Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Am J Physiol Cell Physiol ; 325(2): C519-C537, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37399500

ABSTRACT

V3 is an isoform of the extracellular matrix (ECM) proteoglycan (PG) versican generated through alternative splicing of the versican gene such that the two major exons coding for sequences in the protein core that support chondroitin sulfate (CS) glycosaminoglycan (GAG) chain attachment are excluded. Thus, versican V3 isoform carries no GAGs. A survey of PubMed reveals only 50 publications specifically on V3 versican, so it is a very understudied member of the versican family, partly because to date there are no antibodies that can distinguish V3 from the CS-carrying isoforms of versican, that is, to facilitate functional and mechanistic studies. However, a number of in vitro and in vivo studies have identified the expression of the V3 transcript during different phases of development and in disease, and selective overexpression of V3 has shown dramatic phenotypic effects in "gain and loss of function" studies in experimental models. Thus, we thought it would be useful and instructive to discuss the discovery, characterization, and the putative biological importance of the enigmatic V3 isoform of versican.


Subject(s)
Alternative Splicing , Versicans , Extracellular Matrix , Protein Isoforms/genetics , Versicans/genetics , Humans
2.
Am J Respir Cell Mol Biol ; 59(6): 723-732, 2018 12.
Article in English | MEDLINE | ID: mdl-30095976

ABSTRACT

Pulmonary lymphangioleiomyomatosis (LAM) is a slow-progressing metastatic disease that is driven by mutations in the tumor suppressor tuberous sclerosis complex 1/2 (TSC1/2). Rapamycin inhibits LAM cell proliferation and is the only approved treatment, but it cannot cause the regression of existing lesions and can only stabilize the disease. However, in other cancers, immunotherapies such as checkpoint blockade against PD-1 and its ligand PD-L1 have shown promise in causing tumor regression and even curing some patients. Thus, we asked whether PD-L1 has a role in LAM progression. In vitro, PD-L1 expression in murine Tsc2-null cells is unaffected by mTOR inhibition with torin but can be upregulated by IFN-γ. Using immunohistochemistry and single-cell flow cytometry, we found increased PD-L1 expression both in human lung tissue from patients with LAM and in Tsc2-null lesions in a murine model of LAM. In this model, PD-L1 is highly expressed in the lung by antigen-presenting and stromal cells, and activated T cells expressing PD-1 infiltrate the affected lung. In vivo treatment with anti-PD-1 antibody significantly prolongs mouse survival in the model of LAM. Together, these data demonstrate that PD-1/PD-L1-mediated immunosuppression may occur in LAM, and suggest new opportunities for therapeutic targeting that may provide benefits beyond those of rapamycin.


Subject(s)
B7-H1 Antigen/metabolism , Lung Neoplasms/metabolism , Lung/metabolism , Lymphangioleiomyomatosis/metabolism , Tuberous Sclerosis/metabolism , Animals , Antibodies, Monoclonal/pharmacology , B7-H1 Antigen/immunology , Case-Control Studies , Cell Proliferation , Disease Models, Animal , Humans , Lung/drug effects , Lung/immunology , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lymphangioleiomyomatosis/drug therapy , Lymphangioleiomyomatosis/immunology , Lymphangioleiomyomatosis/pathology , Mice , Mice, Inbred C57BL , Tuberous Sclerosis/drug therapy , Tuberous Sclerosis/immunology , Tuberous Sclerosis/pathology , Up-Regulation
3.
J Biol Chem ; 292(1): 51-63, 2017 Jan 06.
Article in English | MEDLINE | ID: mdl-27895126

ABSTRACT

Viral infection is an exacerbating factor contributing to chronic airway diseases, such as asthma, via mechanisms that are still unclear. Polyinosine-polycytidylic acid (poly(I:C)), a Toll-like receptor 3 (TLR3) agonist used as a mimetic to study viral infection, has been shown to elicit inflammatory responses in lungs and to exacerbate pulmonary allergic reactions in animal models. Previously, we have shown that poly(I:C) stimulates lung fibroblasts to accumulate an extracellular matrix (ECM), enriched in hyaluronan (HA) and its binding partner versican, which promotes monocyte adhesion. In the current study, we aimed to determine the in vivo role of versican in mediating inflammatory responses in poly(I:C)-induced lung inflammation using a tamoxifen-inducible versican-deficient mouse model (Vcan-/- mice). In C57Bl/6 mice, poly(I:C) instillation significantly increased accumulation of versican and HA, especially in the perivascular and peribronchial regions, which were enriched in infiltrating leukocytes. In contrast, versican-deficient (Vcan-/-) lungs did not exhibit increases in versican or HA in these regions and had strikingly reduced numbers of leukocytes in the bronchoalveolar lavage fluid and lower expression of inflammatory chemokines and cytokines. Poly(I:C) stimulation of lung fibroblasts isolated from control mice generated HA-enriched cable structures in the ECM, providing a substrate for monocytic cells in vitro, whereas lung fibroblasts from Vcan-/- mice did not. Moreover, increases in proinflammatory cytokine expression were also greatly attenuated in the Vcan-/- lung fibroblasts. These findings provide strong evidence that versican is a critical inflammatory mediator during poly(I:C)-induced acute lung injury and, in association with HA, generates an ECM that promotes leukocyte infiltration and adhesion.


Subject(s)
Chemokines/metabolism , Cytokines/metabolism , Interferon Inducers/toxicity , Pneumonia/prevention & control , Poly I-C/toxicity , Versicans/physiology , Animals , Bronchoalveolar Lavage Fluid/chemistry , Cells, Cultured , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Pneumonia/chemically induced , Pneumonia/metabolism , Pneumonia/pathology
4.
Respir Res ; 19(1): 146, 2018 08 02.
Article in English | MEDLINE | ID: mdl-30071849

ABSTRACT

BACKGROUND: Airway inflammation is a hallmark of asthma. Alterations in extracellular matrix (ECM) hyaluronan (HA) content have been shown to modulate the recruitment and retention of inflammatory cells. Bronchial epithelial cells (BECs) regulate the activity of human lung fibroblasts (HLFs); however, their contribution in regulating HLF production of HA in asthma is unknown. In this study, we tested the hypothesis that BECs from asthmatic children promote the generation of a pro-inflammatory, HA-enriched ECM by HLFs, which promotes the retention of leukocytes. METHODS: BECs were obtained from well-characterized asthmatic and healthy children ages 6-18 years. HLFs were co-cultured with BECs for 96 h and samples were harvested for analysis of gene expression, synthesis and accumulation of HA, and subjected to a leukocyte adhesion assay with U937 monocytes. RESULTS: We observed increased expression of HA synthases HAS2 and HAS3 in HLFs co-cultured with asthmatic BECs. Furthermore, we demonstrated greater total accumulation and increased synthesis of HA by HLFs co-cultured with asthmatic BECs compared to healthy BEC/HLF co-cultures. ECM generated by HLFs co-cultured with asthmatic BECs displayed increased HA-dependent adhesion of leukocytes in a separate in vitro binding assay. CONCLUSIONS: Our findings demonstrate that BEC regulation of HA production by HLFs is altered in asthma, which may in turn promote the establishment of a more leukocyte-permissive ECM promoting airway inflammation in this disease.


Subject(s)
Asthma/metabolism , Bronchi/metabolism , Extracellular Matrix/metabolism , Hyaluronic Acid/biosynthesis , Leukocytes/metabolism , Respiratory Mucosa/metabolism , Adolescent , Bronchi/cytology , Child , Coculture Techniques , Female , Fibroblasts/metabolism , Humans , Lung/metabolism , Male , Respiratory Mucosa/cytology , U937 Cells
5.
Am J Physiol Lung Cell Mol Physiol ; 313(6): L1069-L1086, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28912382

ABSTRACT

Growing evidence suggests that versican is important in the innate immune response to lung infection. Our goal was to understand the regulation of macrophage-derived versican and the role it plays in innate immunity. We first defined the signaling events that regulate versican expression, using bone marrow-derived macrophages (BMDMs) from mice lacking specific Toll-like receptors (TLRs), TLR adaptor molecules, or the type I interferon receptor (IFNAR1). We show that LPS and polyinosinic-polycytidylic acid [poly(I:C)] trigger a signaling cascade involving TLR3 or TLR4, the Trif adaptor, type I interferons, and IFNAR1, leading to increased expression of versican by macrophages and implicating versican as an interferon-stimulated gene. The signaling events regulating versican are distinct from those for hyaluronan synthase 1 (HAS1) and syndecan-4 in macrophages. HAS1 expression requires TLR2 and MyD88. Syndecan-4 requires TLR2, TLR3, or TLR4 and both MyD88 and Trif. Neither HAS1 nor syndecan-4 is dependent on type I interferons. The importance of macrophage-derived versican in lungs was determined with LysM/Vcan-/- mice. These studies show increased recovery of inflammatory cells in the bronchoalveolar lavage fluid of poly(I:C)-treated LysM/Vcan-/- mice compared with control mice. IFN-ß and IL-10, two important anti-inflammatory molecules, are significantly decreased in both poly(I:C)-treated BMDMs from LysM/Vcan-/- mice and bronchoalveolar lavage fluid from poly(I:C)-treated LysM/Vcan-/- mice compared with control mice. In short, type I interferon signaling regulates versican expression, and versican is necessary for type I interferon production. These findings suggest that macrophage-derived versican is an immunomodulatory molecule with anti-inflammatory properties in acute pulmonary inflammation.


Subject(s)
Adaptor Proteins, Vesicular Transport/immunology , Immunity, Innate , Interferon-beta/immunology , Lung/immunology , Macrophages, Alveolar/immunology , Versicans/immunology , Adaptor Proteins, Vesicular Transport/genetics , Animals , Hyaluronan Synthases/genetics , Hyaluronan Synthases/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Lipopolysaccharides/toxicity , Mice , Mice, Knockout , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/immunology , Syndecan-4/genetics , Syndecan-4/immunology , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Versicans/genetics
6.
J Biol Chem ; 290(35): 21629-41, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26152723

ABSTRACT

Arterial smooth muscle cells (ASMCs) undergo phenotypic changes during development and pathological processes in vivo and during cell culture in vitro. Our previous studies demonstrated that retrovirally mediated expression of the versican V3 splice variant (V3) by ASMCs retards cell proliferation and migration in vitro and reduces neointimal thickening and macrophage and lipid accumulation in animal models of vascular injury and atherosclerosis. However, the molecular pathways induced by V3 expression that are responsible for these changes are not yet clear. In this study, we employed a microarray approach to examine how expression of V3 induced changes in gene expression and the molecular pathways in rat ASMCs. We found that forced expression of V3 by ASMCs affected expression of 521 genes by more than 1.5-fold. Gene ontology analysis showed that components of the extracellular matrix were the most significantly affected by V3 expression. In addition, genes regulating the formation of the cytoskeleton, which also serve as markers of contractile smooth muscle cells (SMCs), were significantly up-regulated. In contrast, components of the complement system, chemokines, chemokine receptors, and transcription factors crucial for regulating inflammatory processes were among the genes most down-regulated. Consistently, we found that the level of myocardin, a key transcription factor promoting contractile SMC phenotype, was greatly increased, and the proinflammatory transcription factors NFκB1 and CCAAT/enhancer-binding protein ß were significantly attenuated in V3-expressing SMCs. Overall, these findings demonstrate that V3 expression reprograms ASMCs promoting differentiated and anti-inflammatory phenotypes.


Subject(s)
Anti-Inflammatory Agents/metabolism , Arteries/cytology , Cell Differentiation , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Versicans/metabolism , Animals , Apoptosis/genetics , Biomarkers/metabolism , Cell Survival/genetics , Cellular Microenvironment , Cluster Analysis , Down-Regulation/genetics , Gene Expression Profiling , Inflammation/genetics , Inflammation/pathology , Molecular Sequence Annotation , Oligonucleotide Array Sequence Analysis , Phenotype , Rats, Inbred F344 , Response Elements/genetics , Software , Up-Regulation/genetics , Versicans/genetics
7.
Arterioscler Thromb Vasc Biol ; 35(12): 2647-56, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26494233

ABSTRACT

OBJECTIVE: Prenatal deletion of the type II transforming growth factor-ß (TGF-ß) receptor (TBRII) prevents normal vascular morphogenesis and smooth muscle cell (SMC) differentiation, causing embryonic death. The role of TBRII in adult SMC is less well studied. Clarification of this role has important clinical implications because TBRII deletion should ablate TGF-ß signaling, and blockade of TGF-ß signaling is envisioned as a treatment for human aortopathies. We hypothesized that postnatal loss of SMC TBRII would cause aortopathy. APPROACH AND RESULTS: We generated mice with either of 2 tamoxifen-inducible SMC-specific Cre (SMC-CreER(T2)) alleles and homozygous floxed Tgfbr2 alleles. Mice were injected with tamoxifen, and their aortas examined 4 and 14 weeks later. Both SMC-CreER(T2) alleles efficiently and specifically rearranged a floxed reporter gene and efficiently rearranged a floxed Tgfbr2 allele, resulting in loss of aortic medial TBRII protein. Loss of SMC TBRII caused severe aortopathy, including hemorrhage, ulceration, dissection, dilation, accumulation of macrophage markers, elastolysis, abnormal proteoglycan accumulation, and aberrant SMC gene expression. All areas of the aorta were affected, with the most severe pathology in the ascending aorta. Cre-mediated loss of SMC TBRII in vitro ablated both canonical and noncanonical TGF-ß signaling and reproduced some of the gene expression abnormalities detected in vivo. CONCLUSIONS: SMC TBRII plays a critical role in maintaining postnatal aortic homeostasis. Loss of SMC TBRII disrupts TGF-ß signaling, acutely alters SMC gene expression, and rapidly results in severe and durable aortopathy. These results suggest that pharmacological blockade of TGF-ß signaling in humans could cause aortic disease rather than prevent it.


