Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Clin Invest ; 120(8): 2767-81, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20628202

ABSTRACT

Studies in mice and humans have revealed that the T cell, immunoglobulin, mucin (TIM) genes are associated with several atopic diseases. TIM-1 is a type I membrane protein that is expressed on T cells upon stimulation and has been shown to modulate their activation. In addition to a recently described interaction with dendritic cells, TIM-1 has also been identified as a phosphatidylserine recognition molecule, and several protein ligands have been proposed. Our understanding of its activity is complicated by the possibility that TIM-1 possesses multiple and diverse binding partners. In order to delineate the function of TIM-1, we generated monoclonal antibodies directed to a cleft formed within the IgV domain of TIM-1. We have shown here that antibodies that bind to this defined cleft antagonize TIM-1 binding to specific ligands and cells. Notably, these antibodies exhibited therapeutic activity in a humanized SCID model of experimental asthma, ameliorating inflammation, and airway hyperresponsiveness. Further experiments demonstrated that the effects of the TIM-1-specific antibodies were mediated via suppression of Th2 cell proliferation and cytokine production. These results demonstrate that modulation of the TIM-1 pathway can critically influence activated T cells in a humanized disease model, suggesting that TIM-1 antagonists may provide potent therapeutic benefit in asthma and other immune-mediated disorders.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Asthma/prevention & control , Membrane Glycoproteins/antagonists & inhibitors , Membrane Proteins/antagonists & inhibitors , Receptors, Virus/antagonists & inhibitors , Animals , Asthma/immunology , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Female , Hepatitis A Virus Cellular Receptor 1 , Humans , Lymphocyte Activation , Membrane Glycoproteins/physiology , Membrane Proteins/physiology , Mice , Mice, SCID , Phosphatidylserines/metabolism , Receptors, Virus/physiology
2.
J Biol Chem ; 282(48): 34748-57, 2007 Nov 30.
Article in English | MEDLINE | ID: mdl-17911103

ABSTRACT

Alefacept is a chimeric protein combining CD58 immunoglobulin-like domain 1 with human IgG1 Fc. Alefacept mediates adhesion by bridging CD2 on T cells to activating Fc receptors on effector cells, but the equilibrium binding parameters have not been determined. Alefacept mediated T cell killing by NK cells and adhesion between CD2- and CD16-expressing cells at an optimum concentration of 100 nM. We introduce novel measurements with supported planer bilayers, from which key two-dimensional and three-dimensional parameters can be determined by data fitting. Alefacept competitively inhibited cell bilayer adhesion mediated by the CD2-CD58 interaction. Alefacept mediated maximal adhesion of CD2(+) T cells to CD16B, an Fc receptor, in planar bilayers at 500 nM. A mechanistic model for alefacept-mediated cell-bilayer adhesion allowed fitting of the data and determination of two-dimensional binding parameters. These included the density of bonds in the adhesion area, which grew to maintain a consistent average bond density of 200 molecules/microm(2) and two-dimensional association constants of 3.1 and 630 microm(2) for bivalently and monovalently bound forms of alefacept, respectively. The maximum number of CD16 bound and the fit value of 4,350 CD2 per cell are much lower than the 40,000 CD2 per cell measured with anti-CD2 Fab. These results suggest that additional information is needed to correctly predict Alefacept-mediated bridge formation.


Subject(s)
CD2 Antigens/biosynthesis , CD58 Antigens/chemistry , Receptors, IgG/biosynthesis , Recombinant Fusion Proteins/chemistry , Alefacept , Animals , CHO Cells , Cell Adhesion , Cricetinae , Cricetulus , Dermatologic Agents/chemistry , Dermatologic Agents/pharmacology , Dose-Response Relationship, Drug , Humans , Jurkat Cells , Kinetics , Lipid Bilayers/chemistry , Protein Binding , Receptors, Fc/chemistry , Receptors, IgG/chemistry , Recombinant Fusion Proteins/pharmacology , T-Lymphocytes/metabolism
3.
J Clin Invest ; 115(11): 3072-82, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16276416

ABSTRACT

Analysis of mononuclear cells in the adult mouse liver revealed that B cells represent as much as half of the intrahepatic lymphocyte population. Intrahepatic B cells (IHB cells) are phenotypically similar to splenic B2 cells but express lower levels of CD23 and CD21 and higher levels of CD5. IHB cells proliferate as well as splenic B cells in response to anti-IgM and LPS stimulation in vitro. VDJ gene rearrangements in IHB cells contain insertions of N,P region nucleotides characteristic of B cells maturing in the adult bone marrow rather than in the fetal liver. To evaluate whether B cells can have an impact on liver pathology, we compared CCl4-induced fibrosis development in B cell-deficient and wild-type mice. CCl4 caused similar acute liver injury in mutant and wild-type mice. However, following 6 weeks of CCl4 treatment, histochemical analyses showed markedly reduced collagen deposition in B cell-deficient as compared with wild-type mice. By analyzing mice that have normal numbers of B cells but lack either T cells or immunoglobulin in the serum, we established that B cells have an impact on fibrosis in an antibody- and T cell-independent manner.


Subject(s)
B-Lymphocyte Subsets/pathology , Liver Cirrhosis/pathology , Liver/pathology , Animals , B-Lymphocyte Subsets/metabolism , Base Sequence , Cells, Cultured , Collagen/biosynthesis , Liver/metabolism , Liver Cirrhosis/metabolism , Lymphopenia/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Rats , Rats, Sprague-Dawley , Spleen/cytology
4.
J Immunol ; 172(12): 7574-82, 2004 Jun 15.
Article in English | MEDLINE | ID: mdl-15187137

ABSTRACT

Type I IFNs (IFN-alphabeta) exert potent antiviral and immunoregulatory activities during viral infections, but their role in bacterial or protozoan infections is poorly understood. In this study, we demonstrate that the application of low, but not of high doses of IFN-beta protects 60 or 100% of BALB/c mice from progressive cutaneous and fatal visceral disease after infection with a high (10(6)) or low (10(4)) number of Leishmania major parasites, respectively. IFN-beta treatment of BALB/c mice restored the NK cell cytotoxic activity, increased the lymphocyte proliferation, and augmented the production of IFN-gamma and IL-12 in the draining lymph node. Low, but not high doses of IFN-beta caused enhanced tyrosine phosphorylation of STAT1 and STAT4, suppressed the levels of suppressor of cytokine signaling-1, and up-regulated the expression of inducible NO synthase in vivo. The IFN-beta-induced increase of IFN-gamma production was dependent on STAT4. Protection by IFN-beta strictly required the presence of inducible NO synthase. In the absence of STAT4 or IL-12, IFN-beta led to an amelioration of the cutaneous and visceral disease, but was unable to prevent its progression. These results identify IFN-beta as a novel cytokine with a strong, dose-dependent protective effect against progressive cutaneous leishmaniasis that results from IL-12- and STAT4-dependent as well as -independent events.


Subject(s)
Interferon-beta/pharmacology , Leishmaniasis/drug therapy , Animals , Cytotoxicity, Immunologic/drug effects , DNA-Binding Proteins/metabolism , Disease Progression , Dose-Response Relationship, Drug , Interferon-beta/administration & dosage , Interferon-gamma/biosynthesis , Interleukin-12/biosynthesis , Killer Cells, Natural/immunology , Leishmaniasis/prevention & control , Leishmaniasis, Cutaneous/prevention & control , Leishmaniasis, Visceral/prevention & control , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Mice, Knockout , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , STAT4 Transcription Factor , Trans-Activators/metabolism
5.
J Immunol ; 171(2): 547-51, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12847217

ABSTRACT

Germinal centers (GCs) form in B cell follicles and require specific signals for development and maintenance. B cell-activating factor belonging to the TNF family (BAFF) is a fundamental B cell survival factor and therefore may influence GC reactions and subsequent Ab responses. To test this possibility, the effect of BAFF neutralization in immunized mice was assessed. Using B cell maturation Ag-Fc, we demonstrate that BAFF blockade does not inhibit GC formation or somatic hypermutation. However, GCs in B cell maturation Ag-Fc-treated mice dissipated more rapidly than those of control mice and did not form a mature follicular dendritic cell reticulum. Examination of immunized BAFF-null mice validated the BAFF-independent nature of GC formation. Furthermore, Ab responses, including high-affinity responses, were attenuated. This is the first evidence that BAFF is required for maintenance, but not initiation, of the GC reaction, and it further hints that somatic hypermutation within the GC and selection of Ag-specific high-affinity Ab could be uncoupled.


Subject(s)
Germinal Center/immunology , Germinal Center/pathology , Membrane Proteins/deficiency , Membrane Proteins/physiology , Multigene Family/immunology , Receptors, Tumor Necrosis Factor/deficiency , Receptors, Tumor Necrosis Factor/genetics , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/physiology , Animals , B-Cell Activating Factor , B-Cell Maturation Antigen , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , B-Lymphocyte Subsets/pathology , Cell Differentiation/genetics , Cell Differentiation/immunology , Dendritic Cells, Follicular/immunology , Dendritic Cells, Follicular/metabolism , Dendritic Cells, Follicular/pathology , Down-Regulation/genetics , Down-Regulation/immunology , Female , Gene Frequency/immunology , Germinal Center/metabolism , Humans , Immunoglobulin G/biosynthesis , Injections, Intraperitoneal , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Multigene Family/genetics , Nitrophenols/administration & dosage , Nitrophenols/immunology , Phenylacetates , Receptors, Tumor Necrosis Factor/administration & dosage , Receptors, Tumor Necrosis Factor/immunology , Somatic Hypermutation, Immunoglobulin/genetics , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics
6.
Eur J Immunol ; 33(3): 666-75, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12616487

ABSTRACT

Modulation of the immune response using immunoglobulin fusion proteins has shown great promise for clinical immunotherapy of autoimmune diseases. Alefacept is an immunoglobulin fusion protein composed of the first extracellular domain of human LFA-3 fused to the hinge, C(H)2 and C(H)3 domains of human IgG(1). Alefacept has previously been reported to inhibit T cell proliferation. Here, we analyzed the effects of alefacept on lymphocytes in vitro and characterized the role of autologous NK cells in its mechanism of action. Alefacept, but not a C(H)2 binding mutant of Alefacept, inhibited CD3-induced T cell proliferation only in the presence of live NK cells, consistent with an important role for FcgammaR engagement. Alefacept caused preferential depletion of CD69+CD45R0+CD25+ T cell subsets. Cytotoxicity assays revealed that alefacept, but not the C(H)2 binding mutant, induced NK cell-mediated death of activated T cells and sorting into CD45R0+ and CD45RA+ subpopulations showed that lymphocyte deletion occurred preferentially in the CD45R0+ subset. Activated CD45R0+ cells expressed higher levels of CD2 than CD45R0- cells, providing a possible explanation for the selective targeting of this subset. Our results suggest that selective targeting of CD45R0+ T cells by NK cells represents a potential therapeutic mechanism of action of alefacept.


Subject(s)
Killer Cells, Natural/immunology , Leukocyte Common Antigens/analysis , Recombinant Fusion Proteins/pharmacology , T-Lymphocyte Subsets/drug effects , Alefacept , Antibody-Dependent Cell Cytotoxicity/drug effects , Cells, Cultured , DNA/biosynthesis , Female , Humans , Lymphocyte Activation/drug effects , Male , Recombinant Fusion Proteins/metabolism , T-Lymphocyte Subsets/immunology
7.
J Immunol ; 168(9): 4462-71, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11970990

ABSTRACT

Alefacept, an immunomodulatory recombinant fusion protein composed of the first extracellular domain of LFA-3 fused to the human IgG1 hinge, C(H)2, and C(H)3 domains, has recently been shown in phase II and III clinical trials to safely reduce disease expression in patients with chronic plaque psoriasis. Alefacept modulates the function of and selectively induces apoptosis of CD2(+) human memory-effector T cells in vivo. We have sought to gain further understanding of the mechanisms of action that influence the biological activity of alefacept and may contribute to its efficacy and patient responsiveness. Specifically evaluated is the ability of alefacept to activate intracellular signals mediated via CD2 and/or Fc gamma RIII (CD16). Experimentation using isoforms of alefacept engineered to have amino acid substitutions in the IgG1 C(H)2 domain that impact Fc gamma R binding indicate that alefacept mediates cognate interactions between cells expressing human CD2 and CD16 to activate cells, e.g., increase extracellular signal-regulated kinase phosphorylation, up-regulate cell surface expression of the activation marker CD25, and induce release of granzyme B. In the systems used, this signaling is shown to require binding to CD2 and CD16 and be mediated through CD16, but not CD2. Experimentation using human CD2-transgenic mice and isoforms of alefacept confirmed the requirement for Fc gamma R binding for detection of the pharmacological effects of alefacept in vivo. Thus alefacept acts as an effector molecule, mediating cognate interactions to activate Fc gamma R(+) cells (e.g., NK cells) to induce apoptosis of sensitive CD2(+) target cells.


Subject(s)
Adjuvants, Immunologic/pharmacology , CD2 Antigens/metabolism , Lymphocytes/immunology , Receptors, IgG/metabolism , Recombinant Fusion Proteins/pharmacology , Alefacept , Animals , Antibodies, Monoclonal/pharmacology , Apoptosis , CD2 Antigens/analysis , CD2 Antigens/genetics , Cells, Cultured , Dose-Response Relationship, Drug , Female , Gene Expression Profiling , Humans , Interleukin-2/pharmacology , Jurkat Cells , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lymphocyte Activation/drug effects , Lymphocytes/drug effects , Male , Mice , Mice, Transgenic , Receptors, IgG/genetics , Receptors, IgG/immunology , Signal Transduction , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , U937 Cells
8.
J Interferon Cytokine Res ; 22(2): 173-88, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11911800

ABSTRACT

We analyzed whether interferon-alpha 2b (IFN-alpha 2b) and IFN-beta 1a engage their common receptor to generate activated receptor complexes possessing distinct signaling properties. Human vascular endothelial cells (HUVEC) are 100-1000-fold more sensitive to IFN-beta 1a than to IFN-alpha 2b in in vitro assays. An nonarray-based expression profiling (GeneCalling) technology was employed to compare the patterns and levels of gene expression induced by these IFN as the broadest means by which signaling events could be measured. To distinguish subtype-related differences from dose-related effects, RNA was prepared from HUVEC treated with 50-5000 pg/ml of each IFN. The results showed that at 50 pg/ml IFN, only a subset of the genes induced by IFN-beta 1a were also induced by IFN-alpha 2b and that individual genes were induced to higher levels by IFN-beta 1a. In contrast, at 5000 pg/ml, both subtypes induced essentially identical sets of genes to similar levels of expression. No genes were seen to be induced uniquely by IFN-alpha 2b but not by IFN-beta 1a. The results show that the two IFN are intrinsically capable of inducing similar gene induction responses and do not provide evidence that they generate activated receptor complexes possessing distinct signaling properties. In contrast, the two IFN generate gene induction patterns that are both qualitatively and quantitatively distinct at subsaturating and potentially physiologically more relevant concentrations.


Subject(s)
Endothelium, Vascular/drug effects , Gene Expression Profiling/methods , Interferon Type I/physiology , Interferon-alpha/pharmacology , Interferon-beta/pharmacology , Receptors, Interferon/physiology , Umbilical Veins , Antineoplastic Agents/pharmacology , Cells, Cultured , Cluster Analysis , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Gene Expression/drug effects , Gene Expression Profiling/statistics & numerical data , Genes/drug effects , Humans , Interferon alpha-2 , Interferon beta-1a , Membrane Proteins , Receptor, Interferon alpha-beta , Recombinant Proteins , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...