Chemokine CCL3 , Chemokine CCL4 , Chemokine CCL5/pharmacology , Chemokine CXCL12 , Chemokines, CXC/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/immunology , Humans , Integrin alpha4beta1 , Integrins/biosynthesis , Interferon-gamma/pharmacology , Lymphocyte Function-Associated Antigen-1/pharmacology , Macrophage Inflammatory Proteins/pharmacology , Receptors, Lymphocyte Homing/biosynthesis , Recombinant Proteins , Stimulation, Chemical , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/pharmacology
8.
J Leukoc Biol ; 65(6): 778-85, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10380899

ABSTRACT

Membrane-permeable proteasome inhibitors, lactacystin (LC) and N-acetyl-Leu-Leu-norleucinal (ALLN), but not calpain inhibitor Z-Leu-leucinal (ZLL), prevented LFA-1/ICAM-1-dependent cellular adhesion of TPA-stimulated HL-60 cells. These proteasome inhibitors affected neither the induction of monocytic differentiation nor the accompanying protein-tyrosine phosphorylation. They suppressed the increase in the avidity of LFA-1 to ICAM-1 without changing the expression of these molecules. Immunoblotting using monoclonal antibody FK-1, which reacts specifically with polyubiquitinated proteins, demonstrated that the proteasome inhibitors caused the drastic accumulation of the polyubiquitinated proteins in the membrane fraction of TPA-treated HL-60 cells. This indicates that accompanying activation of LFA-1, TPA induces the polyubiquitination of the membrane proteins, which are rapidly degraded by proteasomes. These data taken together show that proteolysis mediated by the ubiquitin-proteasome system is a prerequisite for the induction of LFA-1-dependent adhesion of HL-60 cells.


Subject(s)
Cysteine Endopeptidases/physiology , HL-60 Cells/cytology , Intercellular Adhesion Molecule-1/pharmacology , Lymphocyte Function-Associated Antigen-1/pharmacology , Multienzyme Complexes/physiology , Ubiquitins/physiology , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Adenosine Triphosphatases/antagonists & inhibitors , Antibody Affinity/drug effects , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Cysteine Proteinase Inhibitors/pharmacology , HL-60 Cells/chemistry , HL-60 Cells/drug effects , Humans , Leupeptins/pharmacology , Lymphocyte Function-Associated Antigen-1/immunology , Monocytes/cytology , Proteasome Endopeptidase Complex , Tetradecanoylphorbol Acetate/pharmacology , Tretinoin/pharmacology
9.
Atherosclerosis ; 108(1): 73-81, 1994 Jul.
Article in English | MEDLINE | ID: mdl-7526875

ABSTRACT

Although the accumulation of monocyte-derived foam cells in the subendothelium is a key step in early atherogenesis, the mechanism responsible for monocyte adhesion to and subsequent transmigration through endothelial cells (ECs) has not been defined fully. We investigated the kinetics and the role played by adhesion molecules in the adhesion and transmigration of human monocytes using an in vitro three-dimensional model system comprising ECs cultured on collagen gels. Monocyte adhesion to untreated EC layers increased with time, reached a maximum after 3 h, and then declined. Monocyte transmigration through untreated EC layers also increased with time and reached a plateau after 3-4 h. Prestimulation of ECs with interleukin-1 beta (IL-1 beta; 25 U/ml) for 4 h enhanced monocyte adhesion (40.7 +/- 1.4%) and transmigration (37.9 +/- 1.6%) significantly compared with the value for untreated EC layers. In unstimulated EC layers, anti-leukocyte function-associated antigen-1 (LFA-1) plus anti-intercellular adhesion molecule-1 (ICAM-1) monoclonal antibodies (mAbs) inhibited monocyte adhesion and transmigration significantly by 19% and 20%, respectively, whereas anti-very late antigen-4 (VLA-4) plus anti-vascular cell adhesion molecule-1 (VCAM-1) mAbs did not. In IL-1 beta-stimulated EC layers, anti-LFA 1 plus anti-ICAM-1 mAbs inhibited the adhesion and transmigration by 32% and 30%, respectively and anti-VLA-4 plus anti-VCAM-1 mAbs did so by 18% and 27%, respectively. These results suggest that the monocyte-EC interaction in unstimulated ECs is mediated, in part, by the LFA-1-ICAM-1 pathway and in IL-1 beta-stimulated ECs, in part, by both LFA-1-ICAM-1 and VLA-4-VCAM-1 pathways.


Subject(s)
Cell Adhesion Molecules/physiology , Endothelium, Vascular/physiology , Monocytes/physiology , Antibodies, Monoclonal , Cell Adhesion/physiology , Cell Adhesion Molecules/pharmacology , Cell Movement/physiology , Cells, Cultured , Humans , Intercellular Adhesion Molecule-1/pharmacology , Intercellular Adhesion Molecule-1/physiology , Lymphocyte Function-Associated Antigen-1/pharmacology , Lymphocyte Function-Associated Antigen-1/physiology , Receptors, Very Late Antigen/physiology , Vascular Cell Adhesion Molecule-1
10.
Transplantation ; 68(5): 685-92, 1999 Sep 15.
Article in English | MEDLINE | ID: mdl-10507489

ABSTRACT

BACKGROUND: The counter receptors intercellular adhesion molecule (ICAM)-1 and lymphocyte function-associated antigen (LFA)-1 are lymphocyte cell surface adhesion proteins the interaction of which can provide signals for T cell activation. This binding event is important in T cell function, migration, and general immune system regulation. The ability to inhibit this interaction with monoclonal antibodies has proved to be therapeutically useful for several allograft rejection and autoimmune disease models. METHODS: Short peptides representing counter-receptor contact domains of LFA-1 and ICAM-1 were examined for their ability to inhibit T cell adhesion and T cell function. RESULTS: Peptides encompassing amino acids Q1-C21 and D26-K50 of ICAM-1, I237-I261 and G441-G466 of the LFA-1 alpha-subunit, and D134-Q159 of the LFA-1 beta-subunit inhibited LFA-1/ICAM-1-dependent adhesion in a phorbol-12,13-dibutyrate-induced model of tonsil T cell homotypic adhesion. This inhibition was specific to the peptide sequence and occurred without stimulation of T cell proliferation. The peptides also were effective in preventing T cell function using a one-way mixed lymphocyte reaction model for bone marrow transplantation. CONCLUSIONS: Our data suggest that these peptides or their derivatives may be useful as therapeutic modulators of LFA-1/ICAM-1 interaction during organ transplants.


Subject(s)
Intercellular Adhesion Molecule-1/pharmacology , Lymphocyte Culture Test, Mixed , Lymphocyte Function-Associated Antigen-1/pharmacology , Peptide Fragments/pharmacology , T-Lymphocytes/immunology , T-Lymphocytes/physiology , Animals , Antibody Formation/drug effects , COS Cells/metabolism , COS Cells/physiology , Cell Adhesion/drug effects , Cell Division/drug effects , Cell Survival/drug effects , Cells, Cultured , Humans , Intercellular Adhesion Molecule-1/genetics , Lymphocyte Count/drug effects , Transfection
11.
Immunobiology ; 195(3): 323-33, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8877406

ABSTRACT

We designed the present study to clarify the mechanism of superantigen-induced apoptosis of human mature T cells and to elucidate the pivotal roles of monocyte-derived macrophages in induction of T cell apoptosis. Exposure of unfractionated human peripheral blood mononuclear cells to SEA, SEB or PHA elicited apoptosis in T cells after 5-day culture. In purified T cell preparations, SEB was unable to induce apoptosis, but was inductive when the purified T cells were cocultured with monocyte-derived macrophages adhering to plastic culture dishes. Placing the T cells in the insert wells which physically separated them from the adhering macrophages resulted in a complete loss of SEB-induced apoptosis. The addition of blocking antibodies against LFA-1, ICAM-1 and CD2 to the cocultures significantly inhibited the SEB-induced T cell apoptosis. We concluded therefore that direct contact of macrophages with T cells is critical in SEB-induced T cell apoptosis, and that adhesion molecules such as LFA-1/ICAM-1 and CD2 may be involved in the mechanism of this effect.


