Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Immunity ; 42(2): 367-378, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25680276

ABSTRACT

T cell dysfunction is well documented during chronic viral infections but little is known about functional abnormalities in humoral immunity. Here we report that mice persistently infected with lymphocytic choriomeningitis virus (LCMV) exhibit a severe defect in Fcγ-receptor (FcγR)-mediated antibody effector functions. Using transgenic mice expressing human CD20, we found that chronic LCMV infection impaired the depletion of B cells with rituximab, an anti-CD20 antibody widely used for the treatment of B cell lymphomas. In addition, FcγR-dependent activation of dendritic cells by agonistic anti-CD40 antibody was compromised in chronically infected mice. These defects were due to viral antigen-antibody complexes and not the chronic infection per se, because FcγR-mediated effector functions were normal in persistently infected mice that lacked LCMV-specific antibodies. Our findings have implications for the therapeutic use of antibodies and suggest that high levels of pre-existing immune complexes could limit the effectiveness of antibody therapy in humans.


Subject(s)
Antibodies, Viral/immunology , Antigen-Antibody Complex/immunology , Lymphocyte Depletion , Lymphocytic Choriomeningitis/immunology , Receptors, IgG/immunology , Animals , Antibodies, Monoclonal, Murine-Derived/pharmacology , Antigens, CD20/biosynthesis , Antigens, CD20/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/immunology , Dendritic Cells/immunology , Hypergammaglobulinemia/immunology , Immunologic Factors/pharmacology , Lymphocyte Activation/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Rituximab
2.
Invest New Drugs ; 37(5): 837-848, 2019 10.
Article in English | MEDLINE | ID: mdl-30465316

ABSTRACT

Caffeic acid phenethyl ester (CAPE) is a phenolic compound initially identified in bee glue. CAPE is reported to exhibit antitumor activity in many cancer models. However, the effect of CAPE on multiple myeloma (MM) is not well studied. We investigated the anti-myeloma effect of CAPE, and the data showed that CAPE inhibited the growth of human MM cells in a dose (1 ~ 30 µM) and time (24 ~72 h) dependent manner without altering the viability of normal human peripheral blood B cells. Stress and toxicity pathway analysis demonstrated that CAPE, in a dose- and time-related fashion, induced the expression of apoptotic and oxidative stress-response genes including growth arrest and DNA-damage inducible, alpha and gamma (GADD45A and GADD45G) and heme oxygenase-1. Apoptosis of MM cells by CAPE was further confirmed through flow cytometric analysis with up to 50% apoptotic cells induced by 50 µM CAPE within 24 h. Western blot analysis revealed the CAPE-induced activation of apoptosis executioner enzyme caspase-3, and corresponding cleavage of its downstream target poly(ADP-ribose)polymerase (PARP). The oxidative stress caused by CAPE cytotoxicity in MM cells was evaluated through measurement of reactive oxygen species (ROS) level, antioxidant intervention and glutathione depletion. The intracellular ROS level was not elevated by CAPE, but the pretreatment of antioxidant (N-acetyl cysteine) and glutathione synthesis inhibitor (buthionine sulfoximine) suggested that CAPE may cause oxidative stress by decrease of intracellular antioxidant level rather than over production of ROS. These data suggest that CAPE promotes apoptosis through oxidative stress in human multiple myeloma cells.


Subject(s)
Apoptosis/drug effects , Biomarkers, Tumor/genetics , Caffeic Acids/pharmacology , Multiple Myeloma/pathology , Oxidative Stress/drug effects , Phenylethyl Alcohol/analogs & derivatives , Antioxidants/metabolism , Cell Movement , Cell Proliferation/drug effects , Gene Expression Profiling , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Phenylethyl Alcohol/pharmacology , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured
3.
Inflamm Res ; 67(7): 589-596, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29713730

ABSTRACT

BACKGROUND: The chronic inflammation associated with rheumatoid arthritis (RA) leads to focal and systemic bone erosion of the joints resulting in a crippling disability. Recent reports indicate an increase in the incidence of RA in the coming years, placing a significant burden on healthcare resources. The incidence of RA is observed to be increasing with age and a significant proportion of those new cases will be aggressively erosive. FINDINGS: The altered physiology, due to immune disturbances, contributes towards RA pathogenesis. The imbalance of inflammatory cytokines and non-cytokine immune modulators such as prostaglandin E2 (PGE2) and IL-23-induced pathogenic IL-17, plays a crucial role in persistent inflammation and bone degradation during RA. However, the molecular mechanism of IL-23, a key cytokine, and PGE2 in the development and perpetuation of IL-17 producing effector Th17 cells is poorly understood. CONCLUSION: This review focuses on research findings that provide insight into the contribution of PGE2 and IL-23 during the development of pathogenic Th17 cells. We also highlight the key transcriptional factors required for Th17 development and therapeutic strategies to disrupt the interaction between IL-23 and IL-17 to prevent the end-organ damage in RA.


Subject(s)
Arthritis, Rheumatoid/metabolism , Cytokines/metabolism , Dinoprostone/metabolism , Th17 Cells/metabolism , Animals , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Humans , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction
4.
Arthritis Rheum ; 65(4): 1064-73, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23280250

ABSTRACT

OBJECTIVE: Lipocalin 2 (LCN-2) is an innate immune protein that is expressed by a variety of cells and is highly up-regulated during several pathologic conditions, including immune complex (IC)-mediated inflammatory/autoimmune disorders. However, the function of LCN-2 during IC-mediated inflammation is largely unknown. Therefore, this study was undertaken to investigate the role of LCN-2 in IC-mediated diseases. METHODS: The up-regulation of LCN-2 was determined by enzyme-linked immunosorbent assay in 3 different mouse models of IC-mediated autoimmune disease: systemic lupus erythematosus, collagen-induced arthritis, and serum-transfer arthritis. The in vivo role of LCN-2 during IC-mediated inflammation was investigated using LCN-2-knockout mice and their wild-type littermates. RESULTS: LCN-2 levels were significantly elevated in all 3 of the autoimmune disease models. Further, in an acute skin inflammation model, LCN-2-knockout mice exhibited a 50% reduction in inflammation, with histopathologic analysis revealing notably reduced immune cell infiltration as compared to wild-type mice. Administration of recombinant LCN-2 to LCN-2-knockout mice restored inflammation to levels observed in wild-type mice. Neutralization of LCN-2 using a monoclonal antibody significantly reduced inflammation in wild-type mice. In contrast, LCN-2-knockout mice developed more severe serum-induced arthritis compared to wild-type mice. Histologic analysis revealed extensive tissue and bone destruction, with significantly reduced neutrophil infiltration but considerably more macrophage migration, in LCN-2-knockout mice compared to wild-type mice. CONCLUSION: These results demonstrate that LCN-2 may regulate immune cell recruitment to the site of inflammation, a process essential for the controlled initiation, perpetuation, and resolution of inflammatory processes. Thus, LCN-2 may present a promising target in the treatment of IC-mediated inflammatory/autoimmune diseases.


Subject(s)
Acute-Phase Proteins/immunology , Antigen-Antibody Complex/immunology , Arthritis, Experimental/immunology , Dermatitis/immunology , Inflammation/immunology , Lipocalins/immunology , Lupus Erythematosus, Systemic/immunology , Oncogene Proteins/immunology , Acute Disease , Acute-Phase Proteins/genetics , Acute-Phase Proteins/metabolism , Animals , Antigen-Antibody Complex/metabolism , Arthritis, Experimental/metabolism , Chronic Disease , Dermatitis/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Inflammation/metabolism , Lipocalin-2 , Lipocalins/genetics , Lipocalins/metabolism , Lupus Erythematosus, Systemic/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Up-Regulation
5.
J Immunol ; 189(4): 1911-9, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22786765

ABSTRACT

Various states of inflammation, including sepsis, are associated with hypoferremia, which limits iron availability to pathogens and reduces iron-mediated oxidative stress. Lipocalin 2 (Lcn2; siderocalin, 24p3) plays a central role in iron transport. Accordingly, Lcn2-deficient (Lcn2KO) mice exhibit elevated intracellular labile iron. In this study, we report that LPS induced systemic Lcn2 by 150-fold in wild-type mice at 24 h. Relative to wild-type littermates, Lcn2KO mice were markedly more sensitive to endotoxemia, exhibiting elevated indices of organ damage (transaminasemia, lactate dehydrogenase) and increased mortality. Such exacerbated endotoxemia was associated with substantially increased caspase-3 cleavage and concomitantly elevated immune cell apoptosis. Furthermore, cells from Lcn2KO mice were hyperresponsive to LPS ex vivo, exhibiting elevated cytokine secretion. Additionally, Lcn2KO mice exhibited delayed LPS-induced hypoferremia despite normal hepatic hepcidin expression and displayed decreased levels of the tissue redox state indicators cysteine and glutathione in liver and plasma. Desferroxamine, an iron chelator, significantly protects Lcn2KO mice from LPS-induced toxicity, including mortality, suggesting that Lcn2 may act as an antioxidant in vivo by regulating iron homeostasis. Thus, Lcn2-mediated regulation of labile iron protects the host against sepsis. Its small size and simple structure may make Lcn2 a deployable treatment for sepsis.


Subject(s)
Acute-Phase Proteins/metabolism , Homeostasis/physiology , Iron/metabolism , Lipocalins/metabolism , Oncogene Proteins/metabolism , Sepsis/metabolism , Acute-Phase Proteins/deficiency , Acute-Phase Proteins/immunology , Animals , Apoptosis/physiology , Endotoxins/toxicity , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunoblotting , Lipocalin-2 , Lipocalins/immunology , Male , Mice , Mice, Knockout , Oncogene Proteins/deficiency , Oncogene Proteins/immunology , Reverse Transcriptase Polymerase Chain Reaction , Sepsis/chemically induced , Sepsis/immunology
6.
Med Res Rev ; 32(6): 1197-219, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23059764

ABSTRACT

Cancer cells have developed numerous ways to escape immune surveillance and gain unlimited proliferative capacity. Currently, several chemotherapeutic agents and radiotherapy, either alone or in combination, are being used to treat malignancies. However, both of these therapies are associated with several limitations and detrimental side effects. Therefore, recent scientific investigations suggest that immunotherapy is among the most promising new approaches in modern cancer therapy. The focus of cancer immunotherapy is to boost both acquired and innate immunity against malignancies by specifically targeting tumor cells, and leaving healthy cells and tissues unharmed. Cellular, cytokine, gene, and monoclonal antibody therapies have progressively become promising immunotherapeutic approaches that are being tested for several cancers in preclinical models as well as in the clinic. In this review, we discuss recent advances in these immunotherapeutic approaches, focusing on new strategies that allow the expression of specific immunostimulatory molecules on the surface of tumor cells to induce robust antitumor immunity.


Subject(s)
Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Immunotherapy/methods , Neoplasm Proteins/immunology , Neoplasms/immunology , Neoplasms/therapy , Animals , Humans
7.
J Immunol ; 184(11): 6293-8, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20439912

ABSTRACT

Immunologic memory is a hallmark of the vertebrate immune system. The first antigenic exposure leads to a slow and modest immune response, whereas repeated exposure, even many years later, leads to a rapid and exaggerated response that is two to three orders of magnitude greater than the primary. In the case of humoral immunity, the increased efficacy of recall responses is due to the production of amplified levels of Ag-specific Ab, as well as the accelerated kinetics of their production. Current thinking suggests that this is due to selective activation of long-lived, Ag-specific memory B cells. A downside of restricting secondary responses solely to memory cells is that the repertoire of the memory B cell pool remains static while pathogens continue to evolve. In this study, we propose that during secondary responses, naive Ag-specific B cells participate alongside memory cells. We show that immune complexes formed in vivo between the Ag and pre-existing Abs from the primary response activate these naive B cells, inducing them to respond with accelerated kinetics and increased magnitude. Thus, the continued recruitment of new B cell clones after each antigenic exposure enables the immune system to stay abreast of rapidly changing pathogens.


Subject(s)
Antibody Formation/immunology , Antigen-Antibody Complex/immunology , B-Lymphocyte Subsets/immunology , B-Lymphocytes/immunology , Immunologic Memory/immunology , Lymphocyte Activation/immunology , Adoptive Transfer , Animals , Antibodies/immunology , CD4-Positive T-Lymphocytes/immunology , Complement Activation/immunology , Enzyme-Linked Immunosorbent Assay , Immunity, Humoral , Mice , Mice, Inbred C57BL , Receptors, IgG/immunology
8.
J Immunol ; 183(12): 8216-24, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-20007585

ABSTRACT

CD32A, the major phagocytic FcgammaR in humans, exhibits a polymorphism in the ligand binding domain. Individuals homozygous for the R allelic form of CD32A (CD32A(R) allele) are more susceptible to bacterial infections and autoimmune diseases as compared with H allelic CD32A (CD32A(H)) homozygous and CD32A(R/H) heterozygous individuals. To understand the mechanisms behind this differential susceptibility, we have investigated the dynamics of the interaction of these allelic forms of CD32A when they are simultaneously exposed to immune complexes (IC). Binding studies using Ig fusion proteins of CD32A alleles showed that the R allele has significantly lower binding not only to human IgG2, but also to IgG1 and IgG3 subtypes. Competition assays using purified molecules demonstrated that CD32A(H)-Ig outcompetes CD32A(R)-Ig for IC binding when both alleles simultaneously compete for the same ligand. CD32A(H)-Ig blocked the IC binding mediated by both the allelic forms of cell surface CD32A, whereas CD32A(R)-Ig blocked only CD32A(R) and was unable to cross-block IC binding mediated by CD32A(H). Two-dimensional affinity measurements also demonstrated that CD32A(R) has significantly lower affinity toward all three subtypes as compared with CD32A(H). Our data suggest that the lower binding of CD32A(R) not only to IgG2 but also to IgG1 and IgG3 might be responsible for the lack of clearance of IC leading to increased susceptibility to bacterial infections and autoimmune diseases. Our data further suggests that in humans, inflammatory cells from CD32A(R/H) heterozygous individuals may predominantly use the H allele to mediate Ab-coated target cell binding during phagocytosis and Ab-dependent cellular cytotoxicity, resulting in a phenotype similar to CD32A(H) homozygous individuals.


Subject(s)
Alleles , Antigen-Antibody Complex/metabolism , Arginine/genetics , Histidine/genetics , Immunoglobulin G/classification , Immunoglobulin G/metabolism , Receptors, IgG/biosynthesis , Receptors, IgG/genetics , Animals , Antigen-Antibody Complex/genetics , Arginine/biosynthesis , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Bacterial Infections/genetics , Bacterial Infections/immunology , Bacterial Infections/metabolism , Binding, Competitive/genetics , Binding, Competitive/immunology , CHO Cells , Cell Line , Cell Membrane/genetics , Cell Membrane/immunology , Cell Membrane/metabolism , Cricetinae , Cricetulus , Dimerization , Genetic Predisposition to Disease , Histidine/biosynthesis , Humans , Immunoglobulin G/genetics , Ligands , Polymorphism, Genetic/immunology , Protein Binding/genetics , Protein Binding/immunology , Receptors, IgG/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
9.
J Microencapsul ; 28(4): 294-300, 2011.
Article in English | MEDLINE | ID: mdl-21545320

ABSTRACT

In this study, we formulated a microparticulate melanoma cancer vaccine via the transdermal route. The vaccine was delivered using microneedle-based Dermaroller® which is available for cosmetic purposes. Unlike subcutaneous injections, administration using microneedles is painless and in general can increase the permeability of many compounds ranging in size from small molecules to proteins and microparticles that do not normally penetrate the skin. The vaccine microparticles were taken up by the antigen presenting cells which demonstrated a strong IgG titre level of 930 ug/mL in serum samples. The formulation increased the immunogenicity of the vaccine by incorporating the antigen into an albumin matrix having a size range of around 0.63-1.4 µm which acted as a synthetic adjuvant. The animals were vaccinated with 1 prime and 4 booster doses administered every 14 days over 8 weeks duration, followed by challenge with live tumour cells which showed protection after transdermal vaccination.


Subject(s)
Antibodies, Neoplasm , Antigens, Neoplasm , Cancer Vaccines , Drug Carriers/pharmacology , Immunoglobulin G , Melanoma , Animals , Antibodies, Neoplasm/blood , Antigens, Neoplasm/immunology , Antigens, Neoplasm/pharmacology , Cancer Vaccines/immunology , Cancer Vaccines/pharmacology , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Injections, Subcutaneous , Melanoma/blood , Melanoma/immunology , Melanoma/therapy , Mice , Vaccination/methods
10.
Medicines (Basel) ; 6(1)2019 Jan 31.
Article in English | MEDLINE | ID: mdl-30709026

ABSTRACT

Background: Plant-derived phytochemicals have been of emerging interest as anti-obesity compounds due to their apparent effects on promoting reduced lipid accumulation in adipocytes. Despite such promising evidence, little is known about the potential mechanisms behind their anti-obesity effects. The aim of this study is to establish potential anti-obesity effects of the phytochemical guggulsterone (GS). Methods: Mature 3T3-L1 adipocytes were treated with GS, derived from the guggul plant native in northern India, to investigate its effects on mitochondrial biogenesis and adipocyte "beiging." Further, to explore the relationship between macrophages and adipocytes, 3T3-L1s were treated with conditioned media from GS-treated RAW264.7 macrophages. Markers of mitochondrial biogenesis and beiging were measured by western blot. Results: GS treatment in adipocytes resulted in increased mitochondrial density, biogenesis (PGC1α and PPARγ), and increased markers of a beige adipocyte phenotype (UCP1, TBX1, and ß-3AR). This upregulation in mitochondrial expression was accompanied by increases oxygen consumption. In GS-treated macrophages, markers of M2 polarization were elevated (e.g., arginase and IL-10), along with increased catecholamine release into the media. Lastly, 3T3-L1 adipocytes treated with conditioned media from macrophages induced a 167.8% increase in UCP1 expression, suggestive of a role of macrophages in eliciting an anti-adipogenic response to GS. Conclusions: Results from this study provide the first mechanistic understanding of the anti-obesity effects of GS and suggests a role for both direct GS-signaling and indirect stimulation of M2 macrophage polarization in this model.

11.
Hum Vaccin ; 4(5): 384-8, 2008.
Article in English | MEDLINE | ID: mdl-18382145

ABSTRACT

Attempts to create vaccines for humans against invading pathogens such as viruses and bacteria have met with tremendous success. The process of developing vaccines against these pathogens is greatly aided by the fact that they contain antigens that are entirely foreign to humans. Although the knowledge and strategies developed for designing vaccines against these microbes may be of use in developing cancer vaccines, the poor antigenicity and immunosuppressive ability of cancers pose major hurdles to vaccine development. Established tumors have not only withstood immune screening and selection pressure, making them poor stimulators of an immune response, but have also adapted mechanisms to continue evading immune surveillance by creating an immunosuppressive environment. Also, genetic differences in immune responses to an antigen among individuals result in an antigenic profile that varies from patient to patient. Cancers bear such great similarities to normal cells in the body that, on a molecular level, the differences between cancerous and non-cancerous cells are minor. Therefore, developing vaccines which use the host's own tumor tissues carries the risk of breaking tolerance to self-antigens that are present in the tumor tissue. Vaccination strategies that will optimally stimulate the immune system against tumor specific antigens under immunosuppressive conditions need to be developed. In practical terms, this calls for a method by which therapeutic vaccines may be custom-designed to treat cancers case by case. Ex vivo manipulation of dendritic cells and gene transfer of immunostimulatory molecules in ex vivo expanded tumors are being tested in both experimental models and also in human clinical trials. Some of them have met with limited success. Emerging technologies such as protein transfer, which make it possible to express immunostimulatory molecules on tumor cell membranes, offer the means to develop efficient tumor vaccines that are simple and fast, while being easy to store and administer in human patients. Progress in these techniques will move the cancer vaccine field a step closer towards realizing custom designed cancer vaccines in human clinical settings.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cancer Vaccines/immunology , Neoplasms/therapy , Vaccination/methods , Animals , Humans
12.
Nutr Metab (Lond) ; 15: 42, 2018.
Article in English | MEDLINE | ID: mdl-29946343

ABSTRACT

BACKGROUND: Obesity alters adipose tissue metabolic and endocrine functioning, leading to an increased adiposity and release of pro-inflammatory cytokines. Various phytochemicals have been reported to contribute to the beiging of white adipose tissue in order to ameliorate obesity by increasing thermogenesis. Here, we show that the prenylated chalcone, xanthohumol (XN), induces beiging of white adipocytes, stimulates lipolysis, and inhibits adipogenesis of murine 3T3-L1 adipocytes and primary human subcutaneous preadipocytes and these effects are partly mediated by the activation of the AMP-activated protein kinase (AMPK) signaling pathway. METHODS: 3T3-L1 adipocytes and primary human subcutaneous preadipocytes were differentiated using a standard protocol and were treated with various concentrations of XN, dorsomorphin, an AMPK inhibitor, or AICAR, an AMPK activator, to investigate the effects on adipogenesis, beiging and lipolysis. RESULTS: XN induced beiging of white adipocytes as witnessed by the increased expression of beige markers CIDE-A and TBX-1. XN increased mitochondrial biogenesis, as evidenced by increased mitochondrial content, enhanced expression of PGC-1α, and the thermogenic protein UCP1. Following 24 h of treatment, XN also increased oxygen consumption rate. XN stimulated lipolysis of mature 3T3-L1 and primary human subcutaneous adipocytes and inhibited adipogenesis of maturing adipocytes. XN activated AMPK and in turn, XN-induced upregulation of UCP1, p-ACC, HSL, and ATGL was downregulated in the presence of dorsomorphin. Likewise, an XN-induced decrease in adipogenesis was reversed in the presence of dorsomorphin. CONCLUSIONS: Taken together, XN demonstrates anti-obesity effects by not only inducing beiging but also decreasing adipogenesis and inducing lipolysis. The anti-obesity effects of XN are partly mediated by AMPK signaling pathway suggesting that XN may have potential therapeutic implications for obesity.

13.
Mol Biol Cell ; 27(22): 3449-3458, 2016 11 07.
Article in English | MEDLINE | ID: mdl-27582391

ABSTRACT

Isoforms of the Fcγ receptor III (FcγRIII or CD16) are cell surface receptors for the Fc portion of IgG and important regulators of humoral immune responses. Different ligand binding kinetics of FcγRIII isoforms are obtained in three dimensions by surface plasmon resonance and in two dimensions by a micropipette adhesion frequency assay. We show that the anchor structure of CD16 isoforms isolated from the cell membrane affects their binding affinities in a ligand-specific manner. Changing the receptor anchor structure from full to partial to none decreases the ligand binding affinity for human IgG1 (hIgG1) but increases it for murine IgG2a (mIgG2a). Removing N-glycosylation from the CD16 protein core by tunicamycin also increases the ligand binding affinity. Molecular dynamics simulations indicate that deglycosylation at Asn-163 of CD16 removes the steric hindrance for the CD16-hIgG1 Fc binding and thus increases the binding affinity. These results highlight an unexpected sensitivity of ligand binding to the receptor anchor structure and glycosylation and suggest their respective roles in controlling allosterically the conformation of the ligand binding pocket of CD16.


Subject(s)
Receptors, IgG/metabolism , Animals , Cell Membrane/metabolism , GPI-Linked Proteins/metabolism , Glycosylation , Humans , Immunoglobulin G/metabolism , Kinetics , Ligands , Models, Molecular , Protein Binding , Protein Isoforms , Receptors, IgG/physiology , Structure-Activity Relationship , Surface Plasmon Resonance
14.
Cell Mol Gastroenterol Hepatol ; 2(4): 482-498.e6, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27458605

ABSTRACT

BACKGROUND & AIMS: Lipocalin 2 (Lcn2) is a multifunctional innate immune protein whose expression closely correlates with extent of intestinal inflammation. However, whether Lcn2 plays a role in the pathogenesis of gut inflammation is unknown. Herein, we investigated the extent to which Lcn2 regulates inflammation and gut bacterial dysbiosis in mouse models of IBD. METHODS: Lcn2 expression was monitored in murine colitis models and upon microbiota ablation/restoration. WT and Lcn2 knockout (Lcn2KO) mice were analyzed for gut bacterial load, composition by 16S rRNA gene pyrosequencing and, their colitogenic potential by co-housing with Il-10KO mice. Acute (dextran sodium sulfate) and chronic (IL-10R neutralization and T-cell adoptive transfer) colitis was induced in WT and Lcn2KO mice with or without antibiotics. RESULTS: Lcn2 expression was dramatically induced upon inflammation and was dependent upon presence of a gut microbiota and MyD88 signaling. Use of bone-marrow chimeric mice revealed non-immune cells are the major contributors of circulating Lcn2. Lcn2KO mice exhibited elevated levels of entA-expressing gut bacteria burden and, moreover, a broadly distinct bacterial community relative to WT littermates. Lcn2KO mice developed highly colitogenic T-cells and exhibited exacerbated colitis upon exposure to DSS or neutralization of IL-10. Such exacerbated colitis could be prevented by antibiotic treatment. Moreover, exposure to the microbiota of Lcn2KO mice, via cohousing, resulted in severe colitis in Il-10KO mice. CONCLUSION: Lcn2 is a bacterially-induced, MyD88-dependent, protein that play an important role in gut homeostasis and a pivotal role upon challenge. Hence, therapeutic manipulation of Lcn2 levels may provide a strategy to help manage diseases driven by alteration of the gut microbiota.

15.
PLoS One ; 11(4): e0153620, 2016.
Article in English | MEDLINE | ID: mdl-27101012

ABSTRACT

Autoimmune vasculitis is an endothelial inflammatory disease that results from the deposition of immune-complexes (ICs) in blood vessels. The interaction between Fcgamma receptors (FcγRs) expressed on inflammatory cells with ICs is known to cause blood vessel damage. Hence, blocking the interaction of ICs and inflammatory cells is essential to prevent the IC-mediated blood vessel damage. Thus we tested if uncoupling the interaction of FcγRs and ICs prevents endothelium damage. Herein, we demonstrate that dimeric FcγR-Igs prevented nitric oxide (NO) mediated apoptosis of human umbilical vein endothelial cells (HUVECs) in an in vitro vasculitis model. Dimeric FcγR-Igs significantly inhibited the IC-induced upregulation of inducible nitric oxide synthase (iNOS) and nitric oxide (NO) release by murine monocytic cell line. However, FcγR-Igs did not affect the exogenously added NO-induced upregulation of pro-apoptotic genes such as Bax (15 fold), Bak (35 fold), cytochrome-C (11 fold) and caspase-3 (30 fold) in HUVECs. In conclusion, these data suggest that IC-induced NO could be one of the major inflammatory mediator promoting blood vessel inflammation and endothelial cell death during IC-mediated vasculitis which can be effectively blocked by dimeric decoy FcγRs.


Subject(s)
Antigen-Antibody Complex/immunology , Apoptosis/drug effects , Endothelium, Vascular/cytology , Nitric Oxide/pharmacology , Phagocytes/immunology , Receptors, IgG/immunology , Human Umbilical Vein Endothelial Cells , Humans , Inflammation Mediators/metabolism , Nitric Oxide Synthase Type II/metabolism
16.
ACS Med Chem Lett ; 4(7): 616-621, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23914286

ABSTRACT

Prostanoid receptor EP2 is emerging as a novel target for development of anti-inflammatory drugs for the treatment of chronic neurodegenerative and peripheral diseases; however, the availability of EP2 antagonist probes for exploration of peripheral disease models is very limited. We now report identification and characterization of a novel chemical class of compounds that show nanomolar potency and competitive antagonism of the EP2 receptor. A compound in this class, TG6-129, showed prolonged plasma half-life and did not cross the blood brain barrier. This compound also suppressed the induction of inflammatory mRNA markers in a macrophage cell line upon activation of EP2. Thus, this compound could be useful as a probe for a variety of peripheral chronic inflammatory diseases such as rheumatoid arthritis and chronic obstructive pulmonary disease, in which EP2 appears to play a pathogenic role.

17.
Vaccine ; 31(20): 2449-56, 2013 May 07.
Article in English | MEDLINE | ID: mdl-23541884

ABSTRACT

Many studies have shown that the systemic administration of cytokines or vaccination with cytokine-secreting tumors augments an antitumor immune response that can result in eradication of tumors. However, these approaches are hampered by the risk of systemic toxicity induced by soluble cytokines. In this study, we have evaluated the efficacy of 4TO7, a highly tumorigenic murine mammary tumor cell line, expressing glycosyl phosphatidylinositol (GPI)-anchored form of cytokine molecules alone or in combination with the costimulatory molecule B7-1 as a model for potential cell or membrane-based breast cancer vaccines. We observed that the GPI-anchored cytokines expressed on the surface of tumor cells greatly reduced the overall tumorigenicity of the 4TO7 tumor cells following direct live cell challenge as evidenced by transient tumor growth and complete regression within 30 days post challenge. Tumors co-expressing B7-1 and GPI-IL-12 grew the least and for the shortest duration, suggesting that this combination of immunostimulatory molecules is most potent. Protective immune responses were also observed following secondary tumor challenge. Further, the 4TO7-B7-1/GPI-IL-2 and 4TO7-B7-1/GPI-IL-12 transfectants were capable of inducing regression of a wild-type tumor growing at a distant site in a concomitant tumor challenge model, suggesting the tumor immunity elicited by the transfectants can act systemically and inhibit the tumor growth at a distant site. Additionally, when used as irradiated whole cell vaccines, 4TO7-B7-1/GPI-IL-12 led to a significant inhibition in tumor growth of day 7 established tumors. Lastly, we observed a significant decrease in the prevalence of myeloid-derived suppressor cells and regulatory T-cells in the tumor microenvironment on day 7 post challenge with 4TO7-B7-1/GPI-IL-12 cells, which provides mechanistic insight into antitumor efficacy of the tumor-cell membrane expressed IL-12. These studies have implications in designing membrane-based therapeutic vaccines with GPI-anchored cytokines for breast cancer.


Subject(s)
B7-1 Antigen/immunology , Breast Neoplasms/immunology , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Membrane/metabolism , Cytokines/immunology , Tumor Microenvironment/immunology , Animals , B7-1 Antigen/genetics , B7-1 Antigen/metabolism , Breast Neoplasms/complications , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cancer Vaccines/metabolism , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Female , Glycosylphosphatidylinositols/metabolism , Interleukin-12/genetics , Interleukin-12/immunology , Interleukin-12/metabolism , Interleukin-2/genetics , Interleukin-2/immunology , Interleukin-2/metabolism , Mice , Mice, Inbred BALB C , Myeloid Cells/cytology , Myeloid Cells/immunology , Solubility , Splenomegaly/complications , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transfection , Tumor Burden
18.
J Drug Target ; 20(2): 166-73, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21981679

ABSTRACT

Cancer vaccines are limited in their use, because of their inability to mount a robust anti-tumor immune response. Thus, targeting M-cells in the small intestine, which are responsible for entry of many pathogens, will be an attractive way to elicit a strong immune response toward particulate antigens. Therefore, in the present investigation, we demonstrated that efficient oral vaccination against melanoma antigens could be accomplished by incorporating the antigens in an albumin-based microparticle with a ligand AAL (Aleuria aurantia lectin) targeted specifically to M-cells. The oral microparticulate vaccine effectively protected the mice from subcutaneous challenge with tumor cells in prophylactic settings. The animals were vaccinated with antigen microparticles having a size range of around 1-1.25 µm where one prime and four booster doses were administered every 14 days over 10 weeks of duration, followed by challenge with live tumor cells, which showed complete tumor protection after oral vaccination. With the inclusion of ligand in the microparticles, we observed significantly higher IgG titers (1565 µg/mL) as compared to the microparticle formulations without AAL (872 µg/mL). This data suggests that ligand loaded microparticles may have the potential to target antigens to M-cells for an efficient oral vaccination.


Subject(s)
Cancer Vaccines/administration & dosage , Cancer Vaccines/therapeutic use , Drug Delivery Systems/methods , Melanoma/therapy , Microspheres , Peyer's Patches/drug effects , Administration, Oral , Albumins/administration & dosage , Albumins/chemistry , Animals , Cancer Vaccines/chemistry , Cancer Vaccines/immunology , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Drug Screening Assays, Antitumor/methods , Drug Screening Assays, Antitumor/statistics & numerical data , Female , Immunoglobulin G/immunology , Lectins/administration & dosage , Lectins/chemistry , Melanoma/immunology , Mice , Mice, Inbred DBA , Particle Size , Peyer's Patches/immunology , Surface Properties
19.
Front Biosci (Landmark Ed) ; 15(1): 309-20, 2010 01 01.
Article in English | MEDLINE | ID: mdl-20036822

ABSTRACT

Cancer vaccine development is one of the most hopeful and exhilarating areas in cancer research. For this reason, there has been a growing interest in the development and application of novel immunotherapies for the treatment of cancer with the focus being on stimulating the immune system to target tumor cells specifically while leaving normal cells unharmed. From such research has emerged a host of promising immunotherapies such as dendritic cell-based vaccines, cytokine therapies and gene transfer technology. These therapies seek to counteract the poor immunogenicity of tumors by augmenting the host's immune system with a variety of immunostimulatory proteins such as cytokines and costimulatory molecules. While such therapies have proven effective in the induction of anti-tumor immunity in animal models, they are less than optimal and pose a high risk of clinical infeasibility. Herein, we further discuss these immunotherapies as well as a feasible and efficient alternative that, in pre-clinical animal models, allows for the expression of specific immunostimulatory molecules on the surface of tumor cells by a novel protein transfer technology.


Subject(s)
Cancer Vaccines/immunology , Immunity/immunology , Neoplasms/immunology , Animals , Cancer Vaccines/genetics , Cancer Vaccines/therapeutic use , Dendritic Cells/cytology , Dendritic Cells/immunology , Gene Transfer Techniques , Humans , Immunity/drug effects , Immunotherapy/methods , Models, Biological , Neoplasms/pathology , Neoplasms/therapy , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology
20.
J Control Release ; 137(2): 90-7, 2009 Jul 20.
Article in English | MEDLINE | ID: mdl-19328830

ABSTRACT

Biodegradable microparticles can function as an adjuvant by targeting antigens to professional antigen presenting cells such as dendritic cells and macrophages. To enhance targeting of microparticles, we have developed a novel method of attaching immunostimulatory molecules such as B7-1 to the surface of albumin microparticles utilizing the glycosylphosphatidyl inositol (GPI) anchor. GPI-B7-1 attaches to the surface of albumin microparticles in a protein transfer mediated process and is functionally active. This protein transfer was dependent on the concentration of the GPI-anchored protein, and independent of temperature and incubation time. Results show that the binding of the GPI-anchored protein is specifically occurring through an interaction between the GPI-anchor and the albumin microparticle surface. Stability studies indicate that the GPI-anchored protein can remain attached to the surface of the microparticle up to 7 days, with storage at 4 degrees C providing the optimal stability. Finally, we were able to simultaneously attach two different GPI-anchored proteins, GPI-B7-1 and GPI-ICAM-1, to the microparticles, demonstrating the capability of attaching more than one GPI-anchored protein to the microparticle surface. This novel method of attaching proteins to the surface of microparticles has potential implications in using microparticles as an antigen delivery device in vaccines as well as in targeted drug delivery.


Subject(s)
B7-1 Antigen/chemistry , Drug Carriers/chemistry , Glycosylphosphatidylinositols/chemistry , Intercellular Adhesion Molecule-1/chemistry , Serum Albumin/chemistry , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/genetics , Adjuvants, Immunologic/isolation & purification , Animals , B7-1 Antigen/genetics , B7-1 Antigen/immunology , B7-1 Antigen/isolation & purification , Cattle , Cell Line, Tumor , Gene Expression , Glycosylphosphatidylinositols/genetics , Glycosylphosphatidylinositols/isolation & purification , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Intercellular Adhesion Molecule-1/isolation & purification , Lipids/chemistry , Mice , Protein Engineering , Protein Stability , Rats , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL