Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
Am J Physiol Renal Physiol ; 312(6): F1101-F1111, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28228399

ABSTRACT

Thickening of the glomerular basement membrane (GBM) and expansion of the mesangial matrix are hallmarks of diabetic nephropathy (DN), generally considered to emerge from different sites of overproduction: GBM components from podocytes and mesangial matrix from mesangial cells. Reevaluation of 918 biopsies with DN revealed strong evidence that these mechanisms are connected to each other, wherein excess GBM components fail to undergo degradation and are deposited in the mesangium. These data do not exclude that mesangial cells also synthesize components that contribute to the accumulation of matrix in the mesangium. Light, electron microscopic, immunofluorescence, and in situ hybridization studies clearly show that the thickening of the GBM is due not only to overproduction of components of the mature GBM (α3 and α5 chains of collagen IV and agrin) by podocytes but also to resumed increased synthesis of the α1 chain of collagen IV and of perlecan by endothelial cells usually seen during embryonic development. We hypothesize that these abnormal production mechanisms are caused by different processes: overproduction of mature GBM-components by the diabetic milieu and regression of endothelial cells to an embryonic production mode by decreased availability of mediators from podocytes.


Subject(s)
Diabetic Nephropathies/pathology , Glomerular Basement Membrane/ultrastructure , Glomerular Mesangium/ultrastructure , Podocytes/ultrastructure , Agrin/analysis , Autoantigens/analysis , Biopsy , Cellular Microenvironment , Collagen Type IV/analysis , Diabetic Nephropathies/metabolism , Disease Progression , Glomerular Basement Membrane/chemistry , Glomerular Mesangium/chemistry , Heparan Sulfate Proteoglycans/analysis , Humans , Immunohistochemistry , Microscopy, Electron, Transmission , Podocytes/chemistry , Sclerosis
2.
Lab Invest ; 96(3): 283-95, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26568297

ABSTRACT

Adeno-associated virus (AAV) vector-mediated delivery of inhibitors of blood-retinal barrier breakdown (BRBB) offers promise for the treatment of diabetic macular edema. Here, we demonstrated a reversal of blood-retinal barrier pathology mediated by AAV type 2 (AAV2) vectors encoding vasoinhibin or soluble VEGF receptor 1 (sFlt-1) when administered intravitreally to diabetic rats. Efficacy and safety of the AAV2 vasoinhibin vector were tested by monitoring its effect on diabetes-induced changes in the retinal vascular bed and thickness, and in the electroretinogram (ERG). Also, the transduction of AAV2 vectors and expression of AAV2 receptors and co-receptors were compared between the diabetic and the non-diabetic rat retinas. AAV2 vasoinhibin or AAV2 sFlt-1 vectors were injected intravitreally before or after enhanced BRBB due to diabetes induced by streptozotocin. The BRBB was examined by the Evans blue method, the vascular bed by fluorescein angiography, expression of the AAV2 EGFP reporter vector by confocal microscopy, and the AAV2 genome, expression of transgenes, receptors, and co-receptors by quantitative PCR. AAV2 vasoinhibin and sFlt-1 vectors inhibited the diabetes-mediated increase in BRBB when injected after, but not before, diabetes was induced. The AAV2 vasoinhibin vector decreased retinal microvascular abnormalities and the diabetes-induced reduction of the B-wave of the ERG, but it had no effect in non-diabetic controls. Also, retinal thickness was not altered by diabetes or by the AAV2 vasoinhibin vector. The AAV2 genome, vasoinhibin and sFlt-1 transgenes, and EGFP levels were higher in the retinas from diabetic rats and were associated with an elevated expression of AAV2 receptors (syndecan, glypican, and perlecan) and co-receptors (fibroblast growth factor receptor 1, αvß5 integrin, and hepatocyte growth factor receptor). We conclude that retinal transduction and efficacy of AAV2 vectors are enhanced in diabetes, possibly due to their elevated cell entry. AAV2 vectors encoding vasoinhibin and sFlt-1 may be desirable gene therapeutics to target diabetic retinopathy and macular edema.


Subject(s)
Cell Cycle Proteins/genetics , Dependovirus/genetics , Diabetes Mellitus, Experimental/therapy , Diabetic Retinopathy/therapy , Genetic Therapy , Retina/metabolism , Vascular Endothelial Growth Factor Receptor-1/genetics , Animals , Blood-Retinal Barrier , Genetic Vectors , Heparan Sulfate Proteoglycans/analysis , Male , Rats , Rats, Wistar , Streptozocin
3.
Cardiovasc Res ; 107(1): 20-31, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25952902

ABSTRACT

AIMS: Excessive vascular cell proliferation is an important component of pulmonary hypertension (PH). Perlecan is the major heparan sulfate (HS) proteoglycan in the vascular extracellular matrix. It binds growth factors, including FGF2, and either restricts or promotes cell proliferation. In this study, we have explored the effects of perlecan HS deficiency on pulmonary vascular development and in hypoxia-induced PH. METHODS AND RESULTS: In normoxia, Hspg2(Δ3/Δ3) mice, deficient in perlecan HS, had reduced pericytes and muscularization of intra-acinar vessels. Pulmonary angiography revealed a peripheral perfusion defect. Despite these abnormalities, right ventricular systolic pressure (RVSP) and myocardial mass remained normal. After 4 weeks of hypoxia, increases in the proportion of muscularized vessels, RVSP, and right ventricular hypertrophy were significantly less in Hspg2(Δ3/Δ3) compared with wild type. The early phase of hypoxia induced a significantly lower increase in fibroblast growth factor receptor-1 (FGFR1) protein level and receptor phosphorylation, and reduced pulmonary artery smooth muscle cell (PASMC) proliferation in Hspg2(Δ3/Δ3). At 4 weeks, FGF2 mRNA and protein were also significantly reduced in Hspg2(Δ3/Δ3) lungs. Ligand and carbohydrate engagement assay showed that perlecan HS is required for HS-FGF2-FGFR1 ternary complex formation. In vitro, proliferation assays showed that PASMC proliferation is reduced by selective FGFR1 inhibition. PASMC adhesion to fibronectin was higher in Hspg2(Δ3/Δ3) compared with wild type. CONCLUSIONS: Perlecan HS chains are important for normal vascular arborization and recruitment of pericytes to pulmonary vessels. Perlecan HS deficiency also attenuates hypoxia-induced PH, where the underlying mechanisms involve impaired FGF2/FGFR1 interaction, inhibition of PASMC growth, and altered cell-matrix interactions.


Subject(s)
Heparan Sulfate Proteoglycans/metabolism , Hypertension, Pulmonary/etiology , Hypoxia/complications , Pulmonary Artery/physiology , Animals , Cell Adhesion , Cell Proliferation , Cells, Cultured , Female , Fibroblast Growth Factor 2/analysis , Heparan Sulfate Proteoglycans/analysis , Heparan Sulfate Proteoglycans/deficiency , Hypertension, Pulmonary/prevention & control , Mice , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/physiology , Phosphorylation , Pulmonary Artery/diagnostic imaging , Radiography , Receptor, Fibroblast Growth Factor, Type 1/metabolism
4.
J Biol Chem ; 290(11): 7027-39, 2015 Mar 13.
Article in English | MEDLINE | ID: mdl-25589788

ABSTRACT

Hageman factor (FXIIa) initiates the intrinsic coagulation pathway and triggers the kallikrein-kinin and the complement systems. In addition, it functions as a growth factor by expressing promitogenic activities toward several cell types. FXIIa binds to the cell surface via a number of structurally unrelated surface receptors; however, the underlying mechanisms are not yet fully understood. Here, we demonstrate that FXIIa utilizes cell membrane-bound glycosaminoglycans to interact with the cell surface of human lung fibroblasts (HLF). The combination of enzymatic, inhibitory, and overexpression approaches identified a heparan sulfate (HS) component of proteoglycans as an important determinant of the FXIIa binding capacity of HLF. Moreover, cell-free assays and competition experiments revealed preferential binding of FXIIa to HS and heparin over dextran sulfate, dermatan sulfate, and chondroitin sulfate A and C. Finally, we demonstrate that fibroblasts isolated from the lungs of the patients suffering from idiopathic pulmonary fibrosis (IPF) exhibit enhanced FXIIa binding capacity. Increased sulfation of HS resulting from elevated HS 6-O-sulfotransferase-1 expression in IPF HLF accounted, in part, for this phenomenon. Application of RNA interference technology and inhibitors of intracellular sulfation revealed the cooperative action of cell surface-associated HS and urokinase-type plasminogen activator receptor in the accumulation of FXIIa on the cell surface of IPF HLF. Moreover, FXIIa stimulated IPF HLF migration, which was abrogated by pretreatment of cells with heparinase I. Collectively, our study uncovers a novel role of HS-type glycosaminoglycans in a local accumulation of FXIIa on the cell membrane. The enhanced association of FXIIa with IPF HLF suggests its contribution to fibrogenesis.


Subject(s)
Factor XIIa/metabolism , Fibroblasts/pathology , Heparan Sulfate Proteoglycans/metabolism , Lung/pathology , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Cells, Cultured , Factor XIIa/analysis , Fibroblasts/metabolism , Heparan Sulfate Proteoglycans/analysis , Humans , Lung/metabolism , Protein Binding
5.
J Cereb Blood Flow Metab ; 34(10): 1637-45, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25052558

ABSTRACT

In our juvenile traumatic brain injury (jTBI) model, emergence of cognitive dysfunctions was observed up to 6 months after trauma. Here we hypothesize that early brain injury induces changes in the neurovascular unit (NVU) that would be associated with amyloid-beta (Aß) accumulation. We investigated NVU changes for up to 6 months in a rat jTBI model, with a focus on the efflux protein P-glycoprotein (P-gp) and on the basement membrane proteins perlecan and fibronectin, all known to be involved in Aß clearance. Rodent-Aß staining is present and increased after jTBI around cerebral blood microvessels, and the diameter of those is decreased by 25% and 34% at 2 and 6 months, respectively, without significant angiogenesis. P-glycoprotein staining in endothelium is decreased by 22% and parallels an increase of perlecan and fibronectin staining around cerebral blood vessels. Altogether, these results strongly suggest that the emergence of long-term behavioral dysfunctions observed in rodent jTBI may be related to endothelial remodeling at the blood-brain barrier alongside vascular dysfunction and altered Aß trafficking. This study shows that it is important to consider jTBI as a vascular disorder with long-term consequences on cognitive functions.


Subject(s)
Amyloid beta-Peptides/metabolism , Brain Injuries/pathology , Brain/blood supply , Brain/pathology , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain/metabolism , Brain Injuries/metabolism , Fibronectins/analysis , Fibronectins/metabolism , Heparan Sulfate Proteoglycans/analysis , Heparan Sulfate Proteoglycans/metabolism , Male , Microcirculation , Rats , Rats, Sprague-Dawley
6.
Gynecol Obstet Invest ; 73(4): 277-84, 2012.
Article in English | MEDLINE | ID: mdl-22516801

ABSTRACT

BACKGROUND/AIMS: Pre-eclampsia (PE) is one of the leading causes of maternal and perinatal morbidity and mortality. PE is defined clinically as the onset of maternal hypertension and proteinuria following 20 weeks of gestation. It is associated with altered maternal uterine decidual spiral artery remodelling, which may lead to reduced blood flow and increased thrombosis within the uteroplacental vasculature. Proteoglycans (PGs) are macromolecules which have (in combination with glycosaminoglycans) important anticoagulant roles in vascular endothelial environments, including the uteroplacental circulation. The hypothesis under consideration in this study was that differential expression of placental PGs may be associated with PE. METHODS: PE and control placental samples were collected with ethics approval and patient consent. RNA and protein were extracted and real-time PCR and Western immunoblotting were performed to determine the expression of the PGs in the samples. RESULTS: Of the nine PGs investigated, none showed increased expression, whereas the mRNA and protein expression of five of them was significantly decreased in the placentae of pre-eclamptic women compared to gestation-matched controls. CONCLUSION: Therefore, the results of this study support the hypothesis that a placental PG deficiency may contribute to the placental thrombotic lesions characteristic of PE.


Subject(s)
Placenta/metabolism , Pre-Eclampsia/metabolism , Proteoglycans/analysis , Proteoglycans/genetics , Adult , Blotting, Western , Decorin/analysis , Decorin/genetics , Female , Gene Expression , Glypicans/analysis , Glypicans/genetics , Heparan Sulfate Proteoglycans/analysis , Heparan Sulfate Proteoglycans/genetics , Humans , Placenta/chemistry , Pregnancy , RNA, Messenger/analysis , Syndecans/analysis , Syndecans/genetics
7.
Mol Vis ; 17: 2440-54, 2011.
Article in English | MEDLINE | ID: mdl-21976955

ABSTRACT

PURPOSE: The cornea is the major refractive component of the eye and serves as a barrier to the external environment. Understanding how the cornea responds to injury is important to developing therapies to treat vision disorders that affect the integrity and refractive properties of the cornea. Thus, investigation of the wound healing responses of the cornea to injury in a cost-effective animal model is a valuable tool for research. This study characterizes the wound healing responses in the corneas of White Leghorn chicken. METHODS: Linear corneal wounds were induced in post-natal day 7 (P7) chicks and cellular proliferation, apoptosis and regulation of structural proteins were assessed using immunohistochemical techniques. We describe the time course of increased expression of different scar-related markers, including vimentin, vinculin, perlecan and smooth muscle actin. RESULTS: We find evidence for acute necrotic cell death in the corneal region immediately surrounding cite of incision, whereas we failed to find evidence of delayed cell death or apoptosis. We find that the neuronal re-innervation of SV2-positive axon terminals within the corneal stroma and epithelium occurs very quickly after the initial scarring insult. We describe an accumulation of cells within the stroma immediately underlying the scar, which results, at least in part, from the local proliferation of keratocytes. Further, we provide evidence for scar-induced accumulations of CD45-positive monocytes in injured corneas. CONCLUSIONS: We conclude that the chick cornea is an excellent model system in which to study wound healing, formation of scar tissue, and neuronal re-innervation of sensory endings.


Subject(s)
Biomarkers/analysis , Cicatrix/metabolism , Cornea/metabolism , Cornea/pathology , Corneal Keratocytes/metabolism , Neurons/metabolism , Wound Healing/physiology , Actins/analysis , Actins/biosynthesis , Animals , Animals, Newborn , Bromodeoxyuridine/analysis , Cell Proliferation , Chickens , Cornea/innervation , Corneal Injuries , Corneal Keratocytes/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , Heparan Sulfate Proteoglycans/analysis , Heparan Sulfate Proteoglycans/biosynthesis , Immunohistochemistry , Leukocyte Common Antigens/analysis , Microscopy , Monocytes/cytology , Monocytes/metabolism , Necrosis , Neurons/cytology , Vimentin/analysis , Vimentin/biosynthesis , Vinculin/analysis , Vinculin/biosynthesis
8.
Cryobiology ; 63(3): 145-51, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21884690

ABSTRACT

Amniotic membrane (AM) has been used as a scaffold for the ex vivo expansion of different types of cells and a cell delivery matrix in regenerative medicine. Since the preservation procedures can influence the AM properties for experimental and clinical purposes, this study was established to investigate the feasibility of using the AM after different preservation methods to serve as substrates for endothelial cell expansion ex vivo. The effects of cryopreservation and lyophilization were evaluated on mechanical and histological characteristics of the AM, and the results were compared with the fresh AM. The ECM components of the basement membrane were well conserved in all groups. Although lyophilization resulted in more histological changes and lower level of physical variables including thickness, F(max), elongation at break and suture retention than the fresh and cryopreserved AM, endothelial cells grown on the lyophilized AM were better attached to the basement membrane. Cytotoxicity assay by MTT showed that the lyophilized AM is a compatible substrate for endothelial cells cultivation. The findings of this study suggest that the lyophilized AM is a suitable matrix for cultivation of endothelial cells due to this fact that lyophilization led to exposure of basement membrane of the AM.


Subject(s)
Amnion/anatomy & histology , Basement Membrane/anatomy & histology , Cryopreservation/methods , Endothelial Cells/cytology , Freeze Drying/methods , Tissue Engineering/methods , Tissue Scaffolds , Amnion/metabolism , Animals , Basement Membrane/metabolism , Cell Adhesion , Cell Proliferation , Cell Survival , Cesarean Section , Collagen/analysis , Collagen/biosynthesis , Endothelial Cells/physiology , Female , Fibronectins/analysis , Fibronectins/biosynthesis , Heparan Sulfate Proteoglycans/analysis , Heparan Sulfate Proteoglycans/biosynthesis , Humans , Laminin/analysis , Laminin/biosynthesis , Microscopy, Electron, Scanning , Placenta/anatomy & histology , Pregnancy , Rats , Rats, Sprague-Dawley
9.
Histochem Cell Biol ; 136(4): 437-54, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21874555

ABSTRACT

We have colocalized elastin and fibrillin-1 with perlecan in extracellular matrix of tensional and weight-bearing connective tissues. Elastin and fibrillin-1 were identified as prominent components of paraspinal blood vessels, and posterior longitudinal ligament in the human fetal spine and outer annulus fibrosus of the fetal intervertebral disc. We also colocalized perlecan with a synovial elastic basal lamina, where the attached synovial cells were observed to produce perlecan. Elastin, fibrillin-1 and perlecan were co-localized in the intima and media of small blood vessels in the synovium and in human fetal paraspinal blood vessels. Elastic fibers were observed at the insertion point of the anterior cruciate ligament to bone in the ovine stifle joint where they colocalized with perlecan. Elastin has not previously been reported to be spatially associated with perlecan in these tissues. Interactions between the tropoelastin and perlecan heparan sulfate chains were demonstrated using quartz crystal microbalance with dissipation solid phase binding studies. Electrostatic interactions through the heparan sulfate chains of perlecan and core protein mediated the interactions with tropoelastin, and were both important in the coacervation of tropoelastin and deposition of elastin onto perlecan immobilized on the chip surface. This may help us to understand the interactions which are expected to occur in vivo between the tropoelastin and perlecan to facilitate the deposition of elastin and formation of elastic microfibrils in situ and would be consistent with the observed distributions of these components in a number of connective tissues.


Subject(s)
Connective Tissue/chemistry , Elastin/ultrastructure , Heparan Sulfate Proteoglycans/analysis , Lumbar Vertebrae/chemistry , Animals , Elastin/analysis , Fibrillin-1 , Fibrillins , Humans , Immunohistochemistry , Lumbar Vertebrae/cytology , Lumbar Vertebrae/embryology , Microfilament Proteins/analysis , Microfilament Proteins/ultrastructure , Microscopy, Confocal , Sheep
10.
Am J Hum Genet ; 89(1): 15-27, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21763480

ABSTRACT

Proteoglycans are a major component of extracellular matrix and contribute to normal embryonic and postnatal development by ensuring tissue stability and signaling functions. We studied five patients with recessive joint dislocations and congenital heart defects, including bicuspid aortic valve (BAV) and aortic root dilatation. We identified linkage to chromosome 11 and detected a mutation (c.830G>A, p.Arg277Gln) in B3GAT3, the gene coding for glucuronosyltransferase-I (GlcAT-I). The enzyme catalyzes an initial step in the synthesis of glycosaminoglycan side chains of proteoglycans. Patients' cells as well as recombinant mutant protein showed reduced glucuronyltransferase activity. Patient fibroblasts demonstrated decreased levels of dermatan sulfate, chondroitin sulfate, and heparan sulfate proteoglycans, indicating that the defect in linker synthesis affected all three lines of O-glycanated proteoglycans. Further studies demonstrated that GlcAT-I resides in the cis and cis-medial Golgi apparatus and is expressed in the affected tissues, i.e., heart, aorta, and bone. The study shows that reduced GlcAT-I activity impairs skeletal as well as heart development and results in variable combinations of heart malformations, including mitral valve prolapse, ventricular septal defect, and bicuspid aortic valve. The described family constitutes a syndrome characterized by heart defects and joint dislocations resulting from altered initiation of proteoglycan synthesis (Larsen-like syndrome, B3GAT3 type).


Subject(s)
Glucuronosyltransferase/genetics , Heart Defects, Congenital/pathology , Proteoglycans/biosynthesis , Adolescent , Amino Acid Sequence , Aortic Valve/pathology , Case-Control Studies , Child , Chondroitin Sulfates/analysis , Chromosomes, Human, Pair 11/genetics , Consanguinity , Dermatan Sulfate/analysis , Electrophoresis, Polyacrylamide Gel , Fibroblasts/metabolism , Fluorescent Antibody Technique , Heparan Sulfate Proteoglycans/analysis , Humans , Immunoblotting , Male , Mitral Valve/pathology , Models, Molecular , Molecular Sequence Data , Pedigree
11.
J Oral Pathol Med ; 40(7): 552-9, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21198869

ABSTRACT

OBJECTIVES: The deposition of perlecan, a heparan sulfate proteoglycan, is enhanced within oral carcinoma in situ (CIS) foci, while it dynamically switches from CIS foci to the stromal space in squamous cell carcinoma (SCC). Because α-dystroglycan and integrin ß1 have been identified as two of the perlecan receptors, we wanted to determine their differential distributions before and after invasion of oral SCC. METHODS: Eighty-two surgical tissue specimens of oral SCC containing different precancerous stages were examined by immunohistochemistry for perlecan, α-dystroglycan, integrin ß1, and Ki-67. In addition, α-dystroglycan mRNA signals were localized by in situ hybridization. RESULTS: In normal epithelia, α-dystroglycan and integrin ß1 were localized on the cell membrane of basal cells, while perlecan was faintly present in the intercellular spaces of parabasal cells. In epithelial dysplasia and CIS, α-dystroglycan and perlecan were well co-localized in the epithelial layer, especially in its lower half, and this co-localization was mostly overlapped with Ki-67-positive (+) cell zones. However, in SCC, α-dystroglycan was localized neither within carcinoma cell nests nor in the stroma, while perlecan disappeared from SCC foci but emerged in the stromal space, leaving integrin ß1+ and Ki-67+ cells only to the periphery of SCC foci. α-Dystroglycan mRNA signals were basically identical to the α-dystroglycan protein localizations. CONCLUSION: The findings suggest that α-dystroglycan and integrin ß1 act as perlecan receptors in oral precancerous lesions prior to invasion, and that the perlecan signals via the two different receptors function in cellular differentiation and proliferation of CIS cells, respectively.


Subject(s)
Carcinoma, Squamous Cell/pathology , Dystroglycans/analysis , Heparan Sulfate Proteoglycans/analysis , Integrin beta1/analysis , Mouth Neoplasms/pathology , Carcinoma in Situ/pathology , Cell Membrane/pathology , Dystroglycans/genetics , Epithelial Cells/pathology , Epithelium/pathology , Extracellular Space/chemistry , Humans , In Situ Hybridization , Ki-67 Antigen/analysis , Neoplasm Invasiveness , Precancerous Conditions/pathology , RNA, Messenger/analysis , Stromal Cells/pathology
12.
Indian J Dent Res ; 22(6): 823-6, 2011.
Article in English | MEDLINE | ID: mdl-22484878

ABSTRACT

Perlecan means pearl-like structures. Perlecan is a large proteoglycan (400-500 kDa) present in virtually all vascularized tissues with a distribution that is primarily confined to basement membranes including those of oral mucosa. It is a basement membrane-type heparan sulfate proteoglycan. Perlecan is synthesized by basal cells and fibroblasts adjacent to the basal lamina . Perlecan is also synthesized by vascular endothelial and smooth muscle cells present in the extracellular matrix. It has been demonstrated in recent years that perlecan is distributed in the stromal space of various pathophysiological conditions. The complex pleiotropy of perlecan suggests that this gene product is involved in several developmental processes, at both early and late stages of embryogenesis, as well as in cancer and diabetes. In the oral cavity, perlecan expression is reported to basal cells in normal mucosa and its expression increases in precancer and cancerous conditions. It is also expressed in various odontogenic tumors such as ameloblastoma, keratocyst odontogenic tumor, and also salivary gland tumors such as adenoid cystic carcinoma, mucoepidermoid carcinoma, etc.


Subject(s)
Heparan Sulfate Proteoglycans/analysis , Mouth Neoplasms/pathology , Biomarkers, Tumor/analysis , Humans , Mouth Mucosa/pathology , Odontogenic Tumors/pathology , Precancerous Conditions/pathology , Salivary Gland Neoplasms/pathology
13.
Arch Oral Biol ; 55(10): 778-87, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20684947

ABSTRACT

OBJECTIVE: Immunohistochemical and gene expression profiles of heparanase were determined in murine molar tooth germs from their embryonic to postnatal stages, paying special attention to neovascularization within the enamel organ, which is poorly vascularized before birth. DESIGN: Protein and gene expression profiles of heparanase, heparan sulfate (HS), vascular endothelial growth factor (VEGF), transforming growth factor-beta1 (TGF-beta1), and perlecan were comparatively examined by immunohistochemistry and in-situ hybridization, respectively, in mouse mandibular molar tooth germs from embryonic day 11.5 to postnatal day 6. At the same time, their mRNA expression levels were also confirmed by reverse transcriptase-polymerase chain reaction using laser-captured microdissection of enamel organ tissues. RESULTS: Stellate reticulum cells highly expressed perlecan but only slightly expressed heparanase and HS in their embryonic days. On and after postnatal day 1, the expressions of heparanase became dramatically higher in the stellate reticulum, while HS disappeared leaving the immunopositivity for perlecan core protein. Immunohistochemically, HS was enhanced around blood vessels which were newly formed after birth within the enamel organs, whose volume was also regressive. Similar expression patterns were obtained for VEGF and TGF-beta1. CONCLUSIONS: Such synchronized expression modes among the HS metabolism-related molecules suggested that heparanase plays an important role in degradation of HS chains, which is closely related to vascular penetration into the stellate reticulum, which may be one of the driving forces for the postnatal regression of the enamel organ.


Subject(s)
Enamel Organ/blood supply , Enamel Organ/metabolism , Glucuronidase/biosynthesis , Heparan Sulfate Proteoglycans/biosynthesis , Animals , Enamel Organ/cytology , Enamel Organ/embryology , Gene Expression Profiling , Glucuronidase/analysis , Glucuronidase/genetics , Heparan Sulfate Proteoglycans/analysis , Heparan Sulfate Proteoglycans/genetics , In Situ Hybridization , Mice , Microdissection/methods , Neovascularization, Physiologic , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta1/biosynthesis , Transforming Growth Factor beta1/genetics , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics
14.
Braz. j. med. biol. res ; 43(7): 627-633, July 2010. ilus, graf
Article in English | LILACS | ID: lil-550734

ABSTRACT

The objective of the present study was to develop a quantitative method to evaluate laser-induced choroidal neovascularization (CNV) in a rat model using Heidelberg Retina Angiograph 2 (HRA2) imaging. The expression of two heparan sulfate proteoglycans (HSPG) related to inflammation and angiogenesis was also investigated. CNV lesions were induced with argon laser in 21 heterozygous Zucker rats and after three weeks a fluorescein angiogram and autofluorescence exams were performed using HRA2. The area and greatest linear dimension were measured by two observers not aware of the protocol. Bland-Altman plots showed agreement between the observers, suggesting that the technique was reproducible. After fluorescein angiogram, HSPG (perlecan and syndecan-4) were analyzed by real-time RT-PCR and immunohistochemistry. There was a significant increase in the expression of perlecan and syndecan-4 (P < 0.0001) in retinas bearing CNV lesions compared to control retinas. The expression of these two HSPG increased with increasing CNV area. Immunohistochemistry demonstrated that the rat retina damaged with laser shots presented increased expression of perlecan and syndecan-4. Moreover, we observed that the overexpression occurred in the outer layer of the retina, which is related to choroidal damage. It was possible to develop a standardized quantitative method to evaluate CNV in a rat model using HRA2. In addition, we presented data indicating that the expression of HSPG parallels the area of CNV lesion. The understanding of these events offers opportunities for studies of new therapeutic interventions targeting these HSPG.


Subject(s)
Animals , Female , Rats , Choroidal Neovascularization/metabolism , Heparan Sulfate Proteoglycans/metabolism , /analysis , Choroidal Neovascularization/etiology , Choroidal Neovascularization/pathology , Fluorescein Angiography/methods , Heparan Sulfate Proteoglycans/analysis , Immunohistochemistry , Laser Coagulation , Ophthalmoscopy/methods , Rats, Zucker , Reverse Transcriptase Polymerase Chain Reaction , /metabolism
15.
Braz J Med Biol Res ; 43(7): 627-33, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20464343

ABSTRACT

The objective of the present study was to develop a quantitative method to evaluate laser-induced choroidal neovascularization (CNV) in a rat model using Heidelberg Retina Angiograph 2 (HRA2) imaging. The expression of two heparan sulfate proteoglycans (HSPG) related to inflammation and angiogenesis was also investigated. CNV lesions were induced with argon laser in 21 heterozygous Zucker rats and after three weeks a fluorescein angiogram and autofluorescence exams were performed using HRA2. The area and greatest linear dimension were measured by two observers not aware of the protocol. Bland-Altman plots showed agreement between the observers, suggesting that the technique was reproducible. After fluorescein angiogram, HSPG (perlecan and syndecan-4) were analyzed by real-time RT-PCR and immunohistochemistry. There was a significant increase in the expression of perlecan and syndecan-4 (P < 0.0001) in retinas bearing CNV lesions compared to control retinas. The expression of these two HSPG increased with increasing CNV area. Immunohistochemistry demonstrated that the rat retina damaged with laser shots presented increased expression of perlecan and syndecan-4. Moreover, we observed that the overexpression occurred in the outer layer of the retina, which is related to choroidal damage. It was possible to develop a standardized quantitative method to evaluate CNV in a rat model using HRA2. In addition, we presented data indicating that the expression of HSPG parallels the area of CNV lesion. The understanding of these events offers opportunities for studies of new therapeutic interventions targeting these HSPG.


Subject(s)
Choroidal Neovascularization/metabolism , Heparan Sulfate Proteoglycans/metabolism , Syndecan-4/analysis , Animals , Choroidal Neovascularization/etiology , Choroidal Neovascularization/pathology , Female , Fluorescein Angiography/methods , Heparan Sulfate Proteoglycans/analysis , Immunohistochemistry , Laser Coagulation , Ophthalmoscopy/methods , Rats , Rats, Zucker , Reverse Transcriptase Polymerase Chain Reaction , Syndecan-4/metabolism
16.
Matrix Biol ; 29(6): 471-83, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20403434

ABSTRACT

Basement membranes (BMs) are physiologically insoluble extracellular matrix sheets present in all multicellular organisms. They play an important role in providing mechanical strength to tissues and regulating cell behavior. Proteomic analysis of BM proteins is challenged by their high molecular weights and extensive post-translational modifications. Here, we describe the direct analysis of an in vivo BM system using a mass spectrometry (MS) based proteomics approach. Retinal BMs were isolated from embryonic chick eyes. The BM macromolecules were deglycosylated and separated by low percentage gradient SDS PAGE, in-gel digested and analyzed by LC-MS/MS. This identified over 27 extracellular matrix proteins in the retinal BM. A semi-quantitative measure of protein abundance distinguished, nidogens-1 and -2, laminin subunits α1, α5, ß2, and γ1, agrin, collagen XVIII, perlecan, FRAS1 and FREM2 as the most abundant BM protein components. Laminin subunits α3, ß1, γ2, γ3 and collagen IV subunits α5 and α6 were minor constituents. To examine binding interactions that contribute to the stability of the retinal BM, we applied the LC-MS/MS based approach to detect potential BM complexes from the vitreous. Affinity-captured nidogen- and heparin-binding proteins from the vitreous contained >10 and >200 proteins respectively. Comparison of these protein lists with the retinal BM proteome reveals that glycosaminoglycan and nidogen binding interactions play a central role in the internal structure and formation of the retinal BM. In addition, we studied the biomechanical qualities of the retinal BM before and after deglycosylation using atomic force microscopy. These results show that the glycosaminoglycan side chains of the proteoglycans play a dominant role in regulating the thickness and elasticity of the BMs by binding water to the extracellular matrix. To our knowledge, this is the first large-scale investigation of an in vivo BM system using MS-based proteomics.


Subject(s)
Basement Membrane/chemistry , Basement Membrane/metabolism , Extracellular Matrix Proteins/analysis , Proteome/analysis , Proteomics , Retina/metabolism , Agrin/analysis , Agrin/genetics , Agrin/metabolism , Animals , Biomechanical Phenomena , Chick Embryo , Collagen Type IV/analysis , Collagen Type IV/genetics , Collagen Type IV/metabolism , Extracellular Matrix/chemistry , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Glycosaminoglycans/analysis , Glycosaminoglycans/genetics , Glycosaminoglycans/metabolism , Heparan Sulfate Proteoglycans/analysis , Heparan Sulfate Proteoglycans/genetics , Heparan Sulfate Proteoglycans/metabolism , Membrane Glycoproteins/analysis , Microscopy, Atomic Force , Protein Processing, Post-Translational , Proteoglycans/analysis , Proteoglycans/genetics , Proteoglycans/metabolism , Retina/chemistry , Retina/ultrastructure
17.
Cell Mol Life Sci ; 67(17): 2879-95, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20428923

ABSTRACT

More than three decades ago, basement membranes (BMs) were described as membrane-like structures capable of isolating a cell from and connecting a cell to its environment. Since this time, it has been revealed that BMs are specialized extracellular matrices (sECMs) with unique components that support important functions including differentiation, proliferation, migration, and chemotaxis of cells during development. The composition of these sECM is as unique as the tissues to which they are localized, opening the possibility that such matrices can fulfill distinct functions. Changes in BM composition play significant roles in facilitating the development of various diseases. Furthermore, tissues have to provide sECM for their stem cells during development and for their adult life. Here, we briefly review the latest research on these unique sECM and their components with a special emphasis on embryonic and adult stem cells and their niches.


Subject(s)
Basement Membrane/chemistry , Collagen Type IV/metabolism , Extracellular Matrix/chemistry , Heparan Sulfate Proteoglycans/metabolism , Laminin/metabolism , Membrane Glycoproteins/metabolism , Stem Cells/chemistry , Tissue Engineering/methods , Animals , Basement Membrane/physiology , Cell Culture Techniques , Collagen Type IV/analysis , Heparan Sulfate Proteoglycans/analysis , Humans , Laminin/analysis , Membrane Glycoproteins/analysis , Mice
18.
J Proteome Res ; 9(3): 1289-301, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20058866

ABSTRACT

In vitro fertilization (IVF) is fraught with problems and currently proteomics approaches are being tried out to examine the microenvironment of the follicle in order to assess biological and immunological parameters that may affect its development. Additionally, better understanding of reproductive process may help increase IVF birth rate per embryo transfer and at the same time avoid spontaneous miscarriages or life threatening conditions such as ovarian hyperstimulation syndrome. The primary aim of this study was to search for specific differences in protein composition of human follicular fluid (HFF) and plasma in order to identify proteins that accumulate or are absent in HFF. Depletion of abundant proteins combined with multidimensional protein fractionation allowed the study of middle- and lower-abundance proteins. Paired comparison study examining HFF with plasma/serum from women undergoing successful IVF revealed important differences in the protein composition which may improve our knowledge of the follicular microenvironment and its biological role. This study showed involvement of innate immune function of complement cascade in HFF. Complement inhibition and the presence of C-terminal fragment of perlecan suggested possible links to angiogenesis which is a vital process in folliculogenesis and placental development. Differences in proteins associated with blood coagulation were also found in the follicular milieu. Several specific proteins were observed, many of which have not yet been associated with follicle/oocyte maturation. These proteins together with their regulatory pathways may play a vital role in the reproductive process.


Subject(s)
Complement C3/metabolism , Complement C4/metabolism , Fertilization in Vitro , Follicular Fluid/metabolism , Proteome/metabolism , Chromatography, High Pressure Liquid , Cluster Analysis , Clusterin/analysis , Complement C3/analysis , Complement C4/analysis , Electrophoresis, Gel, Two-Dimensional , Female , Follicular Fluid/chemistry , Hemolysis , Heparan Sulfate Proteoglycans/analysis , Humans , Immunoblotting , Proteome/analysis , Reproducibility of Results , Signal Transduction
19.
Cardiovasc Diabetol ; 8: 46, 2009 Aug 20.
Article in English | MEDLINE | ID: mdl-19695080

ABSTRACT

BACKGROUND: Heparan sulfate proteoglycans (HSPGs) contain glycosaminoglycan (GAG) chains made primarily of heparan sulfate (HS). Hyperglycemia in diabetes leads to endothelial injury and nephropathy, retinopathy and atherosclerosis. Decreased HSPG may contribute to diabetic endothelial injury. Decreased tissue HS in diabetes has been reported, however, endothelial HS changes are poorly studied. OBJECTIVE: To determine total GAGs, including HS, in endothelium under hyperglycemic conditions and the protective effect of insulin and heparin. METHODS: Confluent primary porcine aortic endothelial cells (PAECs) were divided into control, glucose (30 mM), insulin (0.01 unit/ml) and glucose plus insulin treatment groups for 24, 48 and 72 hours. Additionally, PAECs were treated with glucose, heparin (0.5 microg/ml) and glucose plus heparin for 72 hours. GAGs were isolated from cells and medium. GAG concentrations were determined by the carbazole assay and agarose gel electrophoresis. RESULTS: GAGs were significantly increased only in control and glucose plus insulin groups at 72 versus 24 hours. Glucose decreased cell GAGs and increased medium GAGs, and insulin alone decreased cell GAGs at all times compared to control. In the glucose plus insulin group, cell GAGs were less than control at 24 hours, and greater than glucose or insulin alone at 48 and 72 hours while GAGs in medium were greater than control at all times and glucose at 72 hours. Heparin increased GAGs in glucose treated cells and medium. CONCLUSION: High glucose and insulin alone reduces endothelial GAGs. In hyperglycemic conditions, heparin or insulin preserves GAGs which may protect cells from injury. Insulin is an effective diabetic therapy since it not only lowers blood glucose, but also protects endothelium.


Subject(s)
Endothelial Cells/drug effects , Glucose/pharmacology , Glycosaminoglycans/metabolism , Heparin/pharmacology , Insulin/pharmacology , Animals , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Drug Interactions , Electrophoresis, Agar Gel , Endothelial Cells/metabolism , Glycosaminoglycans/analysis , Heparan Sulfate Proteoglycans/analysis , Heparan Sulfate Proteoglycans/metabolism , Heparitin Sulfate/analysis , Heparitin Sulfate/metabolism , Sus scrofa , Swine , Syndecans/analysis , Syndecans/metabolism , Time Factors
20.
Histochem Cell Biol ; 132(5): 491-503, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19669783

ABSTRACT

We evaluated the immunohistochemical distribution of three major proteoglycans of cartilage, i.e., aggrecan, versican and perlecan vis-a-vis collagens I and II in the developing human spine of first-trimester foetuses. Aggrecan and perlecan were prominently immunolocalised in the cartilaginous vertebral body rudiments and to a lesser extent within the foetal intervertebral disc. In contrast, versican was only expressed in the developing intervertebral disc interspace. Using domain-specific monoclonal antibodies against the various modules of versican, we discovered the V0 isoform as the predominant form present. Versican immunolocalisations conducted with antibodies directed to epitopes in its N and C termini and GAG-alpha and GAG-beta core protein domains provided evidence that versican in the nucleus pulposus was either synthesised devoid of a G3 domain or this domain was proteolytically removed in situ. The V0 versican isoform was localised with prominent fibrillar components in the annular lamellae of the outer annulus fibrosus. Perlecan was a notable pericellular proteoglycan in the annulus fibrosus and nucleus pulposus but poorly immunolocalised in the marginal tissues of the developing intervertebral disc, apparently delineating the intervertebral disc-vertebral body interface region destined to become the cartilaginous endplate in the mature intervertebral disc. The distribution of collagens I and II in the foetal spine was mutually exclusive with type I present in the outer annulus fibrosus, marginal tissues around the vertebral body rudiment and throughout the developing intervertebral disc, and type II prominent in the vertebral rudiment, absent in the outer annulus fibrosus and diffusely distributed in the inner annulus fibrosus and nucleus pulposus. Collectively, our findings suggest the existence of an intricate and finely balanced interplay between various proteoglycans and collagens and the spinal cell populations which synthesise and assemble these components during spinal development.


Subject(s)
Aggrecans/metabolism , Fetus/metabolism , Heparan Sulfate Proteoglycans/metabolism , Spine/embryology , Spine/metabolism , Versicans/metabolism , Aggrecans/analysis , Collagen Type I/chemistry , Collagen Type I/metabolism , Collagen Type II/chemistry , Collagen Type II/metabolism , Fetus/cytology , Heparan Sulfate Proteoglycans/analysis , Humans , Immunohistochemistry , Spine/cytology , Versicans/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...