Subject(s)
Apoptosis/drug effects , Apoptosis/immunology , Immunologic Memory/immunology , Intercellular Adhesion Molecule-1/pharmacology , Lymphocyte Function-Associated Antigen-1/pharmacology , Macrophages/physiology , T-Lymphocytes/immunology , CD2 Antigens/pharmacology , Cell Adhesion/immunology , Cells, Cultured , Humans , Ligands , Lymphocyte Activation/immunology , Macrophages/cytology , Superantigens/immunology , Superantigens/pharmacology
12.
Mol Cells ; 9(6): 662-7, 1999 Dec 31.
Article in English | MEDLINE | ID: mdl-10672935

ABSTRACT

Cell-cell adhesion is essential for the appropriate immune response, differentiation, and migration of lymphocytes. This important physiological event is reflected in vitro by homotypic cell aggregation. We have previously reported that a 120 kDa cell surface glycoprotein, JL1, is a unique protein specifically expressed by immature double positive (DP) human thymocytes which are in the process of positive and negative selections through the interaction between thymocyte and antigen-presenting cells (APCs). The function of the JL1 molecule, however, is yet to be identified. We show here that anti-JL1 monoclonal antibody (mAb) induced the homotypic aggregation of human thymocytes in a temperature- and Mg2+-dependent manner. It required an intact cytoskeleton and the interaction between leucocyte function associated antigen-1 (LFA-1) and intercellular adhesion molecule-1 (ICAM-1) since it was blocked by cytochalasin B and D, and mAb against LFA-1 and ICAM-1 which are known to be involved in the aggregation of thymocytes. Translocation of phosphatidylserine (PtdSer) through the cell membrane was not detected, implying that the molecular mechanism of JL-1-induced homotypic aggregation is different from that of CD99-induced homotypic aggregation. In summary, JL1 is a cell surface molecule that induces homotypic adhesion mediated by the LFA-1 and ICAM-1 interaction and cytoskeletal reorganization. These findings suggest that JL1 may be an important regulator of thymocyte development and thymocyte-APC interaction.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/metabolism , Cell Aggregation/drug effects , Intercellular Adhesion Molecule-1/pharmacology , Lymphocyte Function-Associated Antigen-1/pharmacology , T-Lymphocytes/metabolism , Antibodies, Monoclonal/immunology , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Cell Adhesion/drug effects , Flow Cytometry , Humans , Leukemia , Phosphatidylserines/metabolism , Signal Transduction/drug effects , Temperature , Tumor Cells, Cultured
13.
Inflammation ; 25(3): 203-14, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11403212

ABSTRACT

Tissue inflammation is characterized by aggravated leukocyte infiltration into the sites of inflammation. The mechanism requires the interactions of leukocyte adhesion-molecules and their ligands in the inflamed tissues. In this study, we demonstrate that a cyclic peptide cLAB.L [cyclol, 12-PenlTDGEATDSGC], derived from the "inserted" or I-domain of LFA-1 is able to inhibit the adherence of T-lymphocytes to the epithelial cell monolayers. This inhibition has been thought to involve the disruption of LFA-1/ICAM-1 interaction. The heterotypic adhesion of phorbol ester-activated Molt-3 cells and IFN-gamma-induced Caco-2 monolayers was inhibited upon treatment of the monolayers with monoclonal antibodies (MAbs) to adhesion molecules or with cLAB.L peptide. The adhesion can be inhibited by MAbs to ICAM-1, ICAM-2, and VCAM-1, and cLAB.L peptide in a concentration-dependent manner. However, none of the individual uses of these molecules led to a total inhibition. The inhibitory activity of cLAB.L is greatly reduced by low temperature and the absence of cell activation. Treatment of cLAB.L peptide may trigger an early event of apoptosis on activated but not on non-activated Molt-3 cells; no indication of peptide-induced apoptosis was found on Caco-2 cells. Taken together, data from this work suggest that cLAB.L may have applications to direct cell-targeted delivery during tissue inflammation.


Subject(s)
Cell Adhesion/drug effects , Lymphocyte Function-Associated Antigen-1/pharmacology , Peptides, Cyclic/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/physiology , Apoptosis/drug effects , Caco-2 Cells , Cell Line , Epithelial Cells/cytology , Epithelial Cells/drug effects , Humans , Inflammation/etiology , Intercellular Adhesion Molecule-1/physiology , Lymphocyte Function-Associated Antigen-1/chemistry , Lymphocyte Function-Associated Antigen-1/physiology , Oligopeptides/pharmacology , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Peptides, Cyclic/chemistry , Protein Structure, Tertiary , T-Lymphocytes/cytology , Temperature
14.
Pathol Res Pract ; 191(1): 1-7, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7651927

ABSTRACT

We examined the in vitro cytokine modulation of the interaction of cultured renal tubular epithelial cells (hKEC) with lymphoid effector cells, including mixed lymphocyte reaction (MLR)-derived blasts, Concanavalin A (Con A)-activated blast, and lymphokine-activated killer (LAK) cells. Blast adhesion to hKEC was augmented by treatment with either interferon-gamma (IFN-gamma), tumor necrosis factor-alpha (TNF-alpha), interleukin-1 beta (IL-1 beta), or MLR supernatant. The augmented adhesion statistically correlated with intercellular adhesion molecule-1 (ICAM-1) upregulation on hKEC cells by the cytokines. Blocking of surface ICAM-1 on hKEC or of lymphocyte-function antigen-1 (LFA-1) on the blasts significantly inhibited adhesion. LFA-1 blocking on LAK or MLR blasts resulted in a significant inhibition in cytotoxic function. TNF-alpha and IL-1 beta treatments on hKEC increased the susceptibility to LAK cytotoxicity, whereas IFN-gamma and MLR supernatant treatment significantly reduced this susceptibility. Absorption of IFN-gamma from the MLR supernatant partially restored the susceptibility of LAK cytotoxicity. The combined data suggest the importance of cytokine regulation of LFA-1/ICAM-1 adhesion molecule in these cellular interactions. Although IFN-gamma augments attachment between hKEC and blastic effector cells, this cytokine endows hKEC with resistance to LAK cytotoxicity. Thus, it is clear that cytokine modulation is a complicated phenomenon involving both adhesion molecule regulation-dependent and independent mechanisms.


Subject(s)
Cytokines/pharmacology , Kidney Tubules/cytology , T-Lymphocytes, Regulatory/cytology , Absorption , Antibodies, Monoclonal/immunology , Antigens, Surface/biosynthesis , Cell Adhesion/drug effects , Cell Communication/drug effects , Cell Communication/immunology , Cells, Cultured , Cytotoxicity Tests, Immunologic , Epithelial Cells , Humans , Intercellular Adhesion Molecule-1/pharmacology , Interferon-gamma/pharmacokinetics , Interferon-gamma/pharmacology , Killer Cells, Lymphokine-Activated/immunology , Lymphocyte Culture Test, Mixed/methods , Lymphocyte Function-Associated Antigen-1/pharmacology , Stem Cells/cytology
15.
J Dermatol ; 20(3): 144-50, 1993 Mar.
Article in English | MEDLINE | ID: mdl-8097520

ABSTRACT

Adhesion molecules such as intercellular adhesion molecule-1 (ICAM-1) and its counter-receptor, lymphocyte function associated antigen-1 (LFA-1), play very important roles in immune responses. In this study, the effects of cytokines on cultured human melanoma cells (MMG2) were examined, especially focussing on the expression of ICAM-1 on MMG2 and lymphocyte adhesion to MMG2. Both the expression of ICAM-1 and HLA-DR on MMG2 increased after treatment with IFN-gamma. ICAM-1 expression began to increase earlier than HLA-DR expression. TNF-alpha and IL-1 beta also increased the expression of ICAM-1 on MMG2. However, these cytokines did not increase the expression of HLA-DR. IFN-gamma had a dose dependent effect on lymphocyte adhesion to MMG2. Pretreatment of IFN-gamma treated MMG2 with 84H10 (anti-ICAM-1 antibody) or pretreatment of lymphocytes with either SPV-L7 (anti-LFA-1 alpha antibody) or IOT10 (anti-LFA-1 beta antibody) inhibited the lymphocyte adhesion to MMG2. These results suggest that ICAM-1 molecules induced on melanoma cells by IFN-gamma can interact with LFA-1 molecules on lymphocytes.


Subject(s)
Antigens, CD/analysis , Cell Adhesion Molecules/analysis , Cytokines/pharmacology , Melanoma/pathology , Skin Neoplasms/pathology , Antigens, CD/genetics , Cell Adhesion/drug effects , Cell Adhesion Molecules/genetics , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression/drug effects , HLA-DR Antigens/analysis , HLA-DR Antigens/genetics , Humans , Intercellular Adhesion Molecule-1 , Interferon Type I/pharmacology , Interferon-beta/pharmacology , Interferon-gamma/pharmacology , Interleukin-1/pharmacology , Interleukin-2/pharmacology , Lymphocyte Function-Associated Antigen-1/pharmacology , Lymphocytes/pathology , Recombinant Proteins , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology
17.
Gastroenterology ; 130(2): 482-92, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16472601

ABSTRACT

BACKGROUND & AIMS: The ability of viruses to escape the host immune response represents a globally important problem related to a wide variety of pathogens. Hepatitis C is one of the major causes of liver disease worldwide. Clearance rates of this virus are low, and this condition normally involves a chronic inflammatory process. This raises a possibility that the virus may have developed mechanisms enabling it to evade T-cell-mediated immune surveillance. The aim of this study was to investigate the effect of the hepatitis C envelope protein E2 on LFA-1-stimulated T-cell migration and macrophage inflammatory protein (MIP-1alpha, MIP-1beta) secretion. METHODS: T cells were stimulated through the leukocyte function-associated molecule-1 (LFA-1) receptor by incubating with either intracellular adhesion molecule 1 (ICAM-1)-Fc fusion protein or anti-LFA-1 immobilized on 8-well chamber slides. Subcellular localization of protein kinase C (PKC)-beta, CD81, and LFA-1 was determined by immunofluorescence analysis. Lipid raft formation was assessed using the Cellomics Kineticscan reader. MIP-1alpha and MIP-1beta levels were detected by enzyme-linked immunosorbent assay. RESULTS: We report that the hepatitis C envelope protein E2 can dramatically inhibit T-lymphocyte motility and chemokine release induced via LFA-1 integrin ligation. We have demonstrated a novel T-lymphocyte-directed viral inhibitory mechanism involving the PKC-beta enzyme as a definitive intracellular target. E2-CD81 interaction stimulates translocation of PKC-beta to lipid rafts, thereby preventing its association with the centrosome and microtubule cytoskeleton, which is crucial to the process of T-cell migration. CONCLUSIONS: These studies identify a mechanism whereby the hepatitis C virus can evade the host immune response by inhibition of T-cell migration.


Subject(s)
Hepacivirus , Lymphocyte Function-Associated Antigen-1/pharmacology , Protein Kinase C/metabolism , T-Lymphocytes/physiology , Viral Envelope Proteins/pharmacology , Cell Movement , Chemokines/metabolism , Humans , Lymphocyte Activation , Membrane Microdomains/physiology , Protein Kinase C beta , Signal Transduction , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tetradecanoylphorbol Acetate/pharmacology
18.
J Immunol ; 176(4): 2105-13, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16455966

ABSTRACT

Lymphokine gene expression is a precisely regulated process in T cell-mediated immune responses. In this study we demonstrate that engagement of the beta(2) integrin LFA-1 in human peripheral T cells markedly extends the half-life of TNF-alpha, GM-CSF, and IL-3 mRNA, as well as a chimeric beta-globin mRNA reporter construct containing a strongly destabilizing class II AU-rich element from the GM-CSF mRNA 3'-untranslated region. This integrin-enhanced mRNA stability leads to augmented protein production, as determined by TNF-alpha ELISPOT assays. Furthermore, T cell stimulation by LFA-1 promotes rapid nuclear-to-cytoplasmic translocation of the mRNA-stabilizing protein HuR, which in turn is capable of binding an AU-rich element sequence in vitro. Abrogation of HuR function by use of inhibitory peptides, or marked reduction of HuR levels by RNA interference, prevents LFA-1 engagement-mediated stabilization of T cell TNF-alpha or IFN-gamma transcripts, respectively. Thus, HuR-mediated mRNA stabilization, stimulated by integrin engagement and controlled at the level of HuR nuclear export, is critically involved in T cell activation.


Subject(s)
Antigens, Surface/metabolism , Cell Nucleus/metabolism , Cytokines/genetics , Lymphocyte Activation , Lymphocyte Function-Associated Antigen-1/pharmacology , RNA Stability , RNA-Binding Proteins/metabolism , T-Lymphocytes/metabolism , Active Transport, Cell Nucleus , Antigens, Surface/genetics , Base Sequence , CD28 Antigens/pharmacology , Cell Nucleus/drug effects , Cells, Cultured , Cytoplasm/drug effects , Cytoplasm/metabolism , ELAV Proteins , ELAV-Like Protein 1 , Humans , Lymphocyte Activation/drug effects , Lymphocyte Function-Associated Antigen-1/metabolism , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
19.
Int Immunol ; 7(5): 763-9, 1995 May.
Article in English | MEDLINE | ID: mdl-7547703

ABSTRACT

The mAb YTA-1, which brightly stains CD3-CD16+ large granular lymphocytes (LGL)/natural killer (NK) cells and CD8+ T cells by immunofluorescence, is specific for leukocyte function-associated antigen (LFA)-1. Some mAbs recognizing the LFA-1 alpha chain (CD11a) or LFA-1 beta chain (CD18) inhibited the binding of YTA-1 to peripheral blood mononuclear cells. YTA-1 mAb could be chemically cross-linked to 170 and 96 kDa molecules, whose molecular weights correspond to those of LFA-1 alpha and beta respectively. YTA-1 bound to COS-7 cells co-transfected with CD11a and CD18 cDNAs, but not to untransfected cells. Reactivities of YTA-1 to K562 cells transfected with LFA-1 alpha and beta (CD11a/CD18) cDNAs and to CHO cells transfected with Mac-1 (CD11b/CD18) or p150, 95 (CD11c/CD18) cDNAs strongly suggest that YTA-1 recognizes either LFA-1 alpha or an epitope formed by a combination of LFA-1 alpha and beta. Treatment of fresh CD3-CD16+ LGL with YTA-1 augmented cytolytic activity and induced proliferation. F(ab')2 fragments of YTA-1 augmented NK cytotoxicity, indicating that the NK activating signal was transmitted through LFA-1 without involvement of Fc gamma receptor III. In contrast, the other mAbs against LFA-1 could not activate NK cells. These results collectively indicate that YTA-1 recognizes a unique epitope of LFA-1, which is involved in activation of fresh NK cells.


Subject(s)
Antibodies, Monoclonal/immunology , Cytotoxicity, Immunologic , Killer Cells, Natural/immunology , Lymphocyte Activation , Lymphocyte Function-Associated Antigen-1/immunology , Animals , Antigen-Antibody Reactions , CD3 Complex/immunology , Humans , Lymphocyte Function-Associated Antigen-1/pharmacology , Mice , Mice, Inbred BALB C
20.
Br J Haematol ; 115(4): 862-71, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11843820

ABSTRACT

The bone marrow (BM) microenvironment supports leukaemia cell survival and proliferation. The roles played by adhesive receptor interactions in the survival of T-lineage acute lymphoblastic leukaemia (T-ALL) cells on BM stromal cells are not well understood. Recently, we have developed an assay that partially recapitulates the BM microenvironment using HS-5 BM stromal cells. In this assay, the magnitude of ex vivo T-ALL lymphoblast survival predicts patient outcome. We examined the molecular basis for cell-cell adhesive events leading to T-ALL lymphoblast survival on HS-5 and on donor-derived BM stroma. Lympho cyte function-associated antigen-1 (LFA-1) on T-ALL cell lines bound intercellular adhesion molecule-1 (ICAM-1) on HS-5 monolayers, and survival was inhibited 85-98% with monoclonal antibodies directed against LFA-1 or ICAM-1. We compared these results with patient-derived T-ALL lymphoblasts co-cultured on either HS-5 BM or normal BM monolayers and found that LFA-1 and ICAM-1 were required, but not alone sufficient for ex vivo leukaemic cell survival. On normal BM stroma, but not HS-5 monolayers, two additional adhesion molecules, vascular cell adhesion molecule-1 (VCAM-1) and E-selectin, were highly expressed and contributed to T-ALL cell survival. This is the first report to demonstrate the importance of LFA-1/ICAM-1-mediated adhesion as a critical event in a cascade of cell surface receptor-ligand interactions that regulate T-ALL survival in the BM microenvironment.


Subject(s)
Bone Marrow Cells/physiology , Intercellular Adhesion Molecule-1/pharmacology , Leukemia-Lymphoma, Adult T-Cell/pathology , Lymphocyte Function-Associated Antigen-1/pharmacology , Stromal Cells/physiology , Apoptosis , Cell Adhesion , Cell Line , Cell Survival , Cells, Cultured , Child , Humans , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL