Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
J Ethnopharmacol ; 324: 117745, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38228231

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Jin-Gui-Shen-Qi Wan (JGSQW) is a traditional Chinese medicine formula that has been traditionally used to alleviate urinary system ailments such as frequent urination and polyuria. Clinical studies have indicated that when combined with hypoglycaemic drugs, JGSQW exhibits a synergistic effect and can improve diabetic nephropathy (DN), yet its underlying mechanism and targets remain unclear. AIM OF THE STUDY: This study aims to investigate the therapeutic efficacy of JGSQW and its underlying mechanisms using a DN db/db mouse model. MATERIALS AND METHODS: Ultrahigh-performance liquid chromatography coupled with mass spectrometry was utilized to analyse the primary active compounds, blood levels, and pharmacokinetics of JGSQW. Additionally, the therapeutic effects of JGSQW and metformin on blood glucose levels, lipid levels, renal function, and renal pathology in diabetic nephropathy mice were investigated using a db/db mouse model. Proteomic analysis was carried out to identify the primary target of JGSQW in treating DN. The mechanism of action was verified by western blotting, immunohistochemistry, and immunofluorescence. Then, molecular docking and molecular dynamics, transfection, drug affinity responsive target stability (DARTS) assay and cell thermal migration assay (CETSA) further validated the targeted binding effect. RESULTS: JGSQW combined with metformin significantly improved the blood glucose levels, blood lipids, renal function, and renal pathology of DN mice. JGSQW mainly exerted its therapeutic effect on DN by targeting major histocompatibility complex class II (MHC class II) molecules. Immunohistochemistry results showed that JGSQW inhibited the expression of collagen I, fibronectin, and alpha smooth muscle actin (α-SMA) expression. Immunofluorescence and Western blot results showed that JGSQW inhibited the expression of H2-Ab1 and H2-Aa, which are MHC class II molecules, thereby suppressing CD4+ T-cell infiltration and improving diabetic kidney fibrosis. The binding ability of paeoniflorin to H2-Aa was predicted and verified by molecular, DARTS, and CETSA assays. Treatment with 80 µM paeoniflorin effectively alleviated high glucose-induced injury in the MPC-5 injury model. H2-Aa was overexpressed at this model concentration, and Western blotting further confirmed that paeoniflorin reduced glomerular podocyte fibrosis by regulating H2-Aa. CONCLUSIONS: JGSQW combined with metformin may have a synergistic effect to alleviates renal fibrosis in diabetic nephropathy by downregulating immune complex MHC class II molecules and attenuating the antigen presentation effect of MHC class II on CD4.


Subject(s)
Diabetes Mellitus , Diabetic Nephropathies , Glucosides , Metformin , Monoterpenes , Mice , Animals , Diabetic Nephropathies/pathology , Blood Glucose , Molecular Docking Simulation , Proteomics , Signal Transduction , Fibrosis , Histocompatibility Antigens Class II/pharmacology , Histocompatibility Antigens Class II/therapeutic use , Metformin/pharmacology , Metformin/therapeutic use
2.
Eur J Pediatr ; 176(9): 1163-1172, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28803259

ABSTRACT

Over the last 50 years, collaborative clinical trials have reduced the number of children dying from pediatric cancer significantly. Unfortunately, certain tumor types have remained resistant to conventional surgical, radiotherapy and chemotherapy combinations, and relapsing and/or refractory disease remains associated with dismal outcomes. Recently, renewed attention has been given to the role for immunotherapies in pediatric oncology. In fact, these combine several attractive features, including (but possibly not limited to) the specificity for cancer cells, potentially in vivo persistence and longevity, and potency against refractory disease. In this narrative review designed for the academic pediatrician, we will concisely review the biological underpinnings behind the immunological therapy of pediatric neoplasms and illustrate the current humoral, cellular approaches, and novel drugs targeting the immune checkpoint, oncolytic viruses, and tumor vaccines. We will also comment on the future directions, challenges, and open questions faced by the field. What is Known: • Cancer immunotherapy drives immune cells and its humoral weaponry to eliminate tumor cells. • This occurs by recognizing antigens ideally expressed only on tumoral, but not normal/healthy, cells. What is New: • Clinical immunotherapy trials have shown responses in children with relapsing/refractory neoplasms. • Novel humoral/cellular immunotherapies, immune checkpoint inhibitors, oncolytic viruses, and tumor vaccines are currently being investigated in pediatric oncology.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Immunotherapy/methods , Neoplasms/therapy , Adjuvants, Immunologic/therapeutic use , Antibodies, Monoclonal/therapeutic use , Cancer Vaccines/therapeutic use , Child , Histocompatibility Antigens Class II/therapeutic use , Humans , Immunotherapy/trends , Oncolytic Virotherapy , T-Lymphocytes/drug effects
5.
Int J Clin Exp Pathol ; 7(8): 4636-44, 2014.
Article in English | MEDLINE | ID: mdl-25197336

ABSTRACT

Dermatophagoides farinae (Der f 1) is one of leading cause for allergic asthma, and allergen-specific immunotherapy (SIT) is currently recognized as the only etiological therapy to ameliorate asthmatic symptom. The current study was designed on the major histocompatibility complex (MHC) class II pathway, invariant chain (Ii)-segment hybrids as vaccine basis to explore the efficacy of Der f 1 hybrid vaccine by virtue of Ii as carrier in enhancing the protective immune response to asthma. Initially, we engineered a fused molecule, DCP-IhC-ProDer f 1, to deliver ProDer f 1 antigen via specific dendritic cell-targeting peptides to dendritic cells (DCs). Then the DCP-IhC-ProDer f 1 was immunized to the asthmatic models of murine induced by ProDer f 1 allergen. The findings showed that the cytokine repertoire in the murine model was shifted after SIT, including stronger secretion of IFN-γ and IL-10, and a decreased production of IL-4 and IL-17. ELISA determination revealed that the hybrid displayed weak IgE and IgG1 reactivities, and IgG2a levels were elevated. Furthermore, DCP-IhC-ProDer f 1 treatment inhibited inflammatory cell infiltration in the lung tissues. Our results suggest that the DCP-Ihc-ProDer f 1 may be used as a candidate SIT against asthma.


Subject(s)
Antigens, Dermatophagoides/immunology , Antigens, Differentiation, B-Lymphocyte/immunology , Arthropod Proteins/immunology , Asthma/prevention & control , Cysteine Endopeptidases/immunology , Desensitization, Immunologic/methods , Histocompatibility Antigens Class II/immunology , Vaccines/immunology , Animals , Antigen Presentation/immunology , Antigens, Dermatophagoides/therapeutic use , Antigens, Differentiation, B-Lymphocyte/therapeutic use , Arthropod Proteins/therapeutic use , Blotting, Western , Cysteine Endopeptidases/therapeutic use , Disease Models, Animal , Female , Histocompatibility Antigens Class II/therapeutic use , Mice , Mice, Inbred BALB C , Polymerase Chain Reaction , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use
6.
Mol Ther ; 22(5): 1039-47, 2014 May.
Article in English | MEDLINE | ID: mdl-24476798

ABSTRACT

Despite viral vectors being potent inducers of antigen-specific T cells, strategies to further improve their immunogenicity are actively pursued. Of the numerous approaches investigated, fusion of the encoded antigen to major histocompatibility complex class II-associated invariant chain (Ii) has been reported to enhance CD8(+) T-cell responses. We have previously shown that adenovirus vaccine encoding nonstructural (NS) hepatitis C virus (HCV) proteins induces potent T-cell responses in humans. However, even higher T-cell responses might be required to achieve efficacy against different HCV genotypes or therapeutic effect in chronically infected HCV patients. In this study, we assessed fusion of the HCV NS antigen to murine and human Ii expressed by the chimpanzee adenovirus vector ChAd3 or recombinant modified vaccinia Ankara in mice and nonhuman primates (NHPs). A dramatic increase was observed in outbred mice in which vaccination with ChAd3 expressing the fusion antigen resulted in a 10-fold increase in interferon-γ(+) CD8(+) T cells. In NHPs, CD8(+) T-cell responses were enhanced and accelerated with vectors encoding the Ii-fused antigen. These data show for the first time that the enhancement induced by vector vaccines encoding li-fused antigen was not species specific and can be translated from mice to NHPs, opening the way for testing in humans.


Subject(s)
Antigens, Viral/immunology , Genes, MHC Class II/immunology , Hepacivirus/immunology , Hepatitis C/therapy , Recombinant Fusion Proteins/immunology , Adenoviridae/genetics , Adenoviridae/immunology , Animals , Antigens, Differentiation, B-Lymphocyte/immunology , Antigens, Differentiation, B-Lymphocyte/therapeutic use , Antigens, Viral/genetics , Antigens, Viral/therapeutic use , CD8-Positive T-Lymphocytes/immunology , Hepatitis C/immunology , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/therapeutic use , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Mice , Pan troglodytes , Recombinant Fusion Proteins/therapeutic use , Vaccines/immunology
7.
Metab Brain Dis ; 27(2): 143-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22392628

ABSTRACT

MHC class II-derived recombinant T cell receptor ligands (RTLs) modulate the behavior of pathogenic T cells and can reverse clinical and histological signs of autoimmune disease in experimental autoimmune encephalomyelitis (EAE), experimental autoimmune uveitis (EAU) and collagen-induced arthritis (CIA), and are currently in clinical trials for treatment of multiple sclerosis (MS). To expand the utility of these rationally-designed biologics and explore their mechanism(s) of activity in vivo, we have engineered RTL constructs bearing cysteine-tethered antigenic peptides and demonstrate that the appropriate cysteine-tethered RTLs effectively treat EAE. The data presented here suggests that the mechanism by which antigen-specific tolerance induction by RTLs bearing cysteine-tethered antigenic peptides in vivo involves delivery of RTL/antigen to endosomal compartments for processing and re-presentation by full-length MHC class II, with RTLs bearing cysteine-tethered antigenic peptides requiring gamma-interferon-inducible lysosomal thiol-reductase (GILT) for therapeutic activity.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Histocompatibility Antigens Class II/therapeutic use , Oxidoreductases/metabolism , Peptide Fragments/therapeutic use , Amino Acid Sequence , Animals , Endosomes/chemistry , Endosomes/metabolism , Genes, MHC Class II/genetics , Ligands , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Mycobacterium tuberculosis/immunology , Oxidoreductases/genetics , Oxidoreductases Acting on Sulfur Group Donors , Protein Engineering , Recombinant Proteins/therapeutic use , T-Lymphocytes/metabolism
8.
J Immunol ; 187(1): 316-24, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21613617

ABSTRACT

CD4 Th cells are critical to the development of coordinated immune responses to infections and tumors. Th cells are activated through interactions of the TCR with MHC class II complexed with peptide. T cell activation is dependent on the density of MHC peptide complexes as well as the duration of interaction of the TCR with APCs. In this study, we sought to determine whether MHC class II peptides could be modified with amino acid sequences that facilitated uptake and presentation with the goal of improving Th cell activation in vitro and in vivo. A model epitope derived from the murine folate receptor α, a self- and tumor Ag, was modified at its carboxyl terminus with the invariant chain-derived Ii-Key peptide and at its N terminus with a peptide that enhances uptake of Ag by APC. Modification of a peptide resulted in enhanced generation of high-avidity murine folate receptor α T cells that persisted in vivo and homed to sites of Ag deposition. The nesting approach was epitope and species independent and specifically excluded expansion of CD4 regulatory T cells. The resulting Th cells were therapeutic, enhanced in vivo helper activity and had an increased ability to resist tolerizing immune microenvironments. In addition to improved immunoadjuvants, this epitope modification strategy may be useful for enhancing ex vivo and in vivo generation of Th cells for preventing and treating diseases.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/immunology , Epitopes, T-Lymphocyte/immunology , Folate Receptor 1/immunology , Histocompatibility Antigens Class II/immunology , T-Lymphocytes, Helper-Inducer/immunology , Amino Acid Sequence , Animals , Cell Adhesion/immunology , Cell Line, Tumor , Cell Movement/immunology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Epitopes, T-Lymphocyte/therapeutic use , Female , Folate Receptor 1/therapeutic use , Histocompatibility Antigens Class II/therapeutic use , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Peptides/immunology , Peptides/therapeutic use , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Helper-Inducer/pathology
9.
Vaccine ; 27(34): 4641-7, 2009 Jul 23.
Article in English | MEDLINE | ID: mdl-19520206

ABSTRACT

Activation of antigen-specific CD4+ T cells is critical for vaccine design. We have advanced a novel technology for enhancing activation of antigen-specific CD4+ T helper cells whereby a fragment of the MHC class II-associated invariant chain (Ii-Key) is linked to an MHC class II epitope. An HLA-DR4-restricted HPV16 E7 epitope, HPV16 E7(8-22), was used to create a homologous series of Ii-Key/HPV16 E7 hybrids testing the influence of spacer length on in vivo enhancement of HPV16 E7(8-22)-specific CD4+ T lymphocyte responses. HLA-DR4-tg mice were immunized with Ii-Key/HPV16 E7(8-22) hybrids or the epitope-only peptide HPV16 E7(8-22). As measured by IFN-gamma ELISPOT assay of splenocytes from immunized mice, one of the Ii-Key/HPV16 E7(8-22) hybrids enhanced epitope-specific CD4+ T cell activation 5-fold compared to the HPV16 E7(8-22) epitope-only peptide. We further demonstrated that enhanced CD4+ T cell activation augments the CTL activity of a H-2D(b)-restricted HPV16 E7(49-57) epitope in HLA-DR4+ mice using an in vivo CTL assay. Binding assays indicated that the Ii-Key/HPV16 hybrid has increased affinity to HLA-DR4+ cells relative to the epitope-only peptide, which may explain its increased potency. In summary, Ii-Key hybrid modification of the HLA-DR4-restricted HPV16 E7(8-22) MHC class II epitope generates a potent immunotherapeutic peptide vaccine that may have potential for treating HPV16+ cancers in HLA-DR4+ patients.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/therapeutic use , Histocompatibility Antigens Class II/therapeutic use , Immunologic Factors/therapeutic use , Immunotherapy/methods , Neoplasms/immunology , Oncogene Proteins, Viral/therapeutic use , Uterine Cervical Neoplasms/immunology , Animals , Antigens, Differentiation, B-Lymphocyte/genetics , CD4-Positive T-Lymphocytes/immunology , Cytotoxicity Tests, Immunologic , Female , Histocompatibility Antigens Class II/genetics , Immunologic Factors/genetics , Immunologic Factors/pharmacology , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , T-Lymphocytes, Cytotoxic/immunology
10.
J Immunol ; 180(4): 2459-65, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18250455

ABSTRACT

Chlamydia infections cause substantial morbidity worldwide and effective prevention will depend on a vaccine. Since Chlamydia immunity is T cell-mediated, a major impediment to developing a molecular vaccine has been the difficulty in identifying relevant T cell Ags. In this study, we used a combination of affinity chromatography and tandem mass spectrometry to identify 13 Chlamydia peptides among 331 self-peptides presented by MHC class II (I-A(b)) molecules from bone marrow-derived murine dendritic cells infected with Chlamydia muridarum. These MHC class II-bound peptides were recognized by Chlamydia-specific CD4 T cells harvested from immune mice and adoptive transfer of dendritic cells pulsed ex vivo with the peptides partially protected mice against intranasal and genital tract Chlamydia infection. The results provide evidence for lead vaccine candidates for a T cell-based subunit molecular vaccine against Chlamydia infection suitable for human study.


Subject(s)
Antigens, Bacterial/isolation & purification , Antigens, Differentiation, T-Lymphocyte/isolation & purification , Chlamydia muridarum/immunology , Intracellular Fluid/immunology , Intracellular Fluid/microbiology , Proteome/immunology , Amino Acid Sequence , Animals , Antigens, Bacterial/metabolism , Antigens, Bacterial/therapeutic use , Antigens, Differentiation, T-Lymphocyte/metabolism , Antigens, Differentiation, T-Lymphocyte/therapeutic use , Cells, Cultured , Chlamydia Infections/immunology , Chlamydia Infections/metabolism , Chlamydia Infections/prevention & control , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Female , H-2 Antigens/isolation & purification , H-2 Antigens/metabolism , HeLa Cells , Histocompatibility Antigens Class II/isolation & purification , Histocompatibility Antigens Class II/metabolism , Histocompatibility Antigens Class II/therapeutic use , Humans , Intracellular Fluid/chemistry , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Peptides/isolation & purification , Peptides/metabolism , Peptides/therapeutic use , Protein Binding/immunology , Proteome/isolation & purification , Proteome/metabolism , Tandem Mass Spectrometry
11.
J Immunol ; 180(2): 1249-57, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-18178865

ABSTRACT

We previously demonstrated the therapeutic effects of MHC class II derived recombinant T cell receptor ligands (RTL), single-chain two domain complexes of the alpha1 and beta1 domains of MHC class II molecules genetically linked with an immunodominant peptide, in experimental autoimmune encephalomyelitis. In the current study, we produced a monomeric murine I-Aq-derived RTL construct covalently linked with bovine collagen type II peptide (bCII257-270) suitable for use in DBA/1LacJ mice that develop collagen-induced arthritis (CIA), an animal model of human rheumatoid arthritis, after immunization with bCII protein in CFA. In this study, we demonstrate that the I-Aq-derived RTLs reduced the incidence of the disease, suppressed the clinical and histological signs of CIA and induced long-term modulation of T cells specific for arthritogenic Ags. Our results showed that the I-Aq/bCII257-270 molecule could systemically reduce proinflammatory IL-17 and IFN-gamma production and significantly increase anti-inflammatory IL-10, IL-13, and FoxP3 gene expression in splenocytes. Moreover, I-Aq/bCII257-270 molecule could also selectively inhibit IL-1beta, IL-6, and IL-23 expression in local joint tissue. This is the first report demonstrating effective prevention of joint inflammation and clinical signs of CIA with an I-Aq-derived RTL, thus supporting the possible clinical use of this approach for treating rheumatoid arthritis in humans.


Subject(s)
Arthritis, Experimental/prevention & control , Receptors, Antigen, T-Cell/agonists , Recombinant Fusion Proteins/therapeutic use , Animals , Antibody Formation , Antigens/immunology , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Cattle , Cytokines/genetics , Cytokines/metabolism , Gene Expression , Hepatocyte Nuclear Factor 3-gamma/genetics , Hepatocyte Nuclear Factor 3-gamma/metabolism , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/therapeutic use , Humans , Joints/pathology , Ligands , Mice , Mice, Inbred DBA , Recombinant Fusion Proteins/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/therapeutic use , Spleen/immunology , Synovial Fluid/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Up-Regulation
12.
J Immunol ; 176(3): 1525-33, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16424181

ABSTRACT

In both collagen-induced arthritis (CIA) and rheumatoid arthritis, T cells recognize a galactosylated peptide from type II collagen (CII). In this study, we demonstrate that the CII259-273 peptide, galactosylated at lysine 264, in complex with Aq molecules prevented development of CIA in mice and ameliorated chronic relapsing disease. In contrast, nonglycosylated CII259-273/Aq complexes had no such effect. CIA dependent on other MHC class II molecules (Ar/Er) was also down-regulated, indicating a bystander vaccination effect. T cells could transfer the amelioration of CIA, showing that the protection is an active process. Thus, a complex between MHC class II molecules and a posttranslationally modified peptide offers a new possibility for treatment of chronically active autoimmune inflammation such as rheumatoid arthritis.


Subject(s)
Arthritis, Experimental/prevention & control , Collagen Type II/therapeutic use , Histocompatibility Antigens Class II/therapeutic use , Immunotherapy, Active , Multiprotein Complexes/therapeutic use , Peptides/therapeutic use , Vaccines/therapeutic use , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/immunology , Bystander Effect/immunology , Cattle , Chronic Disease , Collagen Type II/immunology , Collagen Type II/metabolism , Galactose/metabolism , Glycosylation , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Hybridomas/immunology , Hybridomas/metabolism , Immunodominant Epitopes/immunology , Immunodominant Epitopes/metabolism , Immunodominant Epitopes/therapeutic use , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred BALB C , Multiprotein Complexes/immunology , Multiprotein Complexes/metabolism , Peptides/immunology , Peptides/metabolism , Rats , Solubility , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Vaccines/immunology
13.
J Autoimmun ; 25(1): 21-32, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16005609

ABSTRACT

The self-reactive CD4 T-cells play an essential role in triggering and sustaining organ-specific autoimmune diseases. Silencing or elimination of these cells can prevent and reverse an autoimmune process. We have previously showed that a single dose-administration of a soluble dimeric MHC II-peptide chimera (DEF) in double-transgenic mice delayed the onset autoimmune diabetes, and restored the euglycemia in already diabetic mice for a period of 1 week. DEF dimer protection relied on induction of anergy of diabetogenic CD4 T-cells in spleen, and stimulation of IL-10-secreting T regulatory type 1 cells in pancreas. Herein, we show that an octameric form of DEF has doubled the period of protection and reversal of disease by clonal deletion of diabetogenic CD4 T-cells in both the thymic and peripheral compartments. Deletion occurred by activation-induced cell death subsequent to repartitioning and signaling of FAS-FADD apoptotic module in the plasma membrane lipid rafts. Our previous and present data indicated first, that DEF valence translates into various effects on the antigen-specific CD4 T-cells, i.e., Th2 immune deviation, anergy, and apoptosis. Second, the present findings argue for a better efficacy of clonal deletion than anergy of diabetogenic CD4 T-cells for the protection and reversal of autoimmune diabetes.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Clonal Anergy/immunology , Clonal Deletion/immunology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Animals , CD4-Positive T-Lymphocytes/pathology , Cell Death/genetics , Cell Death/immunology , Cell Membrane/immunology , Cells, Cultured , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/therapeutic use , Membrane Microdomains/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Peptides/genetics , Peptides/therapeutic use , Prediabetic State/genetics , Prediabetic State/immunology , Prediabetic State/pathology , Prediabetic State/prevention & control , Rats , Recombinant Fusion Proteins/therapeutic use , Signal Transduction/immunology , Thymus Gland/cytology , Thymus Gland/immunology
14.
J Immunol ; 172(7): 4556-66, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15034073

ABSTRACT

Our previous studies demonstrated that oligomeric recombinant TCR ligands (RTL) can treat clinical signs of experimental autoimmune encephalomyelitis (EAE) and induce long-term T cell tolerance against encephalitogenic peptides. In the current study, we produced a monomeric I-A(s)/PLP 139-151 peptide construct (RTL401) suitable for use in SJL/J mice that develop relapsing disease after injection of PLP 139-151 peptide in CFA. RTL401 given i.v. or s.c. but not empty RTL400 or free PLP 139-151 peptide prevented relapses and significantly reduced clinical severity of EAE induced by PLP 139-151 peptide in SJL/J or (C57BL/6 x SJL)F(1) mice, but did not inhibit EAE induced by PLP 178-191 or MBP 84-104 peptides in SJL/J mice, or MOG 35-55 peptide in (C57BL/6 x SJL/J)F(1) mice. RTL treatment of EAE caused stable or enhanced T cell proliferation and secretion of IL-10 in the periphery, but reduced secretion of inflammatory cytokines and chemokines. In CNS, there was a modest reduction of inflammatory cells, reduced expression of very late activation Ag-4, lymphocyte function-associated Ag-1, and inflammatory cytokines, chemokines, and chemokine receptors, but enhanced expression of Th2-related factors, IL-10, TGF-beta3, and CCR3. These results suggest that monomeric RTL therapy induces a cytokine switch that curbs the encephalitogenic potential of PLP 139-151-specific T cells without fully preventing their entry into CNS, wherein they reduce the severity of inflammation. This mechanism differs from that observed using oligomeric RTL therapy in other EAE models. These results strongly support the clinical application of this novel class of peptide/MHC class II constructs in patients with multiple sclerosis who have focused T cell responses to known encephalitogenic myelin peptides.


Subject(s)
Cytokines/physiology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Histocompatibility Antigens Class II/metabolism , Histocompatibility Antigens Class II/therapeutic use , Receptors, Antigen, T-Cell/metabolism , Recombinant Proteins/metabolism , Recombinant Proteins/therapeutic use , Amino Acid Sequence , Animals , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Dose-Response Relationship, Immunologic , Encephalomyelitis, Autoimmune, Experimental/pathology , HLA-DR Antigens , Histocompatibility Antigens Class II/chemistry , Injections, Intravenous , Injections, Subcutaneous , Ligands , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Myelin Proteolipid Protein/antagonists & inhibitors , Myelin Proteolipid Protein/toxicity , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/toxicity , Recombinant Proteins/chemistry , Recurrence , Severity of Illness Index , Spinal Cord/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
15.
J Immunol ; 171(10): 5587-95, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-14607967

ABSTRACT

Type 1 diabetes is an autoimmune disorder caused by autoreactive T cells that mediate destruction of insulin-producing beta cells of the pancreas. Studies have shown that T cell tolerance can be restored by inducing a partial or altered signal through the TCR. To investigate the potential of bivalent peptide-MHC class II/Ig fusion proteins as therapeutics to restore Ag-specific tolerance, we have developed soluble peptide I-A(g7) dimers for use in the nonobese diabetic mouse model of diabetes. I-A(g7) dimers with a linked peptide specific for islet-reactive BDC2.5 TCR transgenic CD4(+) T cells were shown to specifically bind BDC2.5 T cells as well as a small population of Ag-specific T cells in nonobese diabetic mice. In vivo treatment with BDC2.5 peptide I-A(g7) dimers protected mice from diabetes mediated by the adoptive transfer of diabetogenic BDC2.5 CD4(+) T cells. The dimer therapy resulted in the activation and increased cell death of transferred BDC2.5 CD4(+) T cells. Surviving cells were hypoproliferative to challenge by Ag and produced increased levels of IL-10 and decreased levels of IFN-gamma compared with cells from control I-A(g7) dimer-treated mice. Anti-IL-10R therapy reversed the tolerogenic effects of the dimer. Thus, peptide I-A(g7) dimers induce tolerance of BDC2.5 TCR T cells through a combination of the induction of clonal anergy and anti-inflammatory cytokines.


Subject(s)
Adjuvants, Immunologic/therapeutic use , CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class II/therapeutic use , Peptide Fragments/immunology , Peptide Fragments/therapeutic use , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/genetics , Adoptive Transfer , Amino Acid Sequence , Animals , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/transplantation , Cell Line , Cells, Cultured , Diabetes Mellitus, Type 1/genetics , Dimerization , Down-Regulation/immunology , Histocompatibility Antigens Class II/administration & dosage , Histocompatibility Antigens Class II/genetics , Immune Tolerance , Interleukin-10/biosynthesis , Lymphocyte Activation/genetics , Mice , Mice, Inbred NOD , Mice, Transgenic , Molecular Mimicry/genetics , Molecular Mimicry/immunology , Molecular Sequence Data , Peptide Fragments/administration & dosage , Peptide Fragments/genetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Transfection
16.
Nucl Med Biol ; 30(7): 715-23, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14499329

ABSTRACT

A (213)Bi-labeled antibody to CD74 was evaluated as a therapeutic agent for B-cell lymphoma. The alpha-particle emission, with a half-life of 46 min, is appropriate for therapy of micrometastases. The labeled Ab retained full immunoreactivity, and was potent at single-cell kill of the Raji B-lymphoma cell line. Approximately 30 decays of cell-bound (213)Bi was required for a cell kill of 99%, and dosimetry calculations suggested that the cGy dose delivered was sufficient to produce the level of toxicity observed. A non-reactive control Ab, labeled similarly, also produced toxicity, due to decays occurring in the medium, but was approximately 3-fold less potent than the reactive Ab. In a SCID mouse xenograft micrometastatic model, Ab injection at day 2 or day 5 after tumor inoculation resulted in strong, specific suppression of tumor growth, with some apparent cures.


Subject(s)
Antibodies/therapeutic use , Bismuth/therapeutic use , Cell Survival/radiation effects , Lymphoma, B-Cell/radiotherapy , Radioimmunotherapy/methods , Radioisotopes/therapeutic use , Animals , Antibodies/immunology , Antigens, Differentiation, B-Lymphocyte/immunology , Antigens, Differentiation, B-Lymphocyte/therapeutic use , Cell Line, Tumor/cytology , Cell Line, Tumor/pathology , Dose-Response Relationship, Radiation , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/therapeutic use , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Mice , Mice, SCID , Radiopharmaceuticals , Radiotherapy Dosage , Survival , Tissue Transplantation , Transplantation, Heterologous , Treatment Outcome
17.
J Immunol ; 164(12): 6610-20, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10843721

ABSTRACT

We explored T cell responses to the self class II MHC (I-Ag7) beta-chain-derived peptides in diabetic and prediabetic nonobese diabetic (NOD) mice. We found that one of these immunodominant epitopes of the beta-chain of I-Ag7 molecule, peptide 54-76, could regulate autoimmunity leading to diabetes in NOD mice. T cells from prediabetic young NOD mice do not respond to the peptide 54-76, but T cells from diabetic NOD mice proliferated in response to this peptide. T cells from older nondiabetic mice or mice protected from diabetes do not respond to this peptide, suggesting a role for peptide 54-76-specific T cells in pathogenesis of diabetes. We show that this peptide is naturally processed and presented by the NOD APCs to self T cells. However, the peptide-specific T cells generated after immunization of young mice regulate autoimmunity in NOD mice by blocking the diabetogenic cells in adoptive transfer experiments. The NOD mice immunized with this peptide are protected from both spontaneous and cyclophosphamide-induced insulin-dependent diabetes mellitus. Immunization of young NOD mice with this peptide elicited T cell proliferation and production of Th2-type cytokines. In addition, immunization with this peptide induced peptide-specific Abs of IgG1 isotype that recognized native I-Ag7 molecule on the cell surface and inhibited the T cell proliferative responses. These results suggest that I-Abetag7(54-76) peptide-reactive T cells are involved in the pathogenesis of diabetes. However, immunization with this peptide at young age induces regulatory cells and the peptide-specific Abs that can modulate autoimmunity in NOD mice and prevent spontaneous and induced diabetes.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/prevention & control , Histocompatibility Antigens Class II/immunology , Peptide Fragments/immunology , Adoptive Transfer , Amino Acid Sequence , Animals , Antigen Presentation/immunology , Cells, Cultured , Cyclophosphamide/antagonists & inhibitors , Cyclophosphamide/pharmacology , Cytokines/biosynthesis , Diabetes Mellitus, Type 1/etiology , Female , Histocompatibility Antigens Class II/administration & dosage , Histocompatibility Antigens Class II/therapeutic use , Immune Sera/biosynthesis , Immune Sera/pharmacology , Immunization , Immunoglobulin G/biosynthesis , Immunosuppressive Agents/pharmacology , Injections, Subcutaneous , Lymphocyte Activation , Male , Mice , Mice, Inbred NOD , Molecular Sequence Data , Peptide Fragments/administration & dosage , Peptide Fragments/metabolism , Peptide Fragments/therapeutic use , Spleen/cytology , Spleen/transplantation , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
18.
J Autoimmun ; 11(3): 241-8, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9693972

ABSTRACT

The expression of a transgenic encoding the I-E alpha-chain, Ea(d), is highly effective in the protection from systemic lupus erythematosus (SLE) in BXSB and (MRL x BXSB)F1 male mice, in which a mutant gene, Yaa (Y-linked autoimmune acceleration), plays a critical role. To gain further insight into the protective role of the Ea(d) transgene, we compared the effect of the transgene in two additional lupus-prone (NZB x BXSB)F1 and (NZW x BXSB)F1 hybrid mice, in which both F1 female mice develop typical SLE in the absence of the Yaa gene and their F1 males bearing the Yaa gene develop a more accelerated form of SLE. Comparative analysis of the clinical development of SLE in these F1 hybrid mice showed that Ea(d) transgene expression was much more effective in the protection from SLE occurring in the F1 females than in their male counterparts. Our results indicate that the Ea(d) transgene is capable of preventing SLE by inhibiting autoimmune responses, independently of the Yaa gene-accelerating effect, and that its protective capacity is strongly influenced by the genetic susceptibility to SLE in individual strains of lupus-prone mice. In addition, this autoimmune inhibitory effect was shown to be selective for IgG, but not IgM, anti-DNA autoantibody production, and is more specific for anti-gp70 autoantibody than for anti-DNA autoantibody. These results favour the hypothesis that the transgene expression may lead to the modulation of self-peptide presentation, thereby preventing excessive T-cell-dependent activation of autoreactive B cells.


Subject(s)
Autoimmunity/genetics , Genetic Therapy/methods , Histocompatibility Antigens Class II/therapeutic use , Lupus Nephritis/prevention & control , Animals , Antigen-Antibody Complex/blood , Autoantibodies/blood , Chimera , DNA/immunology , Female , Genetic Linkage , Glycoproteins/blood , Histocompatibility Antigens Class II/biosynthesis , Histocompatibility Antigens Class II/genetics , Lupus Nephritis/genetics , Male , Mice , Mice, Inbred NZB , Mice, Transgenic , Sex Chromosome Aberrations , Sex Factors , Transgenes , Y Chromosome
19.
J Immunother ; 20(5): 343-53, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9336741

ABSTRACT

We have transfected human melanoma cell line 518A2 with the cDNA encoding interleukin-2 (IL-2) or granulocyte-macrophage colony-stimulating factor (GM-CSF), and compared cytokine-producing clones for their ability to induce melanoma-specific cytotoxic T lymphocytes (CTL) from autologous peripheral blood mononuclear cells (PBMC) in vitro. The parental cell line expressed HLA-A1, HLA-A2, ICAM-1, LFA-3, in addition to the common CTL antigens MAGE-1, MAGE-3, tyrosinase, gp100, and Melan-A/MART-1. Stimulation of autologous PBMC responders with the IL-2-transfected clone 518/IL2.14 specifically induced CTL lines reactive with all cell lines derived from the autologous patient. Strikingly, GM-CSF-transfected 518A2 cells did not induce anti-tumor CTL reactivity. CTL induction against 518/IL2.14 was independent of HLA class II expression or CD4 help. The parental cell line 518A2 gained immunogenic properties when high concentrations of IL-2 were supplied exogenously, indicating that IL-2 produced and present at high levels locally by itself enhanced immunogenicity. From the autologous CTL line reactive with 518/IL2.14, clones were generated against an as yet unknown antigen, which was present in all autologous melanoma cell lines as well as in 7 of 15 HLA-A2+ melanoma cell lines tested, but not in melanocytes. These results will be discussed with respect to the possibility of using IL-2-transfected melanoma cells as a vaccine for treatment of patients with melanoma.


Subject(s)
Cancer Vaccines/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Head and Neck Neoplasms/therapy , Interleukin-2/immunology , Melanoma, Experimental/therapy , T-Lymphocytes, Cytotoxic/immunology , Transfection , Cancer Vaccines/therapeutic use , Genetic Therapy , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/immunology , Histocompatibility Antigens Class II/therapeutic use , Humans , Immunotherapy/methods , Interleukin-2/genetics , Interleukin-2/therapeutic use , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Tumor Cells, Cultured
20.
Eur J Immunol ; 26(12): 3234-7, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8977328

ABSTRACT

To investigate the role of interleukin (IL)-4 during the triggering of collagen-induced arthritis, we examined the effects of the I-A(b) and I-E protective/suppressive genes and passively administered anti-IL-4 monoclonal antibody. In contrast to the action of I-E expression on its own, which has mainly a suppressive effect post-triggering, the combination of I-A(b) and I-E had a marked protective effect. Assuming, on the basis of previous experience with the I-A(b) allele, that it might act through suppressing early IL-4 production, we treated mice with the 11B11 IL-4-neutralizing antibody around the time of initial immunization with collagen. Treatment over a period extending to 6 days post-immunization exacerbated the arthritis, but when curtailed to 2 days post-immunization (and tested in pristane-primed animals), the disease was reduced. We conclude that IL-4 plays an essential role in triggering the disease.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Experimental/prevention & control , Collagen , Histocompatibility Antigens Class II/therapeutic use , Interleukin-4/physiology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Arthritis, Experimental/etiology , Histocompatibility Antigens Class II/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA
SELECTION OF CITATIONS
SEARCH DETAIL