Subject(s)
Aortic Diseases/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Protein Serine-Threonine Kinases/deficiency , Receptors, Transforming Growth Factor beta/deficiency , Age Factors , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Cell Proliferation , Elastin/metabolism , Extracellular Matrix/metabolism , Gene Expression Regulation , Genetic Predisposition to Disease , Macrophages/metabolism , Macrophages/pathology , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Phenotype , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction , Transforming Growth Factor beta1/pharmacology
8.
J Biol Chem ; 289(22): 15393-404, 2014 May 30.
Article in English | MEDLINE | ID: mdl-24719328

ABSTRACT

Monocyte/macrophage accumulation plays a critical role during progression of cardiovascular diseases, such as atherosclerosis. Our previous studies demonstrated that retrovirally mediated expression of the versican V3 splice variant (V3) by arterial smooth muscle cells (ASMCs) decreases monocyte adhesion in vitro and macrophage accumulation in a model of lipid-induced neointimal formation in vivo. We now demonstrate that V3-expressing ASMCs resist monocyte adhesion by altering the composition of the microenvironment surrounding the cells by affecting multiple signaling pathways. Reduction of monocyte adhesion to V3-expressing ASMCs is due to the generation of an extracellular matrix enriched in elastic fibers and depleted in hyaluronan, and reduction of the proinflammatory cell surface vascular cell adhesion molecule 1 (VCAM1). Blocking these changes reverses the protective effect of V3 on monocyte adhesion. The enhanced elastogenesis induced by V3 expression is mediated by TGFß signaling, whereas the reduction in hyaluronan cable formation induced by V3 expression is mediated by the blockade of epidermal growth factor receptor and NFκB activation pathways. In addition, expression of V3 by ASMCs induced a marked decrease in NFκB-responsive proinflammatory cell surface molecules that mediate monocyte adhesion, such as VCAM1. Overall, these results indicate that V3 expression by ASMCs creates a microenvironment resistant to monocyte adhesion via differentially regulating multiple signaling pathways.


Subject(s)
Cell Adhesion/immunology , Inflammation/metabolism , Monocytes/metabolism , Muscle, Smooth, Vascular/metabolism , Versicans/metabolism , Animals , Cells, Cultured , Cellular Microenvironment/immunology , Epidermal Growth Factor/metabolism , Inflammation/immunology , Monocytes/cytology , Monocytes/immunology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/immunology , NF-kappa B/metabolism , Rats , Rats, Inbred F344 , Signal Transduction/immunology , Transforming Growth Factor beta1/metabolism , Versicans/immunology
9.
Arterioscler Thromb Vasc Biol ; 31(6): 1309-16, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21441139

ABSTRACT

OBJECTIVE: Extracellular matrix (ECM) of neointima formed following angioplasty contains elevated levels of versican, loosely arranged collagen, and fragmented deposits of elastin, features associated with lipid and macrophage accumulation. ECM with a low versican content, compact structure, and increased elastic fiber content can be achieved by expression of versican variant V3, which lacks chondroitin sulfate glycosaminoglycans. We hypothesized that V3-expressing arterial smooth muscle cells (ASMC) can be used to form a neointima resistant to lipid and macrophage accumulation associated with hypercholesterolemia. METHODS AND RESULTS: ASMC transduced with V3 cDNA were seeded into ballooned rabbit carotid arteries, and animals were fed a chow diet for 4 weeks, followed by a cholesterol-enriched diet for 4 weeks, achieving plasma cholesterol levels of 20 to 25 mmol/L. V3 neointimae at 8 weeks were compact, multilayered, and elastin enriched. They were significantly thinner (57%) than control neointimae; contained significantly more elastin (118%), less collagen (22%), and less lipid (76%); and showed significantly reduced macrophage infiltration (85%). Mechanistic studies demonstrated that oxidized low-density lipoprotein stimulated the formation of a monocyte-binding ECM, which was inhibited in the presence of V3 expressing ASMC. CONCLUSION: These results demonstrate that expression of V3 in vessel wall creates an elastin-rich neointimal matrix that in the presence of hyperlipidemia is resistant to lipid deposition and macrophage accumulation.


Subject(s)
Macrophages/physiology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/physiology , Neointima/metabolism , Versicans/physiology , Animals , Arteries/cytology , Cells, Cultured , Extracellular Matrix/metabolism , Lipid Metabolism , Lipoproteins, LDL/toxicity , Male , Microscopy, Electron , Neointima/pathology , Rabbits , Versicans/analysis
10.
Matrix Biol Plus ; 14: 100110, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35573706

ABSTRACT

The content and organization of hyaluronan (HA) in the extracellular matrix (ECM) have been identified as strong indicators of inflammation in joint disease, although the source and role of HA as an effector of inflammation is not clear. In this study, we established co-cultures of activated human CD4 T cells with fibroblast-like synoviocytes (FLS) from osteoarthritis (OA) and rheumatoid arthritis (RA) subjects and examined the role of HA in promoting inflammatory events. Co-cultures of RA FLS with activated CD4 T cells generated an HA-enriched ECM that promoted enhanced monocyte adhesion compared to co-cultures of OA FLS with activated CD4 T cells. In addition, both OA FLS and RA FLS co-cultures with activated CD4 T cells elicited significant increases in the expression of IL1ß, TNF, and IL6, with the increase in IL6 expression most prominent in RA co-cultures. Blocking HA synthesis and accumulation with 4-methylumbelliferone reduced expression of IL6, IL1ß, and TNF in both OA FLS and RA FLS co-cultures. The increase in HA synthesis in the co-cultures was mimicked by IL6 trans-signaling of FLS in the absence of CD4 T cells. Inhibition of HA synthesis blocked the increase in IL6 by RA FLS mediated by IL6 trans-signaling, suggesting that the HA synthetic pathway may be a key mediator in IL6 expression by FLS. Overall, our study indicates that HA-enriched ECM generated by co-cultures of activated CD4 T cells with FLS from human joints creates a pathogenic microenvironment by promoting adhesion of leukocytes and expression of inflammatory cytokines including IL6.

11.
Am J Respir Cell Mol Biol ; 43(5): 591-8, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20023207

ABSTRACT

Neutrophil adhesion to pulmonary microvascular endothelial cells (ECs) initiates intracellular signaling, resulting in remodeling of F-actin cytoskeletal structure of ECs. The present study determined the mechanical properties of ECs and the changes induced by neutrophil adhesion by atomic force microscopy. The elastic moduli of ECs were compared before neutrophils were present, as soon as neutrophil adhesion was detected, and 1 minute later. ECs that were adjacent to those with adherent neutrophils were also evaluated. Neutrophil adhesion induced a decrease in the elastic moduli in the 6.25-µm rim of ECs surrounding adherent neutrophils as soon as firmly adherent neutrophils were detected, which was transient and lasted less than 1 minute. Adjacent ECs developed an increase in stiffness that was significant in the central regions of these cells. Intercellular adhesion molecule-1 crosslinking did not induce significant changes in the elastic modulus of ECs in either region, suggesting that crosslinking intercellular adhesion molecule-1 is not sufficient to induce the observed changes. Our results demonstrate that neutrophil adhesion induces regional changes in the stiffness of ECs.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/physiology , Lung/blood supply , Lung/cytology , Microvessels/cytology , Neutrophils/cytology , Biomechanical Phenomena/drug effects , Cell Adhesion/drug effects , Cell Movement/drug effects , Cross-Linking Reagents/pharmacology , Elastic Modulus/drug effects , Endothelial Cells/drug effects , Humans , Intercellular Adhesion Molecule-1/metabolism , Neutrophils/drug effects , Tumor Necrosis Factor-alpha/pharmacology
12.
JCI Insight ; 5(20)2020 09 24.
Article in English | MEDLINE | ID: mdl-32970631

ABSTRACT

The ability of HDL to inhibit inflammation in adipocytes and adipose tissue is reduced when HDL contains serum amyloid A (SAA) that is trapped by proteoglycans at the adipocyte surface. Because we recently found that the major extracellular matrix proteoglycan produced by hypertrophic adipocytes is versican, whereas activated adipose tissue macrophages produce mainly biglycan, we further investigated the role of proteoglycans in determining the antiinflammatory properties of HDL. The distributions of versican, biglycan, apolipoprotein A1 (the major apolipoprotein of HDL), and SAA were similar in adipose tissue from obese mice and obese human subjects. Colocalization of SAA-enriched HDL with versican and biglycan at the cell surface of adipocyte and peritoneal macrophages, respectively, was blocked by silencing these proteoglycans, which also restored the antiinflammatory property of SAA-enriched HDL despite the presence of SAA. Similar to adipocytes, normal HDL exerted its antiinflammatory function in macrophages by reducing lipid rafts, reactive oxygen species generation, and translocation of Toll-like receptor 4 and NADPH oxidase 2 into lipid rafts, effects that were not observed with SAA-enriched HDL. These findings imply that SAA present in HDL can be trapped by adipocyte-derived versican and macrophage-derived biglycan, thereby blunting HDL's antiinflammatory properties.


Subject(s)
Adipocytes/immunology , Biglycan/immunology , Lipoproteins, HDL/immunology , Macrophages, Peritoneal/immunology , Obesity/immunology , Serum Amyloid A Protein/immunology , Versicans/immunology , Adipocytes/pathology , Adult , Animals , Apolipoprotein A-I/genetics , Apolipoprotein A-I/immunology , Biglycan/antagonists & inhibitors , Biglycan/genetics , Diet, High-Fat/adverse effects , Female , Gene Expression Regulation , Humans , Insulin Resistance/immunology , Lipoproteins, HDL/genetics , Macrophages, Peritoneal/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Obesity/etiology , Obesity/genetics , Obesity/pathology , Protein Binding , Protein Transport , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Serum Amyloid A Protein/genetics , Silver Nitrate/administration & dosage , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Versicans/antagonists & inhibitors , Versicans/genetics
13.
J Histochem Cytochem ; 68(11): 797-811, 2020 11.
Article in English | MEDLINE | ID: mdl-33064036

ABSTRACT

Hyaluronan and proteoglycan link protein 1 (HAPLN1) stabilizes interactions between two important extracellular matrix (ECM) macromolecules, versican and hyaluronan, which facilitate proliferation of fibroblasts and their conversion to myofibroblasts. However, the role of HAPLN1 in these events has not been studied. Using immunocytochemistry, cellular and ECM locations of HAPLN1 were evaluated in cultured human lung fibroblasts during proliferation and conversion to myofibroblasts. HAPLN1 localized to pericellular matrices, associating with both versican and hyaluronan in the ECM and on the cell surface. Nuclear and total HAPLN1 immunostaining increased after myofibroblast induction. Confocal microscopy showed HAPLN1 predominant in the ECM under cells while versican predominated above cells. Versican and HAPLN1 were also juxtaposed in columnar inclusions in the cytoplasm and nucleus. Nuclear HAPLN1 staining in interphase cells redistributed to the cytosol during mitosis. In the absence of TGF-ß1, addition of exogenous bovine HAPLN1 (together with aggrecan G1) facilitated myofibroblast formation, as seen by significant upregulation of α-smooth muscle actin (SMA) staining, while adding full-length bovine versican had no effect. Increased compaction of hyaluronan-rich ECM suggests that HAPLN1 plus G1 addition affects hyaluronan networks and myofibroblast formation. These observations demonstrate changes in both extracellular and intracellular localization of HAPLN1 during fibroblast proliferation and myofibroblast conversion suggesting a possible role in fibrotic remodeling.


Subject(s)
Extracellular Matrix Proteins/metabolism , Fibroblasts/metabolism , Lung/cytology , Proteoglycans/metabolism , Cell Proliferation , Cells, Cultured , Fibroblasts/cytology , Humans , Phenotype , Versicans/metabolism
14.
Front Immunol ; 11: 512, 2020.
Article in English | MEDLINE | ID: mdl-32265939

ABSTRACT

The extracellular matrix (ECM) proteoglycan, versican increases along with other ECM versican binding molecules such as hyaluronan, tumor necrosis factor stimulated gene-6 (TSG-6), and inter alpha trypsin inhibitor (IαI) during inflammation in a number of different diseases such as cardiovascular and lung disease, autoimmune diseases, and several different cancers. These interactions form stable scaffolds which can act as "landing strips" for inflammatory cells as they invade tissue from the circulation. The increase in versican is often coincident with the invasion of leukocytes early in the inflammatory process. Versican interacts with inflammatory cells either indirectly via hyaluronan or directly via receptors such as CD44, P-selectin glycoprotein ligand-1 (PSGL-1), and toll-like receptors (TLRs) present on the surface of immune and non-immune cells. These interactions activate signaling pathways that promote the synthesis and secretion of inflammatory cytokines such as TNFα, IL-6, and NFκB. Versican also influences inflammation by interacting with a variety of growth factors and cytokines involved in regulating inflammation thereby influencing their bioavailability and bioactivity. Versican is produced by multiple cell types involved in the inflammatory process. Conditional total knockout of versican in a mouse model of lung inflammation demonstrated significant reduction in leukocyte invasion into the lung and reduced inflammatory cytokine expression. While versican produced by stromal cells tends to be pro-inflammatory, versican expressed by myeloid cells can create anti-inflammatory and immunosuppressive microenvironments. Inflammation in the tumor microenvironment often contains elevated levels of versican. Perturbing the accumulation of versican in tumors can inhibit inflammation and tumor progression in some cancers. Thus versican, as a component of the ECM impacts immunity and inflammation through regulating immune cell trafficking and activation. Versican is emerging as a potential target in the control of inflammation in a number of different diseases.


Subject(s)
Extracellular Matrix/immunology , Hyaluronic Acid/physiology , Inflammation/metabolism , Versicans/physiology , Animals , Humans , Inflammation/immunology , Leukocytes/immunology , Lipopolysaccharides/pharmacology , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Mice , Models, Animal , Myeloid Cells/immunology , Myeloid Cells/metabolism , Rabbits , Rats , Receptors, Cell Surface/physiology , Stromal Cells/immunology , Stromal Cells/ultrastructure , Toll-Like Receptors/agonists , Versicans/deficiency
15.
Cell Rep ; 31(13): 107818, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32610121

ABSTRACT

Obesity is characterized by adipose tissue inflammation. Because proteoglycans regulate inflammation, here we investigate their role in adipose tissue inflammation in obesity. We find that adipose tissue versican and biglycan increase in obesity. Versican is produced mainly by adipocytes and biglycan by adipose tissue macrophages. Both proteoglycans are also present in adipose tissue from obese human subjects undergoing gastric bypass surgery. Deletion of adipocyte-specific versican or macrophage-specific biglycan in mice reduces macrophage accumulation and chemokine and cytokine expression, although only adipocyte-specific versican deletion leads to sustained improvement in glucose tolerance. Macrophage-derived biglycan activates inflammatory genes in adipocytes. Versican expression increases in cultured adipocytes exposed to excess glucose, and adipocyte-conditioned medium stimulates inflammation in resident peritoneal macrophages, in part because of a versican breakdown product, versikine. These findings provide insights into the role of adipocyte- and macrophage-derived proteoglycans in adipose tissue inflammation in obesity.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/pathology , Biglycan/metabolism , Inflammation/pathology , Macrophages/metabolism , Obesity/pathology , Versicans/metabolism , 3T3-L1 Cells , Animals , Bone Marrow/metabolism , Diet, High-Fat , Female , Glucose Tolerance Test , Humans , Hypertrophy , Insulin Resistance , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Middle Aged , Omentum/metabolism , Organ Specificity , RNA, Messenger/genetics , RNA, Messenger/metabolism , Subcutaneous Fat/pathology , Versicans/genetics
16.
Matrix Biol ; 89: 27-42, 2020 07.
Article in English | MEDLINE | ID: mdl-32001344

ABSTRACT

Versican is a large extracellular matrix (ECM) chondroitin sulfate (CS) proteoglycan found in most soft tissues, which is encoded by the VCAN gene. At least four major isoforms (V0, V1, V2, and V3) are generated via alternative splicing. The isoforms of versican are expressed and accumulate in various tissues during development and disease, where they contribute to ECM structure, cell growth and migration, and immune regulation, among their many functions. While several studies have identified the mRNA transcript for the V3 isoform in a number of tissues, little is known about the synthesis, secretion, and targeting of the V3 protein. In this study, we used lentiviral generation of doxycycline-inducible rat V3 with a C-terminal tag in stable NIH 3T3 cell lines and demonstrated that V3 is processed through the classical secretory pathway. We further show that N-linked glycosylation is required for efficient secretion and solubility of the protein. By site-directed mutagenesis, we identified amino acids 57 and 330 as the active N-linked glycosylation sites on V3 when expressed in this cell type. Furthermore, exon deletion constructs of V3 revealed that exons 11-13, which code for portions of the carboxy region of the protein (G3 domain), are essential for V3 processing and secretion. Once secreted, the V3 protein associates with hyaluronan along the cell surface and within the surrounding ECM. These results establish critical parameters for the processing, solubility, and targeting of the V3 isoform by mammalian cells and establishes a role for V3 in the organization of hyaluronan.


Subject(s)
Versicans/chemistry , Versicans/metabolism , Alternative Splicing , Animals , Exons , Glycosylation , HEK293 Cells , Humans , Mice , Mutagenesis, Site-Directed , NIH 3T3 Cells , Protein Domains , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Rats , Versicans/genetics
17.
Front Immunol ; 10: 3159, 2019.
Article in English | MEDLINE | ID: mdl-32047499

