Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 702
Filter
1.
Front Immunol ; 12: 694152, 2021.
Article in English | MEDLINE | ID: mdl-34858388

ABSTRACT

(1→3)-ß-D-Glucan (BDG) represents a potent pathogen-associated molecular pattern (PAMP) in triggering the host response to fungal and some bacterial infections. Monocytes play a key role in recognizing BDG and governing the acute host response to infections. However, the mechanisms regulating monocyte's acute response to BDG are poorly understood. We sought to investigate the response of monocytes to BDG at the epigenetic, transcriptomic, and molecular levels. Response of human monocytes to 1, 4, and 24 hours of BDG exposure was investigated using RNA-seq, ATAC-seq, H3K27ac and H3K4me1 ChIP-seq. We show that pathways including glutathione metabolism, pentose phosphate pathway, and citric acid cycle were upregulated at the epigenetic and transcriptomic levels in response to BDG exposure. Strikingly, unlike bacterial lipopolysaccharides, BDG induced intracellular glutathione synthesis. BDG exposure also induced NADP synthesis, increased NADPH/NADP ratio, and increased expression of genes involved in the pentose phosphate pathway in a GSH-dependent manner. By inhibiting GSH synthesis with L-buthionine sulfoximine (BSO) before BDG exposure we show that the GSH pathway promotes cell survival and regulates monocyte's effector functions including NO production, phagocytosis, and cytokine production. In summary, our work demonstrates that BDG induces glutathione synthesis and metabolism in monocytes, which is a major promoter of the acute functional response of monocytes to infections.


Subject(s)
Glutathione/metabolism , Monocytes/immunology , Pathogen-Associated Molecular Pattern Molecules/immunology , Proteoglycans/immunology , Buthionine Sulfoximine/pharmacology , Cell Survival , Cells, Cultured , Citric Acid/metabolism , Epigenesis, Genetic , Gene Expression Profiling , Humans , Immunity, Innate , Nitric Oxide/metabolism , Pentose Phosphate Pathway , Phagocytosis , Sequence Analysis, RNA
2.
Cell Immunol ; 366: 104383, 2021 08.
Article in English | MEDLINE | ID: mdl-34111646

ABSTRACT

For over 70 years experimental autoimmune encephalomyelitis (EAE) has been induced with myelin autoantigens emulsified in complete Freund's adjuvant (CFA) which has significant side effects such as pain, inflammation, and tissue necrosis at the injection site. ß-1,3-d-glucan particles (GPs) are hollow microcapsules prepared from Saccharomyces cerevisiae cell walls that induce potent Th17 cell responses without causing strong injection site tissue reactions. We evaluated the potential of GPs complexed with neuroantigens to induce EAE while avoiding undesirable side effects. GPs loaded with myelin oligodendrocyte glycoprotein 35-55 (MOG35-55) or proteolipid protein 139-151 (PLP139-151) peptides effectively induced EAE in C57BL/6 mice and SJL mice. Disease severity, CNS pathology and immune responses were comparable between GP- and CFA-immunized mice. Importantly, injection with GPs resulted in significantly decreased inflammation compared with CFA. We posit that use of GPs provides an alternative means for inducing EAE that results in comparable disease, but less discomfort to animals.


Subject(s)
Adjuvants, Immunologic/metabolism , Capsules/metabolism , Cell Wall/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Proteoglycans/metabolism , Saccharomyces cerevisiae/metabolism , Animals , Disease Models, Animal , Female , Freund's Adjuvant , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Myelin Proteolipid Protein/immunology , Myelin-Oligodendrocyte Glycoprotein/immunology , Peptide Fragments/immunology , Proteoglycans/immunology , Th17 Cells/immunology
3.
J Immunol Methods ; 493: 113015, 2021 06.
Article in English | MEDLINE | ID: mdl-33689807

ABSTRACT

BACKGROUND: During eosinophil differentiation, the granule eosinophil major basic protein 1 (eMBP1) is synthesized as a 32-kDa precursor form, referred to as proMBP1, which is processed into the 14-kDa mature form of eMBP1. The prevalence of these two forms of MBP1 in most pathological conditions remains unknown. OBJECTIVE: To develop the immunoassays that differentiate mature eMBP1 and proMBP1 and apply them to analyze their levels in biological fluids from patients with eosinophilia and hematologic disorders. METHODS: We produced a series of monoclonal antibodies and selected pairs capable of discriminating between the two molecular forms of eMBP1. Radioimmunoassay (RIA) was performed to simultaneously quantitate the levels of mature eMBP1 and proMBP1 in secretions from patients with chronic rhinosinusitis (CRS) and sera from patients with hypereosinophilic syndrome (HES) and other myeloproliferative disorders. RESULTS: The novel immunoassays possessed less than 1% crossreactivity between mature eMBP1 and proMBP1. Mature eMBP1, but not proMBP1, was found in nasal secretions of CRS patients. In contrast, elevated serum levels of mature eMBP1 and proMBP1 were observed in approximately 60% and 90% of HES patients, respectively, with proMBP1 present in greater quantities than mature eMBP1. Patients with several myeloproliferative disorders also showed high serum levels of proMBP1 while mature eMBP1 remained at basal levels. CONCLUSION: The novel immunoassays successfully differentiated mature eMBP1 and proMBP1 in human biological fluids. Further studies addressing the clinical correlates of these assays will help to develop biomarkers to diagnose and monitor patients with eosinophilia and myeloproliferative disorders.


Subject(s)
Eosinophil Major Basic Protein/blood , Eosinophilia/diagnosis , Immunoassay/methods , Myeloproliferative Disorders/diagnosis , Proteoglycans/blood , Antibodies, Monoclonal/immunology , Eosinophil Major Basic Protein/immunology , Eosinophilia/immunology , Humans , Myeloproliferative Disorders/immunology , Proteoglycans/immunology
4.
J Allergy Clin Immunol ; 147(4): 1442-1452, 2021 04.
Article in English | MEDLINE | ID: mdl-32791164

ABSTRACT

BACKGROUND: The immunoinhibitory receptor Siglec-8 on the surface of human eosinophils and mast cells binds to sialic acid-containing ligands in the local milieu, resulting in eosinophil apoptosis, inhibition of mast cell degranulation, and suppression of inflammation. Siglec-8 ligands were found on postmortem human trachea and bronchi and on upper airways in 2 compartments, cartilage and submucosal glands, but they were surprisingly absent from the epithelium. We hypothesized that Siglec-8 ligands in submucosal glands and ducts are normally transported to the airway mucus layer, which is lost during tissue preparation. OBJECTIVE: Our aim was to identify the major Siglec-8 sialoglycan ligand on the mucus layer of human airways. METHODS: Human upper airway mucus layer proteins were recovered during presurgical nasal lavage of patients at a sinus clinic. Proteins were resolved by gel electrophoresis and blotted, and Siglec-8 ligands detected. Ligands were purified by size exclusion and affinity chromatography, identified by proteomic mass spectrometry, and validated by electrophoretic and histochemical colocalization. The affinity of Siglec-8 binding to purified human airway ligand was determined by inhibition of glycan binding. RESULTS: A Siglec-8-ligand with a molecular weight of approximately 1000 kDa was found in all patient nasal lavage samples. Purification and identification revealed deleted in malignant brain tumors 1 (DMBT1) (also known by the aliases GP340 and SALSA), a large glycoprotein with multiple O-glycosylation repeats. Immunoblotting, immunohistochemistry, and enzyme treatments confirmed that Siglec-8 ligand on the human airway mucus layer is an isoform of DMBT1 carrying O-linked sialylated keratan sulfate chains (DMBT1S8). Quantitative inhibition revealed that DMBT1S8 has picomolar affinity for Siglec-8. CONCLUSION: A distinct DMBT1 isoform, DMBT1S8, is the major high-avidity ligand for Siglec-8 on human airways.


Subject(s)
Antigens, CD/immunology , Antigens, Differentiation, B-Lymphocyte/immunology , Calcium-Binding Proteins/immunology , DNA-Binding Proteins/immunology , Lectins/immunology , Tumor Suppressor Proteins/immunology , Bronchi/immunology , Calcium-Binding Proteins/chemistry , DNA-Binding Proteins/chemistry , Eosinophils/immunology , Humans , Ligands , Mast Cells/immunology , Nasal Lavage Fluid/immunology , Proteoglycans/immunology , Trachea/immunology , Tumor Suppressor Proteins/chemistry
5.
Front Immunol ; 11: 604413, 2020.
Article in English | MEDLINE | ID: mdl-33363545

ABSTRACT

Legionella pneumophila, an intracellular bacterium, may cause life-threatening pneumonia in immunocompromised individuals. Mononuclear cells and antibodies have been reported to be associated with the host defense response against L. pneumophila. This study is to determine whether Legionella peptidoglycan-associated lipoprotein (PAL)-specific CD8+ T cells are directly associated with protection against L. pneumophila, with a focus on potential epitopes. Synthetic peptides derived from PAL of L. pneumophila were obtained and tested through in vitro and in vivo cytotoxic T lymphocyte (CTL) assays for immunogenicity. PAL DNA vaccines or a peptide epitope with or without CpG-oligodeoxynucleotides (ODN) was evaluated for protection against L. pneumophila infection in animal models. When mice were immunized with DNA vaccines expressing the PAL of L. pneumophila, they were significantly protected against a lethal challenge with L. pneumophila through induction of antigen-specific CD8+ CTLs. Of the 13 PAL peptides tested, PAL92-100 (EYLKTHPGA) was the most immunogenic and induced the strongest CTL responses. When mice were immunized with the PAL92-100 peptide plus CpG-ODN, they were protected against the lethal challenge, while control mice died within 3-6 days after the challenge. Consistent with lung tissue histological data, bacterial counts in the lungs of immunized mice were significantly lower than those in control mice. Also, the amino acid sequence of PAL92-100 peptides is conserved among various Legionella species. To our knowledge, this study is the first to demonstrate that PAL92-100-specific CD8+ T cells play a central role in the host defense response against L. pneumophila.


Subject(s)
Bacterial Outer Membrane Proteins/administration & dosage , Bacterial Vaccines/administration & dosage , Epitopes , Legionella pneumophila/immunology , Legionnaires' Disease/prevention & control , Lung/immunology , Peptide Fragments/administration & dosage , Proteoglycans/administration & dosage , T-Lymphocytes, Cytotoxic/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Bacterial Load , Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/immunology , Cells, Cultured , Cytokines/metabolism , Female , Host-Pathogen Interactions , Immunization , Legionnaires' Disease/immunology , Legionnaires' Disease/metabolism , Legionnaires' Disease/microbiology , Lung/metabolism , Lung/microbiology , Lung/pathology , Lymphocyte Activation , Mice, Inbred BALB C , Oligodeoxyribonucleotides/administration & dosage , Peptide Fragments/immunology , Proteoglycans/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/microbiology
7.
Front Immunol ; 11: 769, 2020.
Article in English | MEDLINE | ID: mdl-32508807

ABSTRACT

Proteoglycans are a specific subset of glycoproteins found at the cell surface and in the extracellular matrix, where they interact with a plethora of proteins involved in metabolic homeostasis and meta-inflammation. Over the last decade, new insights have emerged on the mechanism and biological significance of these interactions in the context of diet-induced disorders such as obesity and type-2 diabetes. Complications of energy metabolism drive most diet-induced metabolic disorders, which results in low-grade chronic inflammation, thereby affecting proper function of many vital organs involved in energy homeostasis, such as the brain, liver, kidney, heart and adipose tissue. Here, we discuss how heparan, chondroitin and keratan sulfate proteoglycans modulate obesity-induced metabolic dysfunction and low-grade inflammation that impact the initiation and progression of obesity-associated morbidities.


Subject(s)
Obesity/immunology , Obesity/metabolism , Proteoglycans/immunology , Proteoglycans/metabolism , Adipocytes , Adipose Tissue , Animals , Diet , Extracellular Matrix/metabolism , Heparan Sulfate Proteoglycans , Humans , Inflammation/metabolism , Mice
8.
Neoplasia ; 22(2): 86-97, 2020 02.
Article in English | MEDLINE | ID: mdl-31896526

ABSTRACT

While recent research points to the importance of glycans in cancer immunity, knowledge on functional mechanisms is lacking. In lung carcinoma among other tumors, anti-tumor immunity is suppressed; and while some recent therapies boost T-cell mediated immunity by targeting immune-checkpoint pathways, robust responses are uncommon. Augmenting tumor antigen-specific immune responses by endogenous dendritic cells (DCs) is appealing from a specificity standpoint, but challenging. Here, we show that restricting a heparan sulfate (HS) loss-of-function mutation in the HS sulfating enzyme Ndst1 to predominantly conventional DCs (Ndst1f/f CD11cCre+ mutation) results in marked inhibition of Lewis lung carcinoma growth along with increased tumor-associated CD8+ T cells. In mice deficient in a major DC HS proteoglycan (syndecan-4), splenic CD8+ T cells showed increased anti-tumor cytotoxic responses relative to controls. Studies examining Ndst1f/f CD11cCre + mutants revealed that mutation was associated with an increase in anti-tumor cytolysis using either splenic CD8+ T cells or tumor-infiltrating (TIL) CD8+ T cells purified ex-vivo, and tested in pooled effector-to-target cytolytic assays against tumor cells from respective animals. On glycan compositional analysis, HS purified from Ndst1f/f CD11cCre + mutant DCs had reduced overall sulfation, including reduced sulfation of a tri-sulfated disaccharide species that was intriguingly abundant on wildtype DC HS. Interestingly, antigen presentation in the context of major histocompatibility complex class-I (MHC-I) was enhanced in mutant DCs, with more striking effects in the setting of HS under-sulfation, pointing to a likely regulatory role by sulfated glycans at the antigen/MHC-I - T-cell interface; and possibly future opportunities to improve antigen-specific T cell responses by immunologic targeting of HS proteoglycans in cancer.


Subject(s)
Carcinoma, Lewis Lung/drug therapy , Major Histocompatibility Complex/genetics , Polysaccharides/genetics , Proteoglycans/genetics , Sulfotransferases/genetics , Animals , CD11c Antigen/genetics , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/pathology , Dendritic Cells/immunology , Dendritic Cells/pathology , Heparitin Sulfate/pharmacology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Immunity, Cellular/genetics , Immunity, Cellular/immunology , Loss of Function Mutation/genetics , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Major Histocompatibility Complex/immunology , Mice , Polysaccharides/antagonists & inhibitors , Proteoglycans/antagonists & inhibitors , Proteoglycans/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
9.
Semin Cancer Biol ; 62: 48-67, 2020 05.
Article in English | MEDLINE | ID: mdl-31336150

ABSTRACT

Over the past few decades, understanding how tumor cells evade the immune system and their communication with their tumor microenvironment, has been the subject of intense investigation, with the aim of developing new cancer immunotherapies. The current therapies against cancer such as monoclonal antibodies against checkpoint inhibitors, adoptive T-cell transfer, cytokines, vaccines, and oncolytic viruses have managed to improve the clinical outcome of the patients. However, in some tumor entities, the response is limited and could benefit from the identification of novel therapeutic targets. It is known that tumor-extracellular matrix interplay and matrix remodeling are necessary for anti-tumor and pro-tumoral immune responses. Proteoglycans are dominant components of the extracellular matrix and are a highly heterogeneous group of proteins characterized by the covalent attachment of a specific linear carbohydrate chain of the glycosaminoglycan type. At cell surfaces, these molecules modulate the expression and activity of cytokines, chemokines, growth factors, adhesion molecules, and function as signaling co-receptors. By these mechanisms, proteoglycans influence the behavior of cancer cells and their microenvironment during the progression of solid tumors and hematopoietic malignancies. In this review, we discuss why cell surface proteoglycans are attractive pharmacological targets in cancer, and we present current and recent developments in cancer immunology and immunotherapy utilizing proteoglycan-targeted strategies.


Subject(s)
Cell Membrane/metabolism , Neoplasms/metabolism , Proteoglycans/metabolism , Animals , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor , Carbohydrate Metabolism , Cell Membrane/immunology , Clinical Trials as Topic , Drug Evaluation, Preclinical , Extracellular Matrix , Glycosaminoglycans , Humans , Immunomodulation/drug effects , Immunotherapy , Molecular Targeted Therapy , Neoplasms/etiology , Neoplasms/immunology , Neoplasms/therapy , Proteoglycans/antagonists & inhibitors , Proteoglycans/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Microenvironment/immunology
10.
Med Mol Morphol ; 53(2): 94-103, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31595373

ABSTRACT

Atopic cataracts develop under the ages of 40 years, after which visual acuity rapidly declines. However, the mechanism underlying the development of atopic cataracts is not yet clear. We focused on the eosinophil granule major basic protein (MBP), which was detected in the aqueous humor of atopic cataracts previously, and which was cytotoxic. Specifically, we investigated its origin in this fluid and its effects on lens epithelial cells (LECs). MBP immunostaining was positive in atopic cataract-derived LECs, but negative in age-related cataract-derived LECs. MBP mRNA was not detected in either type of cataract, but protein was detected in the aqueous humor. Furthermore, the flare values associated with atopic cataracts were higher than those with age-related cataracts. When MBP was purified from eosinophils or recombinant MBP was added to LEC culture medium, cell viability decreased in a concentration-dependent manner, but an MBP antibody neutralized the cytotoxic effect of this protein towards these cells. These results were consistent with the flow of MBP into the aqueous humor from the blood due to a compromised blood-aqueous barrier. Thus, MBP could further penetrate the lens capsule and adhere to LECs, resulting in decreased cell viability and the development of atopic cataracts.


Subject(s)
Cataract/immunology , Eosinophil Major Basic Protein/metabolism , Eosinophils/metabolism , Proteoglycans/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Aqueous Humor/immunology , Aqueous Humor/metabolism , Case-Control Studies , Cataract/blood , Cataract/pathology , Cataract Extraction , Cell Survival/immunology , Cells, Cultured , Eosinophil Major Basic Protein/analysis , Eosinophil Major Basic Protein/immunology , Eosinophil Major Basic Protein/isolation & purification , Eosinophils/immunology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Female , Humans , Lens, Crystalline/cytology , Lens, Crystalline/immunology , Lens, Crystalline/pathology , Lens, Crystalline/surgery , Male , Primary Cell Culture , Proteoglycans/analysis , Proteoglycans/immunology , Proteoglycans/isolation & purification , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Young Adult
11.
Sci Rep ; 9(1): 11689, 2019 08 12.
Article in English | MEDLINE | ID: mdl-31406213

ABSTRACT

Canine distemper virus (CDV) causes a fatal demyelinating leukoencephalitis in young dogs resembling human multiple sclerosis. Astrocytes are the main cellular target of CDV and undergo reactive changes already in pre-demyelinating brain lesions. Based on their broad range of beneficial and detrimental effects in the injured brain reactive astrogliosis is in need of intensive investigation. The aim of the study was to characterize astrocyte plasticity during the course of CDV-induced demyelinating leukoencephalitis by the aid of immunohistochemistry, immunofluorescence and gene expression analysis. Immunohistochemistry revealed the presence of reactive glial fibrillary acidic protein (GFAP)+ astrocytes with increased survivin and reduced aquaporin 4, and glutamine synthetase protein levels, indicating disturbed blood brain barrier function, glutamate homeostasis and astrocyte maladaptation, respectively. Gene expression analysis revealed 81 differentially expressed astrocyte-related genes with a dominance of genes associated with neurotoxic A1-polarized astrocytes. Accordingly, acyl-coA synthetase long-chain family member 5+/GFAP+, and serglycin+/GFAP+ cells, characteristic of A1-astrocytes, were found in demyelinating lesions by immunofluorescence. In addition, gene expression revealed a dysregulation of astrocytic function including disturbed glutamate homeostasis and altered immune function. Observed findings indicate an astrocyte polarization towards a neurotoxic phenotype likely contributing to lesion initiation and progression in canine distemper leukoencephalitis.


Subject(s)
Astrocytes/virology , Demyelinating Diseases/veterinary , Distemper Virus, Canine/pathogenicity , Distemper/virology , Encephalomyelitis, Acute Disseminated/veterinary , Glial Fibrillary Acidic Protein/genetics , Animals , Aquaporin 4/genetics , Aquaporin 4/immunology , Astrocytes/immunology , Astrocytes/pathology , Blood-Brain Barrier/immunology , Blood-Brain Barrier/pathology , Blood-Brain Barrier/virology , Coenzyme A Ligases/genetics , Coenzyme A Ligases/immunology , Demyelinating Diseases/genetics , Demyelinating Diseases/pathology , Demyelinating Diseases/virology , Disease Progression , Distemper/genetics , Distemper/immunology , Distemper/pathology , Distemper Virus, Canine/immunology , Dogs , Encephalomyelitis, Acute Disseminated/genetics , Encephalomyelitis, Acute Disseminated/pathology , Encephalomyelitis, Acute Disseminated/virology , Gene Expression Regulation , Glial Fibrillary Acidic Protein/immunology , Glutamate-Ammonia Ligase/genetics , Glutamate-Ammonia Ligase/immunology , Glutamic Acid/immunology , Glutamic Acid/metabolism , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Proteoglycans/genetics , Proteoglycans/immunology , Signal Transduction , Survivin/genetics , Survivin/immunology , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/immunology
12.
J Invest Dermatol ; 139(11): 2343-2351.e3, 2019 11.
Article in English | MEDLINE | ID: mdl-31128201

ABSTRACT

Reactivation of functionally-impaired anticancer T cells by programmed cell death protein 1 (PD-1) and programmed cell death receptor ligand-1 (PD-L1)-blocking antibodies shows prominent therapeutic benefit in advanced melanoma and patients with non-small cell lung cancer. However, current PD-L1-blocking antibodies lack intrinsic tumor selectivity. Therefore, efficacy may be reduced resulting from on-target and off-tumor binding to PD-L1-expressing normal cells. This may lead to indiscriminate activation of antigen-experienced T cells, including those implicated in autoimmune-related adverse events. To direct PD-L1 blockade to chondroitin sulfate proteoglycan 4 (CSPG4)-expressing cancers and to reactivate anticancer T cells more selectively, we constructed bispecific antibody PD-L1xCSPG4. CSPG4 is an established target antigen that is selectively overexpressed on malignant melanoma and various other difficult-to-treat cancers. PD-L1xCSPG4 showed enhanced capacity for CSPG4-directed blockade of PD-L1 on cancer cells. Importantly, treatment of mixed cultures containing primary patient-derived CSPG4-expressing melanoma cells and autologous tumor-infiltrating lymphocytes with PD-L1xCSPG4 significantly enhanced activation status, IFN-γ production, and cytolytic activity of anticancer T cells. In conclusion, tumor-directed blockade of PD-L1 by PD-L1xCSPG4 may improve efficacy and safety of PD-1/PD-L1 checkpoint blockade for treatment of melanoma and other CSPG4-overexpressing malignancies.


Subject(s)
Antibodies, Bispecific/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Immunotherapy/methods , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/therapy , T-Lymphocytes/immunology , Antibodies, Bispecific/genetics , Antigens/immunology , Antigens, Neoplasm/immunology , B7-H1 Antigen/immunology , Cell Line, Tumor , Cell Proliferation , Cytotoxicity, Immunologic , Epitopes , Humans , Interferon-gamma/metabolism , Lymphocyte Activation , Lymphocyte Culture Test, Mixed , Melanoma/immunology , Protein Engineering , Proteoglycans/immunology
13.
Bioessays ; 41(1): e1800166, 2019 01.
Article in English | MEDLINE | ID: mdl-30485469

ABSTRACT

Proteoglycan 4 (PRG4), first identified in synovial fluid, is an extracellular matrix structural protein in the joint implicated in reducing shear at the cartilage surface as well as controlling adhesion-dependent synovial growth and regulating bulk protein deposition onto the cartilage. However, recent evidence suggests that it can bind to and effect downstream signaling of a number of cell surface receptors implicated in regulating the inflammatory response. Therefore, we pose the hypothesis: Does PRG4 regulate the inflammatory response and maintain tissue homeostasis? Based on these novel findings implicating PRG4 as an inflammatory signaling molecule, we will present and discuss several hypotheses regarding potential mechanisms by which PRG4 may be able to regulate inflammation. If future studies can demonstrate that PRG4 is a potent inflammatory mediator, this will change current paradigms in the musculoskeletal and ophthalmological fields regarding the how the inflammatory microenvironment is regulated in these tissues and potentially others.


Subject(s)
Homeostasis , Inflammation/metabolism , Proteoglycans/physiology , Animals , Humans , Proteoglycans/immunology , Proteoglycans/metabolism , Signal Transduction
14.
J Innate Immun ; 11(2): 136-149, 2019.
Article in English | MEDLINE | ID: mdl-30205385

ABSTRACT

It has emerged that neutrophils can play important roles in the host response following infection with helminth parasites. Mice infected with the tapeworm, Hymenolepis diminuta, are protected from some inflammatory conditions, accompanied by reduced neutrophil tissue infiltration. Thus, the ability of a phosphate-buffered saline-soluble extract of the worm (H. diminuta extract [HdE]) was tested for (1) its ability to activate murine neutrophils (Ca2+ mobilization, reactive oxygen species (ROS) and cytokine production); and (2) affect neutrophil chemotaxis in vitro to the penta-peptide, WKYMVm, the chemokine, KC, and leukotriene B4. HdE was not cytotoxic to neutrophils, elicited a Ca2+ response and ROS, but not, cytokine (KC, interleukin-10, tumour necrosis factor-α) generation. HdE is not a chemotactic stimulus for murine neutrophils. However, a heat- and trypsin-sensitive, acid-insensitive proteoglycan (sensitive to sodium metaperiodate) in the HdE significantly reduced neutrophil chemotaxis towards WKYMVm or KC, but not LTB4. The latter suggested that the HdE interfered with p38 mitogen-activated protein kinase signalling, which is important in WKYMVm chemotaxis. Corroborating this, immunoblotting revealed reduced phosphorylated p38, and the downstream signal heat-shock protein-27, in protein extracts from HdE + WkYMVm treated cells compared to those exposed to the penta-peptide only. We speculate that HdE can be used to modify the outcome of neutrophilic disease and that purification of the bioactive proteoglycan(s) from the extract could be used as a template to design immunomodulatory drugs targeting neutrophils.


Subject(s)
Antigens, Helminth/metabolism , Hymenolepiasis/immunology , Hymenolepis diminuta/physiology , Neutrophils/immunology , Proteoglycans/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Antigens, Helminth/immunology , Calcium Signaling , Cell Extracts/pharmacology , Cells, Cultured , Chemotaxis , Cytokines/metabolism , Down-Regulation , Enzyme Activation , Host-Parasite Interactions , Male , Mice , Mice, Inbred BALB C , Neutrophil Activation , Proteoglycans/immunology , Trypsin/metabolism
15.
Int J Chron Obstruct Pulmon Dis ; 13: 3367-3372, 2018.
Article in English | MEDLINE | ID: mdl-30410323

ABSTRACT

BACKGROUND: Endothelial cell specific molecule-1, also called as endocan, is a dermatan sulfate proteoglycan, which is expressed by endothelial cells in alveolar walls of the lung and kidney. High endocan levels are found associated with endothelial dysfunction and inflammation. We hypothesize that endocan level is also high in COPD due to systemic inflammation and endothelial dysfunction. We aimed to investigate the expression of endocan in patients with stable COPD. MATERIAL AND METHODS: The study included patients with COPD and control subjects. COPD patients were classified according to the Global Initiative for Chronic Obstructive Lung Disease (GOLD) 2017 criteria. Demographics, body mass index, smoking history, and comorbidities were recorded. Endocan levels of COPD patients and controls were compared. RESULTS: Totally, 88 subjects (47 stable COPD patients, 41 controls) were evaluated. Endocan levels were significantly higher in COPD patients than control group (860.1±259.8 vs 647.3±316.9 pg/mL, P=0.001). There was no relationship between GOLD COPD categories and endocan levels. Also endocan levels were similar between COPD patients with or without hypoxemia. CONCLUSION: Serum endocan level was significantly higher in patients with stable COPD. Further studies should be performed to better understand the relationship between endocan and COPD.


Subject(s)
Alveolar Epithelial Cells/immunology , Neoplasm Proteins , Proteoglycans , Pulmonary Disease, Chronic Obstructive , Adult , Aged , Correlation of Data , Female , Humans , Inflammation/immunology , Male , Middle Aged , Neoplasm Proteins/analysis , Neoplasm Proteins/immunology , Patient Acuity , Proteoglycans/analysis , Proteoglycans/immunology , Pulmonary Disease, Chronic Obstructive/diagnosis , Pulmonary Disease, Chronic Obstructive/immunology
16.
J Immunol ; 201(9): 2721-2730, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30266767

ABSTRACT

The respiratory pathogen Moraxella catarrhalis is a human-specific commensal that frequently causes acute otitis media in children and stimulates acute exacerbations in chronic obstructive pulmonary disease patients. The exact molecular mechanisms defining host-pathogen interactions promoting pathogenesis are not clearly understood. Limited knowledge hampers vaccine and immunotherapeutic development required to treat this emerging pathogen. In this study, we reveal in detail a novel antibacterial role displayed by short leucine-rich proteoglycans (SLRPs) in concert with complement. We show that fibromodulin (FMOD), osteoadherin (OSAD), and biglycan (BGN) but not decorin (DCN) enhance serum killing of M. catarrhalis. Our results suggest that M. catarrhalis binding to SLRPs is a conserved feature, as the overwhelming majority of clinical and laboratory strains bound all four SLRPs. Furthermore, we resolve the binding mechanism responsible for this interaction and highlight the role of the ubiquitous surface protein (Usp) A2/A2H in mediating binding to host SLRPs. A conserved immune evasive strategy used by M. catarrhalis and other pathogens is the surface acquisition of host complement inhibitors such as C4b-binding protein (C4BP). We observed that FMOD, OSAD, and BGN competitively inhibit binding of C4BP to the surface of M. catarrhalis, resulting in increased C3b/iC3b deposition, membrane attack complex (MAC) formation, and subsequently decreased bacterial survival. Furthermore, both OSAD and BGN promote enhanced neutrophil killing in vitro, both in a complement-dependent and independent fashion. In summary, our results illustrate that SLRPs, FMOD, OSAD, and BGN portray complement-modulating activity enhancing M. catarrhalis killing, defining a new antibacterial role supplied by SLRPs.


Subject(s)
Complement Activation/immunology , Host-Pathogen Interactions/immunology , Moraxella catarrhalis/immunology , Moraxellaceae Infections/immunology , Proteoglycans/immunology , Humans , Leucine
17.
Int J Mol Sci ; 19(4)2018 Mar 21.
Article in English | MEDLINE | ID: mdl-29561763

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer with a poor prognosis. There is a clinical need for effective, targeted therapy strategies that destroy both differentiated TNBC cells and TNBC cancer initiating cells (CICs), as the latter are implicated in the metastasis and recurrence of TNBC. Chondroitin sulfate proteoglycan 4 (CSPG4) is overexpressed on differentiated tumor cells and CICs obtained from TNBC patient specimens, suggesting that CSPG4 may be a clinically relevant target for the imaging and therapy of TNBC. The purpose of this study was to determine whether α-particle radioimmunotherapy (RIT) targeting TNBC cells using the CSPG4-specific monoclonal antibody (mAb) 225.28 as a carrier was effective at eliminating TNBC tumors in preclinical models. To this end, mAb 225.28 labeled with 212Pb (212Pb-225.28) as a source of α-particles for RIT was used for in vitro Scatchard assays and clonogenic survival assays with human TNBC cells (SUM159 and 2LMP) grown as adherent cells or non-adherent CIC-enriched mammospheres. Immune-deficient mice bearing orthotopic SUM159 or 2LMP xenografts were injected i.v. with the targeted (225.28) or irrelevant isotype-matched control (F3-C25) mAbs, labeled with 99mTc, 125I, or 212Pb for in vivo imaging, biodistribution, or tumor growth inhibition studies. 212Pb-225.28 bound to adherent SUM159 and 2LMP cells and to CICs from SUM159 and 2LMP mammospheres with a mean affinity of 0.5 nM. Nearly ten times more binding sites per cell were present on SUM159 cells and CICs compared with 2LMP cells. 212Pb-225.28 was six to seven times more effective than 212Pb-F3-C25 at inhibiting SUM159 cell and CIC clonogenic survival (p < 0.05). Radiolabeled mAb 225.28 showed significantly higher uptake than radiolabeled mAb F3-C25 in SUM159 and 2LMP xenografts (p < 0.05), and the uptake of 212Pb-225.28 in TNBC xenografts was correlated with target epitope expression. 212Pb-225.28 caused dose-dependent growth inhibition of SUM159 xenografts; 0.30 MBq 212Pb-225.28 was significantly more effective than 0.33 MBq 212Pb-F3-C25 at inhibiting tumor growth (p < 0.01). These results suggest that CSPG4-specific 212Pb-225.28 is a useful reagent for RIT of CSPG4-expressing tumors, including metastatic TNBC.


Subject(s)
Antibodies/therapeutic use , Antigens/immunology , Lead Radioisotopes/chemistry , Proteoglycans/immunology , Triple Negative Breast Neoplasms/drug therapy , Animals , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Cell Survival , Clone Cells , Disease Models, Animal , Female , Humans , Mice, Nude , Neoplastic Stem Cells/pathology , Tissue Distribution , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
18.
Arch Oral Biol ; 90: 67-73, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29567548

ABSTRACT

Proteoglycans are glycosylated proteins which have covalently attached highly anionic glycosaminoglycans. They can be located on the extracellular matrix, cell membrane or intracellular granules. To date, few studies have reported the presence of proteoglycans in human dental pulp. OBJECTIVE: The aim of this study was, therefore, to analyze the expression of lumican, versican and glypican proteoglycans in deciduous and permanent human dental pulp by real-time polymerase chain reaction (q-PCR) and immunofluorescence. DESIGN: Healthy human dental pulps were used: 13 from permanent teeth (group 1) and eight from deciduous teeth (group 2). Versican, lumican and glypican (glypican-1 to 6) gene expressions were quantitatively evaluated by real-time PCR technique, using the expression of the endogenous gene GAPDH as control. Pulp sections were submitted to immunostaining procedure with fluorescence labelling, the tissues being fixed and incubated with well-characterized monoclonal and polyclonal antibodies against proteoglycan epitopes, including anti-versican and anti-lumican. Comparisons among the groups of the quantitative scores for each proteoglycan were analyzed using the t-test and ANOVA (P < 0.05). RESULTS: The real-time PCR analysis showed expression of versican and lumican proteoglycans in the two groups, with significant predominance of lumican gene (P = 0.03). Considering the glypican genes, glypican-3 was the proteoglycan most significantly expressed in permanent pulps (P < 0.001), while glypican-2 was not expressed in this tissue. The immunofluorescence quantification exhibited no significant differences between lumican and versican among the pulps and groups. CONCLUSIONS: The lumican gene was more expressed than versican and glypican-3 was the isoform more expressed in permanent pulp compared to deciduous.


Subject(s)
Dental Pulp/metabolism , Lumican/metabolism , Proteoglycans/metabolism , Actin Cytoskeleton , Antibodies , Dental Pulp/diagnostic imaging , Dental Pulp/pathology , Dentition, Permanent , Epitopes , Extracellular Matrix/metabolism , Gene Expression , Glypicans/genetics , Glypicans/metabolism , Humans , Lumican/genetics , Lumican/immunology , Protein Isoforms , Proteoglycans/genetics , Proteoglycans/immunology , Tooth Extraction , Tooth, Deciduous , Versicans/genetics , Versicans/metabolism
19.
J Histochem Cytochem ; 66(4): 241-259, 2018 04.
Article in English | MEDLINE | ID: mdl-29328866

ABSTRACT

Proteoglycans (PGs) are complex, multifaceted molecules that participate in diverse interactions vital for physiological and pathological processes. As structural components, they provide a scaffold for cells and structural organization that helps define tissue architecture. Through interactions with water, PGs enable molecular and cellular movement through tissues. Through selective ionic interactions with growth factors, chemokines, cytokines, and proteases, PGs facilitate the ability of these soluble ligands to regulate intracellular signaling events and to influence the inflammatory response. In addition, recent findings now demonstrate that PGs can activate danger-associated molecular patterns (DAMPs) and other signaling pathways to influence production of many of these soluble ligands, indicating a more direct role for PGs in influencing the immune response and tissue inflammation. This review will focus on PGs that are selectively expressed during lung inflammation and will examine the novel emerging concept of PGs as immunomodulatory regulators of the innate immune responses in lungs.


Subject(s)
Immunity, Innate , Lung/immunology , Pneumonia/immunology , Proteoglycans/immunology , Animals , Communicable Diseases/immunology , Communicable Diseases/pathology , Extracellular Matrix/immunology , Extracellular Matrix/pathology , Humans , Immunomodulation , Lung/pathology , Pneumonia/pathology , Signal Transduction
20.
J Immunol Methods ; 454: 6-14, 2018 03.
Article in English | MEDLINE | ID: mdl-29128424

ABSTRACT

Rheumatoid arthritis (RA) is an autoimmune disease of unknown etiology. A characteristic feature of RA is the presence of anti-citrullinated protein antibodies (ACPA). Since ACPAs are highly specific for RA and are often present before the onset of RA symptoms, they have become valuable diagnostic and prognostic. As a result, several assays for detection of ACPAs exist, which vary in sensitivity and specificity. In this study, we analyzed the reactivity of RA sera to selected peptides by solid-phase immunoassays in order to develop an ACPA assay with improved sensitivity and specificity. ACPA levels were determined with respect to sensitivity and specificity in 332 serum samples using the newly developed peptide panel, which was compared to the commercial assays CCPlus (Eurodiagnostica) and CCP3.1 (Inova Diagnostics). A primary panel (peptides 814, 33062 and 33156) was identified, which obtained a sensitivity of 71%, while the complete peptide panel reacted with 79% of RA sera screened. Total specificities of 89% and 80% were obtained for the primary peptide panel and the complete peptide panel. Sensitivities for the commercial assays ranged between 71% and 76% and specificities between 88% and 90%. These findings indicate that the generated peptide panel is optimal for ACPA detection and able to compete with commercial available assays. Collectively, this study may contribute to characterize autoimmunity towards citrullinated proteins and to the development of new and improved diagnostic assays for detection of ACPA and determination of RA.


Subject(s)
Anti-Citrullinated Protein Antibodies/metabolism , Arthritis, Rheumatoid/diagnosis , Enzyme-Linked Immunosorbent Assay/methods , Peptides/immunology , Proteoglycans/immunology , Arthritis, Rheumatoid/immunology , Early Diagnosis , Humans , Peptides/chemical synthesis , Predictive Value of Tests , Prognosis , Proteoglycans/chemical synthesis , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...