Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 17(8): 1739-1751, 2018 08.
Article in English | MEDLINE | ID: mdl-29743205

ABSTRACT

CD47, an ubiquitously expressed innate immune checkpoint receptor that serves as a universal "don't eat me" signal of phagocytosis, is often upregulated by hematologic and solid cancers to evade immune surveillance. Development of CD47-targeted modalities is hindered by the ubiquitous expression of the target, often leading to rapid drug elimination and hemotoxicity including anemia. To overcome such liabilities, we have developed a fully human bispecific antibody, NI-1701, designed to coengage CD47 and CD19 selectively on B cells. NI-1701 demonstrates favorable elimination kinetics with no deleterious effects seen on hematologic parameters following single or multiple administrations to nonhuman primates. Potent in vitro and in vivo activity is induced by NI-1701 to kill cancer cells across a plethora of B-cell malignancies and control tumor growth in xenograft mouse models. The mechanism affording maximal tumor growth inhibition by NI-1701 is dependent on the coengagement of CD47/CD19 on B cells inducing potent antibody-dependent cellular phagocytosis of the targeted cells. NI-1701-induced control of tumor growth in immunodeficient NOD/SCID mice was more effective than that achieved with the anti-CD20 targeted antibody, rituximab. Interestingly, a synergistic effect was seen when tumor-implanted mice were coadministered NI-1701 and rituximab leading to significantly improved tumor growth inhibition and regression in some animals. We describe herein, a novel bispecific antibody approach aimed at sensitizing B cells to become more readily phagocytosed and eliminated thus offering an alternative or adjunct therapeutic option to patients with B-cell malignancies refractory/resistant to anti-CD20-targeted therapy. Mol Cancer Ther; 17(8); 1739-51. ©2018 AACR.


Subject(s)
Antibodies, Bispecific/genetics , Leukemia/genetics , Leukemia/therapy , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/therapy , Animals , Antigens, CD19 , CD47 Antigen , Humans , Leukemia/pathology , Lymphoma, B-Cell/pathology , Mice , Xenograft Model Antitumor Assays
2.
Antibodies (Basel) ; 7(1)2018 Jan 03.
Article in English | MEDLINE | ID: mdl-31544856

ABSTRACT

CD47 serves as an anti-phagocytic receptor that is upregulated by cancer to promote immune escape. As such, CD47 is the focus of intense immuno-oncology drug development efforts. However, as CD47 is expressed ubiquitously, clinical development of conventional drugs, e.g., monoclonal antibodies, is confronted with patient safety issues and poor pharmacology due to the widespread CD47 "antigen sink". A potential solution is tumor-directed blockade of CD47, which can be achieved with bispecific antibodies (biAbs). Using mouse CD47-blocking biAbs in a syngeneic tumor model allowed us to evaluate the efficacy of tumor-directed blockade of CD47 in the presence of the CD47 antigen sink and a functional adaptive immune system. We show here that CD47-targeting biAbs inhibited tumor growth in vivo, promoting durable antitumor responses and stimulating CD8+ T cell activation in vitro. In vivo efficacy of the biAbs could be further enhanced when combined with chemotherapy or PD-1/PD-L1 immune checkpoint blockade. We also show that selectivity and pharmacological properties of the biAb are dependent on the affinity of the anti-CD47 arm. Taken together, our study validates the approach to use CD47-blocking biAbs either as a monotherapy or part of a multi-drug approach to enhance antitumor immunity.

3.
Transl Res ; 180: 37-52.e2, 2017 02.
Article in English | MEDLINE | ID: mdl-27559680

ABSTRACT

Life-threatening cytokine release syndromes include primary (p) and secondary (s) forms of hemophagocytic lymphohistiocytosis (HLH). Below detection in healthy individuals, interferon γ (IFNγ) levels are elevated to measurable concentrations in these afflictions suggesting a central role for this cytokine in the development and maintenance of HLH. Mimicking an infection-driven model of sHLH in mice, we observed that the tissue-derived levels of IFNγ are actually 500- to 2000-fold higher than those measured in the blood. To identify a blood biomarker, we postulated that the IFNγ gene products, CXCL9 and CXCL10 would correlate with disease parameters in the mouse model. To translate this into a disease relevant biomarker, we investigated whether CXCL9 and CXCL10 levels correlated with disease activity in pediatric sHLH patients. Our data demonstrate that disease control in mice correlates with neutralization of IFNγ activity in tissues and that the 2 chemokines serve as serum biomarkers to reflect disease status. Importantly, CXCL9 and CXCL10 levels in pediatric sHLH were shown to correlate with key disease parameters and severity in these patients. Thus, the translatability of the IFNγ-biomarker correlates from mouse to human, advocating the use of serum CXCL9 or CXCL10 as a means to monitor total IFNγ activity in patients with sHLH.


Subject(s)
Interferon-gamma/blood , Lymphohistiocytosis, Hemophagocytic/blood , Animals , Biomarkers/blood , Chemokine CXCL10/blood , Chemokine CXCL9/blood , Child , Disease Models, Animal , Female , Humans , Lymphohistiocytosis, Hemophagocytic/pathology , Mice, Inbred C57BL , Neutralization Tests , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/pharmacology , Syndrome
4.
Ann Rheum Dis ; 75(6): 1177-86, 2016 06.
Article in English | MEDLINE | ID: mdl-26698852

ABSTRACT

BACKGROUND: Interferon γ (IFNγ) is considered a seminal cytokine in the pathogenesis of giant cell arteritis (GCA), but its functional role has not been investigated. We explored changes in infiltrating cells and biomarkers elicited by blocking IFNγ with a neutralising monoclonal antibody, A6, in temporal arteries from patients with GCA. METHODS: Temporal arteries from 34 patients with GCA (positive histology) and 21 controls were cultured on 3D matrix (Matrigel) and exposed to A6 or recombinant IFNγ. Changes in gene/protein expression were measured by qRT-PCR/western blot or immunoassay. Changes in infiltrating cells were assessed by immunohistochemistry/immunofluorescence. Chemotaxis/adhesion assays were performed with temporal artery-derived vascular smooth muscle cells (VSMCs) and peripheral blood mononuclear cells (PBMCs). RESULTS: Blocking endogenous IFNγ with A6 abrogated STAT-1 phosphorylation in cultured GCA arteries. Furthermore, selective reduction in CXCL9, CXCL10 and CXCL11 chemokine expression was observed along with reduction in infiltrating CD68 macrophages. Adding IFNγ elicited consistent opposite effects. IFNγ induced CXCL9, CXCL10, CXCL11, CCL2 and intracellular adhesion molecule-1 expression by cultured VSMC, resulting in increased PBMC chemotaxis/adhesion. Spontaneous expression of chemokines was higher in VSMC isolated from GCA-involved arteries than in those obtained from controls. Incubation of IFNγ-treated control arteries with PBMC resulted in adhesion/infiltration by CD68 macrophages, which did not occur in untreated arteries. CONCLUSIONS: Our ex vivo system suggests that IFNγ may play an important role in the recruitment of macrophages in GCA by inducing production of specific chemokines and adhesion molecules. Vascular wall components (ie, VSMC) are mediators of these functions and may facilitate progression of inflammatory infiltrates through the vessel wall.


Subject(s)
Chemokines, CXC/metabolism , Giant Cell Arteritis/immunology , Interferon-gamma/antagonists & inhibitors , Macrophages/immunology , Aged , Aged, 80 and over , Cells, Cultured , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Chemokine CXCL11/genetics , Chemokine CXCL11/metabolism , Chemokine CXCL9/genetics , Chemokine CXCL9/metabolism , Chemokines, CXC/genetics , Chemotaxis/immunology , Down-Regulation/immunology , Female , Gene Expression Regulation/drug effects , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/pharmacology , Male , Muscle, Smooth, Vascular/immunology , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Temporal Arteries/immunology , Tissue Culture Techniques
5.
J Immunol ; 192(4): 1641-50, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24442438

ABSTRACT

B cells play a major role in the pathogenesis of many autoimmune disorders, including rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, and type I diabetes mellitus, as indicated by the efficacy of B cell-targeted therapies in these diseases. Therapeutic effects of the most commonly used B cell-targeted therapy, anti-CD20 mAb, are contingent upon long-term depletion of peripheral B cells. In this article, we describe an alternative approach involving the targeting of CD79, the transducer subunit of the B cell AgR. Unlike anti-CD20 mAbs, the protective effects of CD79-targeted mAbs do not require cell depletion; rather, they act by inducing an anergic-like state. Thus, we describe a novel B cell-targeted approach predicated on the induction of B cell anergy.


Subject(s)
Autoimmune Diseases/prevention & control , B-Lymphocytes/immunology , CD79 Antigens/immunology , Clonal Anergy/immunology , Animals , Antibodies, Monoclonal/immunology , Autoimmunity/immunology , Female , Lymphocyte Activation/immunology , Lymphocyte Count , Lymphocyte Depletion , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout
6.
J Biol Chem ; 287(2): 1458-67, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22041899

ABSTRACT

Dual-specific antibodies are characterized by an antigen-combining site mediating specific interactions with two different antigens. We have generated five dual-specific single chain variable fragments (scFv) that neutralize the activity of the two chemokines, CXCL9 and CXCL10, to bind to their receptor CXCR3. To better understand how these dual-specific scFvs bind these two chemokines that only share a 37% sequence identity, we mapped their epitopes on human CXCL9 and CXCL10 and identified serine 13 (Ser(13)) as a critical residue. It is conserved between the two chemokines but not in the third ligand for CXCR3, CXCL11. Furthermore, Ser(13) is exposed in the tetrameric structure of CXCL10, which is consistent with our finding that the scFvs are able to bind to CXCL9 and CXCL10 immobilized on glycosaminoglycans. Overall, the data indicate that these dual-specific scFvs bind to a conserved surface involved in CXCR3 receptor interaction for CXCL10 and CXCL9. Thus, structural mimicry between the two targets is likely to be responsible for the observed dual specificity of these antibody fragments.


Subject(s)
Antibody Specificity , Chemokine CXCL10/chemistry , Chemokine CXCL9/chemistry , Molecular Mimicry , Single-Chain Antibodies/chemistry , Animals , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Chemokine CXCL11/chemistry , Chemokine CXCL11/genetics , Chemokine CXCL11/immunology , Chemokine CXCL9/genetics , Chemokine CXCL9/immunology , Humans , Macaca fascicularis , Macaca mulatta , Mice , Rabbits , Receptors, CXCR3/chemistry , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology
7.
J Immunol ; 185(9): 5512-21, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20870936

ABSTRACT

IL-6-mediated T cell-driven immune responses are associated with signaling occurring through the membrane-bound cognate receptor α-chain (mIL-6Rα). Once formed, IL-6-mIL-6Rα complexes induce the homodimerization and subsequent phosphorylation of the ubiquitously expressed signal-transducing protein, gp130. This signaling event is defined as classical IL-6 signaling. However, many inflammatory processes assigned to IL-6 may be mediated via binding a naturally occurring soluble IL-6Rα, which forms an agonistic complex (IL-6/soluble IL-6Rα) capable of evoking responses on a wide range of cell types that lack mIL-6Rα (IL-6 trans-signaling). To dissect the differential contribution of the two IL-6 signaling pathways in cell-mediated inflammatory processes, we pharmaceutically targeted each using two murine models of human arthritis. Whereas intra-articular neutralization of trans-signaling attenuated local inflammatory responses, the classical pathway was found to be obligate and sufficient to induce pathogenic T cells and humoral responses, leading to systemic disease. Our data illustrate that mechanisms occurring in the secondary lymphoid organs underlying arthropathies are mediated via the classical pathway of IL-6 signaling, whereas trans-signaling contributes only at the local site, that is, in the affected tissues.


Subject(s)
Arthritis, Experimental/immunology , Autoimmunity/immunology , Interleukin-6/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Animals , Arthritis, Experimental/metabolism , Cell Separation , Cytokines/biosynthesis , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression , Interleukin-6/metabolism , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Receptors, Interleukin-6/immunology , Receptors, Interleukin-6/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Surface Plasmon Resonance , Transfection
8.
Immunity ; 33(1): 84-95, 2010 Jul 23.
Article in English | MEDLINE | ID: mdl-20643339

ABSTRACT

Germinal centers (GCs) are specialized microenvironments where antigen-activated B cells undergo proliferation, immunoglobulin (Ig) class switch recombination, somatic hypermutation (SHM), and affinity maturation. Within GCs, follicular dendritic cells (FDCs) are key players in driving these events via direct interaction with GC B cells. Here, we provide in vivo evidence that FDCs express and upregulate Toll-like-receptor (TLR) 4 in situ during germinal center reactions, confirm that their maturation is driven by TLR4, and associate the role of FDC-expressed TLR4 with quantitative and qualitative affects of GC biology. In iterative cycles of predictions by in silico modeling subsequently verified by in vivo experiments, we demonstrated that TLR4 signaling modulates FDC activation, strongly impacting SHM and generation of Ig class-switched high-affinity plasma and memory B cells. Thus, our data place TLR4 in the heart of adaptive humoral immunity, providing further insight into mechanisms driving GCs arising in both health and disease.


Subject(s)
B-Lymphocytes/metabolism , Dendritic Cells, Follicular/metabolism , Germinal Center/metabolism , Toll-Like Receptor 4/metabolism , Animals , Antibodies, Blocking , Antibody Affinity , Antigens, Differentiation/biosynthesis , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Bone Marrow Transplantation , Cell Differentiation/genetics , Cell Differentiation/immunology , Dendritic Cells, Follicular/pathology , Germinal Center/pathology , Immunoglobulin Class Switching/genetics , Immunoglobulin Class Switching/immunology , Immunologic Memory , Ligands , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mutation/genetics , Radiation Chimera , Signal Transduction/immunology , Somatic Hypermutation, Immunoglobulin/genetics , Somatic Hypermutation, Immunoglobulin/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology
9.
J Immunol ; 185(4): 2544-54, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20644170

ABSTRACT

Chemokines are key regulators of leukocyte trafficking and play a crucial role under homeostatic and inflammatory conditions. Because chemokines are involved in multiple pathologies, they represent an attractive class of therapeutic targets. However, because of the redundancy of this system, neutralizing a single chemokine may be insufficient to achieve therapeutic benefit. Our strategy was to use a Fc-fusion recombinant protein form of the poxvirus-derived viral CC chemokine inhibitor protein (vCCI-Fc) that has the ability to specifically bind to multiple CC chemokines and neutralize their activity. In this study, we demonstrate first that, in vivo, vCCI-Fc prevents CC chemokine-dependent migration of macrophages into inflamed tissue of carageenan-challenged mice. We next studied this effect of inhibiting CC chemokine activity in a model more relevant to human disease, collagen-induced arthritis. Mice receiving vCCI-Fc revealed a striking retention of splenocytes, including activated and IFN-gamma-secreting CD4(+) and CD8(+) T cells, that was associated with a concomitant decrease of cells in the draining lymph nodes. These phenomena resulted in a significant decrease in the incidence of disease and a reduction in clinical score, joint inflammation, and cartilage destruction as compared with mice receiving isotype control. Taken together, these results define a role for CC chemokines in the control of disease, as interfering with their function leads to a previously unappreciated role of controlling inflammatory cell trafficking in and out of secondary lymphoid organs.


Subject(s)
Arthritis, Experimental/immunology , Chemokines, CC/immunology , Inflammation/immunology , Recombinant Fusion Proteins/immunology , Animals , Arthritis, Experimental/prevention & control , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/immunology , Chemokines, CC/antagonists & inhibitors , Chemokines, CC/metabolism , Female , Flow Cytometry , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Inflammation/prevention & control , Interferon-gamma/immunology , Interferon-gamma/metabolism , Lymphocyte Count , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Protein Binding , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Spleen/cytology , Spleen/immunology , Spleen/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Vaccinia virus/genetics , Vaccinia virus/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
10.
MAbs ; 1(3): 288-96, 2009.
Article in English | MEDLINE | ID: mdl-20069756

ABSTRACT

Chemokines are important mediators of the immune response that are responsible for the trafficking of immune cells between lymphoid organs and migration towards sites of inflammation.Using phage display selection and a functional screening approach, we have isolated a panel of single-chain fragment variable (scFv) capable of neutralizing the activity of the human chemokine CXCL10 (hCXCL10). One of the isolated scFv was weakly cross-reactive against another human chemokine CXCL9,but was unable to block its biological activity. We diversified the complementarity determining region 3 (CDR3) of the light chain variable domain (VL) of this scFv and combined phage display with high throughput antibody array screening to identify variants capable of neutralizing both chemokines. Using this approach it is therefore possible to engineer pan-specific antibodies that could prove very useful to antagonize redundant signaling pathways such as the chemokine signaling network.


Subject(s)
Antibodies, Blocking/metabolism , Chemokine CXCL10/immunology , Chemokine CXCL9/immunology , Epitopes/metabolism , Single-Chain Antibodies/metabolism , Antibodies, Blocking/chemistry , Antibodies, Blocking/immunology , Antibody Affinity , Combinatorial Chemistry Techniques , Complementarity Determining Regions/chemistry , Cross Reactions , Epitopes/chemistry , Epitopes/immunology , High-Throughput Screening Assays , Humans , Immunoglobulin Light Chains/chemistry , Peptide Library , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/immunology
11.
J Exp Med ; 205(6): 1293-302, 2008 Jun 09.
Article in English | MEDLINE | ID: mdl-18490487

ABSTRACT

The secreted phosphatidylserine-binding protein milk fat globule epidermal growth factor 8 (Mfge8) mediates engulfment of apoptotic germinal center B cells by tingible-body macrophages (TBMphis). Impairment of this process can contribute to autoimmunity. We show that Mfge8 is identical to the mouse follicular dendritic cell (FDC) marker FDC-M1. In bone-marrow chimeras between wild-type and Mfge8(-/-) mice, all splenic Mfge8 was derived from FDCs rather than TBMphis. However, Mfge8(-/-) TBMphis acquired and displayed Mfge8 only when embedded in Mfge8(+/+) stroma, or when situated in lymph nodes draining exogenous recombinant Mfge8. These findings indicate a licensing role for FDCs in TBMphi-mediated removal of excess B cells. Lymphotoxin-deficient mice lacked FDCs and splenic Mfge8, and suffer from autoimmunity similar to Mfge8(-/-) mice. Hence, FDCs facilitate TBMphi-mediated corpse removal, and their malfunction may be involved in autoimmunity.


Subject(s)
Antigens, Surface/genetics , Apoptosis/physiology , Dendritic Cells, Follicular/physiology , Dendritic Cells/immunology , Macrophages/immunology , Milk Proteins/genetics , Animals , Antibodies/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , B-Lymphocytes/physiology , Bone Marrow Transplantation , Crosses, Genetic , DNA Primers , Dendritic Cells, Follicular/cytology , In Situ Hybridization , In Situ Nick-End Labeling , Leukocyte Common Antigens/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Milk Proteins/metabolism , RNA/genetics , Receptors, Complement 3d/immunology
12.
Nature ; 446(7131): 83-7, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17268470

ABSTRACT

Germinal centres are specialized structures wherein B lymphocytes undergo clonal expansion, class switch recombination, antibody gene diversification and affinity maturation. Three to four antigen-specific B cells colonize a follicle to establish a germinal centre and become rapidly dividing germinal-centre centroblasts that give rise to dark zones. Centroblasts produce non-proliferating centrocytes that are thought to migrate to the light zone of the germinal centre, which is rich in antigen-trapping follicular dendritic cells and CD4+ T cells. It has been proposed that centrocytes are selected in the light zone on the basis of their ability to bind cognate antigen. However, there have been no studies of germinal-centre dynamics or the migratory behaviour of germinal-centre cells in vivo. Here we report the direct visualization of B cells in lymph node germinal centres by two-photon laser-scanning microscopy in mice. Nearly all antigen-specific B cells participating in a germinal-centre reaction were motile and physically restricted to the germinal centre but migrated bi-directionally between dark and light zones. Notably, follicular B cells were frequent visitors to the germinal-centre compartment, suggesting that all B cells scan antigen trapped in germinal centres. Consistent with this observation, we found that high-affinity antigen-specific B cells can be recruited to an ongoing germinal-centre reaction. We conclude that the open structure of germinal centres enhances competition and ensures that rare high-affinity B cells can participate in antibody responses.


Subject(s)
B-Lymphocytes/cytology , Cell Movement , Germinal Center/cytology , Germinal Center/immunology , Animals , B-Lymphocytes/immunology , Cell Adhesion , Cell Communication , Mice , Mice, Inbred C57BL
13.
Eur J Immunol ; 36(7): 1665-73, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16783845

ABSTRACT

In this report, we show that the formation of germinal center (GC)-like structures to thymus-independent type 2 antigens in mice depends on intact signals through CD40, but does not depend on T cell-derived CD40-ligand (CD154). In addition, we show that follicular dendritic cells (FDC) are also critical to thymus-independent GC formation, as their depletion by blockade of lymphotoxin-beta receptor signals abrogated GC development unless the responding B cells bound antigen with high affinity. Further evidence that immune complexes drove this CD40-dependent B cell proliferation was provided by the observation that an antibody that detects immune complexes containing complement component 4 on FDC also inhibited thymus-independent GC formation when injected in vivo at the time of immunization. Finally, we show that thymus-independent B cell proliferation was associated with class switching to IgG3, as IgG3(+) antigen-specific switched B cells could be visualized directly in GC, suggesting that immune complexes can provide the signals for class switching within GC in the absence of CD154.


Subject(s)
Antigens, T-Independent/immunology , CD40 Antigens/physiology , CD40 Ligand/physiology , Germinal Center/immunology , Germinal Center/metabolism , Thymus Gland/immunology , Animals , CD40 Antigens/genetics , CD40 Ligand/genetics , Female , Immunoglobulin Class Switching/genetics , Immunoglobulin G/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism
14.
Eur J Immunol ; 35(5): 1501-9, 2005 May.
Article in English | MEDLINE | ID: mdl-15789355

ABSTRACT

Treatment (from 5 to 25 weeks of age) with a novel blocking monoclonal antibody, mAb I-10, directed against the plasma membrane (pm) form of LAMP-1, protected against development of autoimmune diabetes in the NOD mouse. A shorter course of treatment, i.e. from 5 to 12 weeks of age, significantly reduced the occurrence of insulitis as well as disease onset. Interfering with pm-LAMP-1 required continuous treatment as tolerance was not observed when treatment was stopped, and no higher proportion of cells with a T regulatory phenotype (e.g. CD4(+)CD25(+)) were induced. The mechanism appears to involve modulating a proinflammatory cytokine, as the proportion of pancreatic-infiltrating IFN-gamma-positive cells was significantly reduced in the mAb I-10-treated group. These results demonstrate an unexpected role for pm-LAMP-1 in autoimmune disease progression, and suggest that further investigation should be performed to understand how this molecule modulates IFN-gamma-driven responses.


Subject(s)
Antibodies, Blocking/immunology , Antibodies, Monoclonal/immunology , Antigens, CD/immunology , Diabetes Mellitus, Type 1/immunology , Membrane Proteins/immunology , Amino Acid Sequence , Animals , Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , Blotting, Western , Diabetes Mellitus, Type 1/prevention & control , Disease Models, Animal , Electrophoresis, Polyacrylamide Gel , Female , Flow Cytometry , Interferon-gamma/biosynthesis , Interferon-gamma/drug effects , Interferon-gamma/immunology , Lysosomal Membrane Proteins , Membrane Proteins/drug effects , Mice , Mice, Inbred NOD , Molecular Sequence Data , Pancreas/drug effects , Pancreas/immunology , Th1 Cells/drug effects , Th1 Cells/immunology
15.
J Immunol ; 173(9): 5776-85, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15494530

ABSTRACT

A hallmark of autoimmunity and other chronic diseases is the overexpression of chemokines resulting in a detrimental local accumulation of proinflammatory immune cells. Chemokines play a pivotal role in cellular recruitment through interactions with both cell surface receptors and glycosaminoglycans (GAGs). Anti-inflammatory strategies aimed at neutralizing the chemokine system have to-date targeted inhibition of the receptor-ligand interaction with receptor antagonists. In this study, we describe a novel strategy to modulate the inflammatory process in vivo through mutation of the essential heparin-binding site of a proinflammatory chemokine, which abrogates the ability of the protein to form higher-order oligomers, but retains receptor activation. Using well-established protocols to induce inflammatory cell recruitment into the peritoneal cavity, bronchoalveolar air spaces, and CNS in mice, this non-GAG binding variant of RANTES/CCL5 designated [44AANA47]-RANTES demonstrated potent inhibitory capacity. Through a combination of techniques in vitro and in vivo, [44AANA47]-RANTES appears to act as a dominant-negative inhibitor for endogenous RANTES, thereby impairing cellular recruitment, not through a mechanism of desensitization. [44AANA47]-RANTES is unable to form higher-order oligomers (necessary for the biological activity of RANTES in vivo) and importantly forms nonfunctional heterodimers with the parent chemokine, RANTES. Therefore, although retaining receptor-binding capacity, altering the GAG-associated interactive site of a proinflammatory chemokine renders it a dominant-negative inhibitor, suggesting a powerful novel approach to generate disease-modifying anti-inflammatory reagents.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Bronchoalveolar Lavage Fluid/immunology , Chemokine CCL5/antagonists & inhibitors , Chemokine CCL5/metabolism , Heparin/metabolism , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Peritoneum/immunology , Animals , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Binding Sites/genetics , Binding Sites/immunology , Bronchoalveolar Lavage Fluid/cytology , Cell Migration Inhibition , Chemokine CCL5/administration & dosage , Chemokine CCL5/genetics , Desensitization, Immunologic , Female , Humans , Inflammation Mediators/administration & dosage , Leukocytes/cytology , Leukocytes/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microscopy, Video , Mutation , Peritoneum/cytology , Peritoneum/metabolism , Rats , Rats, Sprague-Dawley
17.
Am J Pathol ; 164(4): 1253-62, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15039214

ABSTRACT

Wasting and renal diseases are frequent complications of HIV (human immunodeficiency virus) infection and are associated with accelerated disease progression and increased mortality. Transgenic mice expressing HIV1 under control of the CD4 promoter develop an AIDS-like disease and were used in the present work to study HIV1-induced wasting and kidney pathology. In this study, we reported that disease evolution paralleled increases in serum urea and creatinine levels, indicating an early and progressive deterioration of kidney function; meanwhile the wasting syndrome characterized by up-regulation of the ubiquitine-proteasome pathway and increased level of serum 3-methyl-histidine levels occurred at later stages just prior to death. Further examination of kidney and muscle pathologies revealed a progressive accumulation of CD45(+) cells, first affecting the kidneys. In addition, the onset of disease is accompanied by elevated levels of circulating "regulated on activation, normal and secreted T cell expressed and secreted" (RANTES). These results prompted us to assess the effects of AS602868, a specific small molecule inhibitor of IkappaB kinase 2 (IKK2) on disease progression. Inhibition of the NF-kappaB pathway indeed resulted in increased lifespan, kidney and lean body mass preservation. These beneficial results were associated with a reduction of CD45(+) cells infiltrating the kidneys, amelioration of the renal architecture, and reduced level of circulating RANTES. Together our data provide evidence that IKK2 inhibitors have therapeutic relevance in the treatment of HIV1-associated disorders.


Subject(s)
Acquired Immunodeficiency Syndrome/drug therapy , Enzyme Inhibitors/pharmacology , HIV Wasting Syndrome/drug therapy , Kidney Diseases/drug therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Acquired Immunodeficiency Syndrome/blood , Acquired Immunodeficiency Syndrome/complications , Animals , Blotting, Northern , Cells, Cultured , Chemokine CCL5/blood , Creatinine/blood , Disease Models, Animal , Female , HIV Wasting Syndrome/etiology , HIV-1 , Humans , I-kappa B Kinase , Kidney Diseases/blood , Kidney Diseases/etiology , Kidney Diseases/pathology , Leukocyte Common Antigens/metabolism , Leukocytes/metabolism , Male , Mice , Mice, Transgenic , Muscle, Skeletal/pathology , Urea/blood
18.
Nat Rev Immunol ; 3(9): 764-9, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12949500

ABSTRACT

Follicular dendritic cells (FDCs), which reside in the primary B-cell follicles and germinal centres of lymphoid tissues, can sequester antigen in the form of immune complexes and are thought to be pivotal to the germinal-centre reaction and the maintenance of immunological memory. But, many recent studies question the importance of FDCs and their bound immune complexes in B-cell responses. This article asks whether we can truly rule out a requirement for these cells in host defence.


Subject(s)
Dendritic Cells, Follicular/immunology , Animals , Antigen-Antibody Complex/immunology , Apoptosis/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Dendritic Cells, Follicular/cytology , Germinal Center/cytology , Germinal Center/immunology , Humans , Immunologic Memory/immunology , Receptors, Antigen, B-Cell/immunology
19.
J Immunol ; 171(3): 1164-71, 2003 Aug 01.
Article in English | MEDLINE | ID: mdl-12874202

ABSTRACT

Allergic contact dermatitis, the clinical manifestation of contact hypersensitivity, is one of the most common disorders of the skin. It is elicited upon multiple cutaneous re-exposure of sensitized individuals to the sensitizing agent. In this study, we demonstrate that using IL-18 binding protein (IL-18BP) to neutralize IL-18 significantly reduced clinical symptoms in a murine model of contact hypersensitivity. Furthermore, IL-18BP alleviated the relapses during established disease, as indicated by significant protection during re-exposure of mice that had previously undergone a contact hypersensitivity response without treatment. Although edema was not influenced, IL-18BP reduced the number of T cells homing to sites of inflammation, resulting in diminished local production of IFN-gamma. Thus, by preventing the accumulation of effector T cells to the target tissue, IL-18BP appears to be a potent protective mediator to counter skin inflammation during contact hypersensitivity. Taken together with the evidence that IL-18 is present in tissue samples of the human disease, our data reinforces IL-18BP as a candidate for this therapeutic indication.


Subject(s)
Dermatitis, Contact/immunology , Dermatitis, Contact/prevention & control , Glycoproteins/therapeutic use , Interleukin-18/metabolism , Administration, Cutaneous , Animals , Capillary Permeability/immunology , Cell Movement/immunology , Cells, Cultured , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Dermatitis, Contact/pathology , Dermatitis, Contact/physiopathology , Dinitrofluorobenzene/administration & dosage , Down-Regulation/immunology , Ear, External/immunology , Ear, External/pathology , Glycoproteins/administration & dosage , Haptens/administration & dosage , Humans , Inflammation Mediators/administration & dosage , Inflammation Mediators/therapeutic use , Intercellular Signaling Peptides and Proteins , Interleukin-18/antagonists & inhibitors , Interleukin-18/biosynthesis , Lymphocyte Count , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism
20.
Proc Natl Acad Sci U S A ; 100(4): 1885-90, 2003 Feb 18.
Article in English | MEDLINE | ID: mdl-12571364

ABSTRACT

During organogenesis, immunosurveillance, and inflammation, chemokines selectively recruit leukocytes by activating seven-transmembrane-spanning receptors. It has been suggested that an important component of this process is the formation of a haptotactic gradient by immobilization of chemokines on cell surface glycosaminoglycans (GAGs). However, this hypothesis has not been experimentally demonstrated in vivo. In the present study we investigated the effect of mutations in the GAG binding sites of three chemokines, monocyte chemoattractant protein-1/CC chemokine ligand (CCL)2, macrophage-inflammatory protein-1beta/CCL4, and RANTES/CCL5, on their ability to recruit cells in vivo. These mutant chemokines retain chemotactic activity in vitro, but they are unable to recruit cells when administered intraperitoneally. Additionally, monomeric variants, although fully active in vitro, are devoid of activity in vivo. These data demonstrate that both GAG binding and the ability to form higher-order oligomers are essential for the activity of particular chemokines in vivo, although they are not required for receptor activation in vitro. Thus, quaternary structure of chemokines and their interaction with GAGs may significantly contribute to the localization of leukocytes beyond migration patterns defined by chemokine receptor interactions.


Subject(s)
Chemokines/physiology , Glycosaminoglycans/metabolism , Animals , Base Sequence , Binding Sites , Biopolymers , CHO Cells , Chemokines/metabolism , Chemotaxis, Leukocyte , Cricetinae , DNA Primers , Female , Glycosaminoglycans/chemistry , In Vitro Techniques , Mice , Mice, Inbred BALB C , Peritoneal Cavity/cytology , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...