ABSTRACT

Human lung fibroblasts (HLFs) treated with the viral mimetic polyinosine-polycytidylic acid (poly I:C) form an extracellular matrix (ECM) enriched in hyaluronan (HA) that avidly binds monocytes and lymphocytes. Mast cells are important innate immune cells in both asthma and acute respiratory infections including respiratory syncytial virus (RSV); however, the effect of RSV on HA dependent mast cell adhesion and/or function is unknown. To determine if RSV infection of HLFs leads to the formation of a HA-enriched ECM that binds and enhances mast cell activity primary HLFs were infected with RSV for 48 h prior to leukocyte binding studies using a fluorescently labeled human mast cell line (LUVA). Parallel HLFs were harvested for characterization of HA production by ELISA and size exclusion chromatography. In separate experiments, HLFs were infected as above for 48 h prior to adding LUVA cells to HLF wells. Co-cultures were incubated for 48 h at which point media and cell pellets were collected for analysis. The role of the hyaladherin tumor necrosis factor-stimulated gene 6 (TSG-6) was also assessed using siRNA knockdown. RSV infection of primary HLFs for 48 h enhanced HA-dependent LUVA binding assessed by quantitative fluorescent microscopy. This coincided with increased HLF HA synthase (HAS) 2 and HAS3 expression and decreased hyaluronidase (HYAL) 2 expression leading to increased HA accumulation in the HLF cell layer and the presence of larger HA fragments. Separately, LUVAs co-cultured with RSV-infected HLFs for 48 h displayed enhanced production of the mast cell proteases, chymase, and tryptase. Pre-treatment with the HA inhibitor 4-methylumbelliferone (4-MU) and neutralizing antibodies to CD44 (HA receptor) decreased mast cell protease expression in co-cultured LUVAs implicating a direct role for HA. TSG-6 expression was increased over the 48-h infection. Inhibition of HLF TSG-6 expression by siRNA knockdown led to decreased LUVA binding suggesting an important role for this hyaladherin for LUVA adhesion in the setting of RSV infection. In summary, RSV infection of HLFs contributes to inflammation via HA-dependent mechanisms that enhance mast cell binding as well as mast cell protease expression via direct interactions with the ECM.


Subject(s)
Extracellular Matrix/immunology , Fibroblasts , Hyaluronic Acid/metabolism , Mast Cells , Respiratory Syncytial Virus Infections/immunology , Cell Adhesion/immunology , Cells, Cultured , Chymases/biosynthesis , Coculture Techniques , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Fibroblasts/immunology , Fibroblasts/metabolism , Fibroblasts/virology , Humans , Lung/immunology , Lung/virology , Mast Cells/immunology , Mast Cells/metabolism , Respiratory Syncytial Virus, Human , Tryptases/biosynthesis
18.
Biophys J ; 94(8): 3273-85, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18199670

ABSTRACT

Mechanical properties of living cells can be determined using atomic force microscopy (AFM). In this study, a novel analysis was developed to determine the mechanical properties of adherent monolayers of pulmonary microvascular endothelial cells (ECs) using AFM and finite element modeling, which considers both the finite thickness of ECs and their nonlinear elastic properties, as well as the large strain induced by AFM. Comparison of this model with the more traditional Hertzian model, which assumes linear elastic behavior, small strains, and infinite cell thickness, suggests that the new analysis can predict the mechanical response of ECs during AFM indentation better than Hertz's model, especially when using force-displacement data obtained from large indentations (>100 nm). The shear moduli and distensibility of ECs were greater when using small indentations (<100 nm) compared to large indentations (>100 nm). Tumor necrosis factor-alpha induced changes in the mechanical properties of ECs, which included a decrease in the average shear moduli that occurred in all regions of the ECs and an increase in distensibility in the central regions when measured using small indentations. These changes can be modeled as changes in a chain network structure within the ECs.


Subject(s)
Endothelial Cells/physiology , Mechanotransduction, Cellular/physiology , Tumor Necrosis Factor-alpha/pharmacology , Cells, Cultured , Elasticity/drug effects , Endothelial Cells/drug effects , Humans , Mechanotransduction, Cellular/drug effects , Stress, Mechanical
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.
Thromb Res ; 119(6): 731-40, 2007.
Article in English | MEDLINE | ID: mdl-17010412

ABSTRACT

Adsorption of plasma proteins such as von Willebrand factor (vWF) on thrombogenic surfaces can induce conformational changes in tertiary structure so that the prothrombotic functional epitopes are exposed for interactions with platelets, resulting in platelet adhesion and thrombus formation. Thus, understanding platelet binding following changes in the structure of vWF is critical in understanding the mechanisms of thrombogenesis. The present study examined the accessibility of platelet binding epitopes within vWF adsorbed on two different thrombogenic surfaces, a hydrophobic synthetic surface and collagen VI coated substrates, under physiological buffer conditions using atomic force microscopy (AFM) in combination with immunogold labeling. Our results demonstrated that the glycoprotein Ib (GPIb) binding domain in vWF undergoes changes when adsorbed on collagen VI compared to vWF on a hydrophobic synthetic surface. This study provides a basis for a novel approach to understand the molecular mechanisms of surface-induced thrombosis by directly examining the structure-function relationships of plasma proteins involved in the thrombus formation.


Subject(s)
Blood Platelets/metabolism , Platelet Glycoprotein GPIb-IX Complex/metabolism , von Willebrand Factor/genetics , von Willebrand Factor/metabolism , Adsorption , Binding Sites , Collagen Type VI/chemistry , Epitopes , Humans , Immunohistochemistry , Microscopy, Atomic Force , Protein Structure, Tertiary , Silanes/chemistry , Structure-Activity Relationship , Surface Properties , Thrombosis/etiology , von Willebrand Factor/immunology , von Willebrand Factor/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL