Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Curr Med Chem ; 28(25): 5066-5083, 2021.
Article in English | MEDLINE | ID: mdl-34191695

ABSTRACT

The successive stages of breast cancer growth and dissemination depend on cell-autonomous factors and the communication between tumor cells and their surrounding cellular and extracellular matrix microenvironment. The cell surface heparan sulfate proteoglycan Syndecan-1 is dysregulated both in tumor cells and cells of the breast tumor stroma, indicating a potential role in the pathogenesis of this most frequent malignancy in women. Indeed, Syndecan-1 interacts with numerous ligands and receptors relevant to tumor progression, affecting processes as diverse as cancer stem cell function, cell proliferation, apoptosis, cell adhesion, migration and invasion, tumor angiogenesis, and leukocyte function in the tumor stroma. The present review summarizes the current understanding of breast carcinogenesis in correlation with their Syndecan-1 expression, involved mechanisms, and proposed therapeutic strategies against Syndecan-1-related malignancy.


Subject(s)
Breast Neoplasms , Syndecan-1 , Breast Neoplasms/drug therapy , Cell Proliferation , Female , Humans , Neovascularization, Pathologic , Syndecan-1/physiology , Tumor Microenvironment
2.
BMC Anesthesiol ; 21(1): 83, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33740916

ABSTRACT

BACKGROUND: The mechanisms of trauma induced coagulopathy (TIC) are considered multifactorial. Amongst others, however, shedding of the endothelial glycocalyx resulting in increased concentrations of glycocalyx fragments in plasma might also play a role. Thus, we hypothesized that shedded glycocalyx components affect coagulation and may act as humoral mediators of TIC. METHODS: To investigate effects of heparan sulfate, chondroitin sulfate, syndecan-1, versican, and thrombomodulin we added these fragments to in vitro assays of whole blood from healthy volunteers to yield concentrations observed in trauma patients. Platelet function, whole blood coagulation, and fibrinolysis were measured by standard coagulation tests, impedance aggregometry (IA), and viscoelastic tests (VET). To assess dose-response relationships, we performed IA with increasing concentrations of versican and VET with increasing concentrations of thrombomodulin. RESULTS: Intrinsically activated clotting times (i.e., activated partial thromboplastin time and intrinsically activated VET with and without heparinase) were unaffected by any glycocalyx fragment. Thrombomodulin, however, significantly and dose-dependently diminished fibrinolysis as assessed by VET with exogenously added rt-PA, and increased rt-PA-induced lysis Indices after 30 (up to 108% of control, p <  0,0001), 45 (up to 368% of control, p <  0,0001), and 60 min (up to 950% of control, p <  0,0001) in VET. Versican impaired platelet aggregation in response to arachidonic acid (up to - 37,6%, p <  0,0001), ADP (up to - 14,5%, p <  0,0001), and collagen (up to - 31,8%, p <  0,0001) in a dose-dependent manner, but did not affect TRAP-6 induced platelet aggregation. Clotting time in extrinsically activated VET was shortened by heparan sulfate (- 7,2%, p = 0,024), chondroitin sulfate (- 11,6%, p = 0,016), versican (- 13%, p = 0,012%), and when combined (- 7,2%, p = 0,007). CONCLUSIONS: Glycocalyx components exert distinct inhibitory effects on platelet function, coagulation, and fibrinolysis. These data do not support a 'heparin-like auto-anticoagulation' by shed glycosaminoglycans but suggest a possible role of versican in trauma-induced thrombocytopathy and of thrombomodulin in trauma-associated impairment of endogenous fibrinolysis.


Subject(s)
Fibrinolysis/physiology , Glycocalyx/physiology , Partial Thromboplastin Time , Platelet Aggregation/physiology , Adult , Chondroitin Sulfates/physiology , Female , Heparitin Sulfate/physiology , Humans , In Vitro Techniques , Male , Syndecan-1/physiology , Thrombomodulin/physiology , Versicans/physiology
3.
Hum Cell ; 33(3): 721-729, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32314115

ABSTRACT

This study is to analyze the potential contribution of Syndecan 1 (SDC1) to cisplatin resistance in hepatic carcinoma. Cell proliferation and viability were determined by direct counting and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, respectively. The protein levels of SDC1, p-AKT, AKT and ß-actin were quantified by western blotting. The SDC1 transcript abundance was measured by real-time polymerase chain reaction. The relative expression of SDC1 in clinical liver tumor samples was analyzed with immunohistochemistry. SDC1 was up-regulated in cisplatin-resistant HepG2 cells (denoted as HepG2 CR hereafter). SDC1-knockdown re-sensitized HepG2 CR cells to cisplatin treatment. Ectopic over-expression of SDC1 conferred drug resistance to naïve HepG2 cells. PI3K/AKT pathway was over-activated in HepG2 CR cells, and simultaneous administration with PI3K inhibitor greatly surmounted the resistance. We also demonstrated that SDC1 was aberrantly up-regulated in clinical hepatocellular carcinoma samples. Our study highlighted the importance of SDC1-PI3K/AKT signaling in the cisplatin resistance in hepatocellular carcinoma.


Subject(s)
Cisplatin/pharmacology , Drug Resistance, Neoplasm/genetics , Liver Neoplasms/pathology , Signal Transduction/genetics , Signal Transduction/physiology , Syndecan-1/physiology , Cell Proliferation/genetics , Cell Survival/genetics , Gene Expression , Hep G2 Cells , Humans , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Syndecan-1/genetics , Syndecan-1/metabolism
4.
Shock ; 53(5): 575-584, 2020 05.
Article in English | MEDLINE | ID: mdl-31090680

ABSTRACT

Clinical data has supported the early use of plasma in high ratios of plasma to red cells to patients in hemorrhagic shock. The benefit from plasma seems to extend beyond its hemostatic effects to include protection to the post-shock dysfunctional endothelium. Resuscitation of the endothelium by plasma and one of its major constituents, fibrinogen, involves cell surface stabilization of syndecan-1, a transmembrane proteoglycan and the protein backbone of the endothelial glycocalyx. The pathogenic role of miRNA-19b to the endothelium is explored along with the PAK-1-mediated intracellular pathway that may link syndecan-1 to cytoskeletal protection. Additionally, clinical studies using fibrinogen and cyroprecipitate to aid in hemostasis of the bleeding patient are reviewed and new data to suggest a role for plasma and its byproducts to treat the dysfunctional endothelium associated with nonbleeding diseases is presented.


Subject(s)
Endothelium/physiopathology , Resuscitation , Shock, Hemorrhagic/physiopathology , Shock, Hemorrhagic/therapy , Wounds and Injuries/complications , Glycocalyx/physiology , Humans , Shock, Hemorrhagic/etiology , Syndecan-1/physiology , Wounds and Injuries/physiopathology
5.
Invest Ophthalmol Vis Sci ; 58(12): 4959-4975, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28973369

ABSTRACT

Purpose: To determine the impact of the loss of syndecan 1 (SDC1) on intraepithelial corneal nerves (ICNs) during homeostasis, aging, and in response to 1.5-mm trephine and debridement injury. Methods: Whole-mount corneas are used to quantify ICN density and thickness over time after birth and in response to injury in SDC1-null and wild-type (WT) mice. High-resolution three-dimensional imaging is used to visualize intraepithelial nerve terminals (INTs), axon fragments, and lysosomes in corneal epithelial cells using antibodies against growth associated protein 43 (GAP43), ßIII tubulin, and LAMP1. Quantitative PCR was performed to quantify expression of SDC1, SDC2, SDC3, and SDC4 in corneal epithelial mRNA. Phagocytosis was assessed by quantifying internalization of fluorescently labeled 1-µm latex beads. Results: Intraepithelial corneal nerves innervate the corneas of SDC1-null mice more slowly. At 8 weeks, ICN density is less but thickness is greater. Apically projecting intraepithelial nerve terminals and lysosome-associated membrane glycoprotein 1 (LAMP1) are also reduced in unwounded SDC1-null corneas. Quantitative PCR and immunofluorescence studies show that SDC3 expression and localization are increased in SDC1-null ICNs. Wild-type and SDC1-null corneas lose ICN density and thickness as they age. Recovery of axon density and thickness after trephine but not debridement wounds is slower in SDC1-null corneas compared with WT. Experiments assessing phagocytosis show reduced bead internalization by SDC1-null epithelial cells. Conclusions: Syndecan-1 deficiency alters ICN morphology and homeostasis during aging, reduces epithelial phagocytosis, and impairs reinnervation after trephine but not debridement injury. These data provide insight into the mechanisms used by sensory nerves to reinnervate after injury.


Subject(s)
Aging/physiology , Cornea/innervation , Corneal Injuries/pathology , Homeostasis/physiology , Nerve Fibers/pathology , Syndecan-1/physiology , Animals , Axons , Corneal Injuries/metabolism , Disease Models, Animal , Epithelium, Corneal/metabolism , Mice , Mice, Inbred BALB C , Syndecan-1/deficiency , Syndecans/metabolism
6.
Tumour Biol ; 37(9): 11573-11588, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27317256

ABSTRACT

Syndecans (SDC, SYND) comprise a group of four structurally related type 1 transmembrane heparan sulfate proteoglycans (HSPGs) that play important roles in tumorigenic processes. SDCs exert signaling via their protein cores and their conserved transmembrane and cytoplasmic domains or by forming complexes with growth factors (GFs). In classical Hodgkin's lymphoma (cHL), a lymphoid neoplasm of predominantly B cell origin, SDC1 and SDC4 are the active SDCs, and a number of GF (vascular endothelial growth factor, fibroblast growth factor, etc.) signaling pathways have been studied. However, despite extensive pre-clinical and clinical research on SDC-mediated GF signaling in many cancer types, there is very limited data for this interaction in cHL. Thus, this review highlights the relevant literature focusing on the potential interactions of SDCs and GFs in cHL pathogenesis. Also discussed are the pre-clinical and clinical studies targeting signaling through these pathways.


Subject(s)
Hodgkin Disease/etiology , Signal Transduction/physiology , Syndecan-1/physiology , Animals , Humans , Lysophospholipids/physiology , Neovascularization, Physiologic , Receptor, IGF Type 1/physiology , Receptors, Platelet-Derived Growth Factor/physiology , Receptors, Vascular Endothelial Growth Factor/physiology , Sphingosine/analogs & derivatives , Sphingosine/physiology , Syndecan-1/analysis , Tumor Necrosis Factor Ligand Superfamily Member 13/physiology , Vascular Endothelial Growth Factor A/physiology
7.
Arch Oral Biol ; 66: 155-64, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26954096

ABSTRACT

AIMS: Pathogenic mechanisms involved in early submerged implant failure are poorly understood. In this study we immunohistochemically analyse differences in proliferation, apoptosis and inflammation in edentulous ridge oral mucosa (ERM) of successful and early failed submerged implants. MATERIALS AND METHODS: 30 samples of ERM covering successful and early failed submerged implants were obtained at the end of osseointegration period along with control samples of healthy ERM. Sections were stained with Ki-67 (proliferation), caspase-3 (apoptosis) and syndecan-1 (epithelial marker). Percentage of positive cells was analysed by Kruskal-Wallis test and Dunn's post hoc test. Co-localization of Ki-67 and caspase-3 with α-SMA, CD68 and TGF-ß was done by double immunofluorescence. RESULTS: There was no significant difference in number of Ki-67 positive cells within surface epithelium (SE) in all groups. Proliferation was significantly higher in underlying connective tissue (UCT) of ERM of early failed submerged implants (26%) compared to ERM of successful submerged implants (3%) and controls (4%). More apoptotic cells appeared in UCT of early failed submerged implants (8%) compared to UCT of successful submerged implants (4%) and UCT of control ERM (3%). Co-localization of Ki-67 and α-SMA in ERM of early failed submerged implants disclosed proliferating fibroblasts and pericytes of blood vessels. Macrophages and cells expressing TGF-ß appeared in UCT of failed implants. Expression of syndecan-1 was significantly weaker in SE of early failed submerged implants. CONCLUSIONS: Imbalance between proliferation and apoptosis, changes in syndecan-1 expression and inflammation are histopathological features of ERM of early failed submerged implants.


Subject(s)
Actins/biosynthesis , Dental Implants/adverse effects , Dental Restoration Failure , Mouth Mucosa/metabolism , Mouth, Edentulous/metabolism , Syndecan-1/biosynthesis , Actins/physiology , Aged , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Apoptosis/physiology , Caspase 3/metabolism , Cell Proliferation/physiology , Dental Implantation, Endosseous , Female , Humans , Macrophages/metabolism , Male , Middle Aged , Mouth Mucosa/blood supply , Mouth Mucosa/pathology , Mouth, Edentulous/pathology , Osseointegration , Syndecan-1/physiology , Transforming Growth Factor beta/analysis , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/physiology
8.
Biochim Biophys Acta ; 1863(4): 717-26, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26852939

ABSTRACT

Surface expressed proteoglycans mediate the binding of cytokines and chemokines to the cell surface and promote migration of various tumor cell types including epithelial tumor cells. We here demonstrate that binding of the chemokine-like inflammatory cytokine macrophage migration inhibitory factor (MIF) to epithelial lung and breast tumor cell lines A549 and MDA-MB231 is sensitive to enzymatic digestion of heparan sulphate chains and competitive inhibition with heparin. Moreover, MIF interaction with heparin was confirmed by chromatography and a structural comparison indicated a possible heparin binding site. These results suggested that proteoglycans carrying heparan sulphate chains are involved in MIF binding. Using shRNA-mediated gene silencing, we identified syndecan-1 as the predominant proteoglycan required for the interaction with MIF. MIF binding was decreased by induction of proteolytic shedding of syndecan-1, which could be prevented by inhibition of the metalloproteinases involved in this process. Finally, MIF induced the chemotactic migration of A549 cells, wound closure and invasion into matrigel without affecting cell proliferation. These MIF-induced responses were abrogated by heparin or by silencing of syndecan-1. Thus, our study indicates that syndecan-1 on epithelial tumor cells promotes MIF binding and MIF-mediated cell migration. This may represent a relevant mechanism through which MIF enhances tumor cell motility and metastasis.


Subject(s)
Epithelial Cells/metabolism , Intramolecular Oxidoreductases/metabolism , Intramolecular Oxidoreductases/physiology , Macrophage Migration-Inhibitory Factors/metabolism , Macrophage Migration-Inhibitory Factors/physiology , Neoplasms/metabolism , Syndecan-1/physiology , Cell Adhesion , Cell Membrane/metabolism , Cell Movement , Epithelial Cells/pathology , HEK293 Cells , Heparitin Sulfate/metabolism , Humans , Neoplasm Metastasis , Neoplasms/pathology , Protein Binding , Syndecan-1/metabolism , Tumor Cells, Cultured
9.
Eur J Gynaecol Oncol ; 36(5): 506-13, 2015.
Article in English | MEDLINE | ID: mdl-26513873

ABSTRACT

PURPOSE: Syndecan-1 (SDC-1) promotes the proliferation of cancer cells and plays a role in angiogenesis by binding to a variety of extracellular effectors. The present study was designed to compare the expression of SDC-1 in the normal ovary and in ovarian tumors, to better understand its roles in the progression of epithelial ovarian carcinoma (EOC). MATERIALS AND METHODS: The expression of SDC- 1, fibroblast growth factor 2 (FGF-2), and FGF receptor 1 (FGFRI) and their transcripts in 65 samples including the normal ovary, benign tumors, borderline ovarian tumors, and EOC was assessed using immunohistochemistry and the reverse transcription-polymerase chain reaction. The influence of FGF-2 on the expression of SDC-1 mRNA syndecan-1 in a human ovarian carcinoma cell line was determined using an FGF-2-neutralizing antibody. RESULTS: SDC-l was not detected in normal ovarian tissue but was present in the epithelial cells of benign or borderline tumors and in ovarian adenocarcinomas. The levels of expression were significantly different in ovarian tissues derived from benign or malignant cases. Coordinate stromal expression of SDC-1 and its mRNA was detected at the original site of the tumor, as well as in metastatic foci in the greater omentum of ovarian adenocarcinomas. FGF-2 reduced the level of expression of SDC-1 mRNA when added exogenously to SKOV3 cells. This effect was abolished in the presence of an FGF-2-neutralizing antibody. CONCLUSION: SDC-l contributes to the role of FGF-2 in proliferation and angiogenesis but may also play a role in the invasive properties of EOC. To the present authors' knowledge, this study is the first to report the presence of distinct patterns ofexpression of SDC-1 in local and metastatic foci in the greater omentum in patients with EOC. These data reinforce the role of the tumor stroma in the invasive properties of ovarian adenocarcinoma and suggest that stromal changes in the expression of SDC-1 may originate from the stroma and contribute to the pathogenesis and metastatic potential of EOC.


Subject(s)
Biomarkers, Tumor/analysis , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Syndecan-1/analysis , Adult , Biomarkers, Tumor/genetics , Biomarkers, Tumor/physiology , Carcinoma, Ovarian Epithelial , Disease Progression , Female , Fibroblast Growth Factor 2/analysis , Humans , Middle Aged , Neoplasm Invasiveness , Neoplasms, Glandular and Epithelial/chemistry , Ovarian Neoplasms/chemistry , Ovary/chemistry , RNA, Messenger/analysis , Receptor, Fibroblast Growth Factor, Type 1/analysis , Syndecan-1/genetics , Syndecan-1/physiology
10.
Shock ; 44(5): 452-7, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26263434

ABSTRACT

We have shown in a rodent model of hemorrhagic shock (HS) that fresh frozen plasma (FFP) reduces lung inflammation and injury that are correlated with restitution of syndecan-1. As the gut is believed to contribute to distant organ injury and inflammation after shock, the current study sought to determine if the protective effects of plasma would extend to the gut and to elucidate the contribution of syndecan-1 to this protective effect. We also examined the potential role of TNFα, and a disintegrin and metalloproteinase (ADAM)-17, both intestinal sheddases of syndecan-1. Wild-type (WT) and syndecan-1 (KO) mice were subjected to HS followed by resuscitation with lactated Ringer's (LR) or FFP and compared with shock alone and shams. Small bowel and blood were obtained after 3  h for analysis of mucosal injury and inflammation and TNFα and ADAM-17 protein expression and activity. After HS, gut injury and inflammation were significantly increased compared with shams. Resuscitation with LR decreased both injury and inflammation that were further lessened by FFP. KO mice displayed worsened gut injury and inflammation after HS compared with WT mice, and LR and FFP equivalently inhibited injury and inflammation. Both systemic and intestinal TNFα and ADAM-17 followed similar trends, with increases after HS, reduction by LR, and a further decrease by FFP in WT but not KO mice. In conclusion, FFP decreased gut injury and inflammation after hemorrhagic shock, an effect that was abrogated in syndecan-1 mice. Plasma also decreased TNFα and ADAM-17, representing a potential mechanistic link to its protection via syndecan-1.


Subject(s)
Intestinal Diseases/prevention & control , Plasma , Shock, Hemorrhagic/therapy , Syndecan-1/physiology , ADAM Proteins/metabolism , ADAM17 Protein , Animals , Disease Models, Animal , Enteritis/etiology , Enteritis/metabolism , Enteritis/pathology , Enteritis/prevention & control , Intestinal Diseases/etiology , Intestinal Diseases/metabolism , Intestinal Diseases/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestine, Small/metabolism , Intestine, Small/pathology , Male , Mice, Knockout , Shock, Hemorrhagic/complications , Shock, Hemorrhagic/metabolism , Syndecan-1/deficiency , Syndecan-1/genetics , Tumor Necrosis Factor-alpha/metabolism
11.
Cell Signal ; 27(10): 2054-67, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26210886

ABSTRACT

Syndecan-1 is a proteoglycan that acts as co-receptor through its heparan sulfate (HS) chains and plays important roles in cancer. HS chains are highly variable in length and sulfation pattern. This variability is enhanced by the SULF1/2 enzymes, which remove 6-O-sulfates from HS. We used malignant mesothelioma, an aggressive tumor with poor prognosis, as a model and demonstrated that syndecan-1 over-expression down-regulates SULF1 and alters the HS biosynthetic machinery. Biochemical characterization revealed a 2.7-fold reduction in HS content upon syndecan-1 over-expression, but an overall increase in sulfation. Consistent with low SULF1 levels, trisulfated disaccharides increased 2.5-fold. ERK1/2 activity was enhanced 6-fold. Counteracting ERK activation, Akt, WNK1, and c-Jun were inhibited. The net effect of these changes manifested in G1 cell cycle arrest. Studies of pleural effusions showed that SULF1 levels are lower in pleural malignancies compared to benign conditions and inversely correlate with the amounts of syndecan-1, suggesting important roles for syndecan-1 and SULF1 in malignant mesothelioma.


Subject(s)
Heparitin Sulfate/metabolism , Lung Neoplasms/metabolism , Mesothelioma/metabolism , Signal Transduction , Syndecan-1/physiology , Biosynthetic Pathways , Cell Cycle , Cell Line, Tumor , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Lung Neoplasms/mortality , Mesothelioma/mortality , Mesothelioma, Malignant , Pleural Effusion, Malignant/metabolism , Proportional Hazards Models , Sulfotransferases/metabolism
12.
Br J Cancer ; 113(2): 321-6, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26057450

ABSTRACT

BACKGROUND: Tongue squamous cell carcinoma (TSCC) is increasing in incidence, especially among young patients and preferably females. Infection with human papilloma virus (HPV) has been suggested as a cause of SCC in the head and neck, and the proportion of oropharyngeal cancers caused by HPV has steadily increased. METHODS: Samples from 109 patients with primary TSCC were analysed for the presence of HPV16 by in situ hybridisation and for expression of its surrogate marker p16 and the HPV receptor syndecan-1 by immunhistochemistry. RESULTS: No evidence of HPV16 DNA was observed in the tumours, although one-third showed p16 staining. There was no difference in the expression of the primary HPV receptor, syndecan-1, between TSCC and a group of tonsil SCC. CONCLUSION: Whereas p16 is expressed in some TSCCs, HPV16 is undetectable, therefore, p16 cannot be used as a surrogate marker for high-risk HPV-infection in this tumour. Despite presence of the HPV-receptor syndecan-1 in TSCC, HPV prefers the tonsillar environment. Lack of p16 associates with worse prognosis primarily in patients aged ⩽40 years with tongue SCC. The improved prognosis seen in p16-positive TSCC can be due to induction of a senescent phenotype or an inherent radiosensitivity due to the ability of p16 to inhibit homologous recombination repair.


Subject(s)
Carcinoma, Squamous Cell/virology , Head and Neck Neoplasms/virology , Human papillomavirus 16/isolation & purification , Neoplasm Proteins/physiology , Papillomavirus Infections/complications , Receptors, Virus/physiology , Syndecan-1/physiology , Tongue Neoplasms/virology , Adult , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/mortality , Cyclin-Dependent Kinase Inhibitor p16 , DNA, Viral/analysis , Female , Head and Neck Neoplasms/mortality , Humans , In Situ Hybridization , Male , Middle Aged , Neoplasm Proteins/analysis , Squamous Cell Carcinoma of Head and Neck , Syndecan-1/analysis , Tongue Neoplasms/mortality
13.
Biochem Biophys Res Commun ; 456(1): 183-9, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25462564

ABSTRACT

MicroRNAs (miRNAs) are short (19-24 nt), low molecular weight RNAs that play important roles in the regulation of target genes associated with cell proliferation, differentiation, and development, by binding to the 3'-untranslated region of the target mRNAs. In this study, we examined the expression of miRNA-126 (miR-126) and miR-149 in prostate cancer, and investigated the molecular mechanisms by which they affect syndecan-1 in prostate cancer. Functional analysis of miR-126 and miR-149 was conducted in the prostate cancer cell lines, PC3, Du145, and LNCaP. The expression levels of SOX2, NANOG, Oct4, miR-126 and miR-149 were evaluated by quantitative RT-PCR. After silencing syndecan-1, miR-126, and/or miR-149 in the PC3 cells, cell proliferation, senescence, and p21 induction were assessed using the MTS assay, senescence-associated ß-galactosidase (SA-ß-Gal) assay, and immunocytochemistry, respectively. Compared to the Du145 and LNCaP cells, PC3 cells exhibited higher expression of syndecan-1. When syndecan-1 was silenced, the PC3 cells showed reduced expression of miR-126 and miR-149 most effectively. Suppression of miR-126 and/or miR-149 significantly inhibited cell growth via p21 induction and subsequently, induced senescence. The mRNA expression levels of SOX2, NANOG, and Oct4 were significantly increased in response to the silencing of miR-126 and/or miR-149. Our results suggest that miR-126 and miR-149 are associated with the expression of syndecan-1 in prostate cancer cells. These miRNAs promote cell proliferation by suppressing SOX2, NANOG, and Oct4. The regulation of these factors by miR-126 and miR-149 is essential for syndecan-1-mediated development of androgen-refractory prostate cancer.


Subject(s)
Cell Proliferation , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Prostatic Neoplasms/metabolism , Syndecan-1/physiology , Cell Line, Tumor , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , Male , Nanog Homeobox Protein , Octamer Transcription Factor-3/metabolism , RNA, Messenger/metabolism , SOXB1 Transcription Factors/metabolism , Transfection , beta-Galactosidase/metabolism
14.
PLoS One ; 9(3): e92378, 2014.
Article in English | MEDLINE | ID: mdl-24658332

ABSTRACT

Despite recent advances in the treatment of multiple myeloma (MM), it remains an incurable disease potentially due to the presence of resistant myeloma cancer stem cells (MM-CSC). Although the presence of clonogenic cells in MM was described three decades ago, the phenotype of MM-CSC is still controversial, especially with respect to the expression of syndecan-1 (CD138). Here, we demonstrate the presence of two subpopulations--CD138++ (95-99%) and CD138low (1-5%)--in eight MM cell lines. To find out possible stem-cell-like features, we have phenotypically, genomic and functionally characterized the two subpopulations. Our results show that the minor CD138low subpopulation is morphologically identical to the CD138++ fraction and does not represent a more immature B-cell compartment (with lack of CD19, CD20 and CD27 expression). Moreover, both subpopulations have similar gene expression and genomic profiles. Importantly, both CD138++ and CD138low subpopulations have similar sensitivity to bortezomib, melphalan and doxorubicin. Finally, serial engraftment in CB17-SCID mice shows that CD138++ as well as CD138low cells have self-renewal potential and they are phenotypically interconvertible. Overall, our results differ from previously published data in MM cell lines which attribute a B-cell phenotype to MM-CSC. Future characterization of clonal plasma cell subpopulations in MM patients' samples will guarantee the discovery of more reliable markers able to discriminate true clonogenic myeloma cells.


Subject(s)
Multiple Myeloma/physiopathology , Syndecan-1/genetics , Syndecan-1/physiology , Animals , Boronic Acids/pharmacology , Bortezomib , Cell Line, Tumor , DNA Copy Number Variations , Doxorubicin/pharmacology , Heterografts , Humans , Immunophenotyping , Melphalan/pharmacology , Mice, SCID , Multiple Myeloma/immunology , Neoplastic Stem Cells , Phenotype , Plasma Cells/immunology , Precursor Cells, B-Lymphoid , Pyrazines/pharmacology , Syndecan-1/biosynthesis
15.
Int J Clin Oncol ; 19(2): 247-53, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23553620

ABSTRACT

BACKGROUND: Syndecan-1 (SDC1) is reported to modulate several key processes of tumorigenesis and to show variable expression in many cancers. The cause of these variations in expression is not known to date. In this study, we compared SDC1 status with clinicopathologic parameters to evaluate the prognostic implications of SDC1 status on squamous cell carcinoma (SCC) of the tonsil. METHODS: In 56 cases of tonsillar SCC, we screened SDC1 expression using immunohistochemistry and analyzed the relationships between SDC1 expression and clinicopathological parameters. To identify the cause of the changes in SDC1 expression seen in tumors, we measured the gene dosage of SDC1 in tumor cells using fluorescent in situ hybridization. RESULTS: SDC1 expression was found in cancer cells in 36 cases (64.3 %) of tonsillar SCC. It was associated with lymph node metastasis (p = 0.010) and a positive surgical resection margin (p = 0.014). On the other hand, it was not significantly correlated with sex, age, smoking status, degree of differentiation, T stage, or distant metastasis. We could not find any copy-number variation of SDC1 in the cases showing increased SDC1 immunopositivity. In addition, strong SDC1 expression in the tumor cells predicted a shorter overall survival (p = 0.020, log-rank). CONCLUSIONS: We showed that SDC1 expression is associated with N stage and the status of resection margin involvement in SCC of the tonsil. With respect to survival, there were unfavorable outcomes in cases with SDC1 positivity. More studies are needed to better understand the role of SDC1 in the progression and invasiveness of tonsillar SCC.


Subject(s)
Carcinoma, Squamous Cell/mortality , Syndecan-1/physiology , Tonsillar Neoplasms/mortality , Adult , Aged , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/virology , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Neoplasm Staging , Papillomaviridae/isolation & purification , Prognosis , Syndecan-1/analysis , Syndecan-1/genetics , Tonsillar Neoplasms/pathology , Tonsillar Neoplasms/virology
16.
Res Vet Sci ; 95(3): 1021-5, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24128845

ABSTRACT

CD138 (syndecan-1) interacts with various components of the extracellular matrix and associates with the actin cytoskeleton. In this study, we cloned pig CD138 cDNA and determined its complete cDNA sequence. Pig CD138 cDNA contained an open reading frame (930 bp) encoding 309 amino acids with five well conserved putative glycosaminoglycan attachment sites, a putative cleavage site for matrix metalloproteinases, and conserved motifs involved in signal transduction among mammalian species. Pig CD138 mRNA was detected in various tissues, including lymphoid and non-lymphoid organs, indicating the multicellular functions of CD138 in pigs. Western blot and flow cytometry analyses detected an approximate 35 kDa pig CD138 protein expressed on the cell surface. Further immunohistochemistry analysis revealed that CD138 expression was mainly observed in submucosa and lamina propria of the pig small intestine. Further study will be necessary to define the functional importance of CD138 during specific infectious diseases in pigs.


Subject(s)
Syndecan-1/genetics , Amino Acid Sequence , Animals , Base Sequence , Blotting, Western/veterinary , Cloning, Molecular , Flow Cytometry/veterinary , Molecular Sequence Data , Phylogeny , Sequence Alignment/veterinary , Swine/genetics , Syndecan-1/physiology , Tissue Distribution
17.
J Pathol ; 231(4): 495-504, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24549646

ABSTRACT

Increasing evidence suggests that tumour-initiating cells (TICs) contribute to the development of prostate cancer. Here, we identified syndecan-1 as a key molecule maintaining the stability of prostate cancer TICs. Holoclones harbouring the biological properties of stemness were derived from single-cell cultures of the PC3 human prostate cancer cell line. These holoclones over-expressed syndecan-1, but showed reduced expression of NADPH oxidase (NOX) and synthesis of hydrogen peroxide and oxygen radicals. Stable RNA-mediated silencing of syndecan-1 gene expression up-regulated NOX-dependent generation of reactive oxygen species and reduced the survival of holoclones in vitro. Syndecan-1 down-regulation also strongly reduced the number of CD133(+)/CD44(+) primitive cancer cells and tumour growth in vivo. Interestingly, syndecan-1 gene knockdown significantly enhanced the tumour-suppressive effects of docetaxel by inhibiting the docetaxel-induced increase in CD133(+)/CD44(+) cells in vivo. In the transgenic adenocarcinoma of the mouse prostate (TRAMP) mouse model of prostate cancer, early intervention with a syndecan-1 inhibitor (OGT2115) or syndecan-1 RNAi reduced the incidence of adenocarcinoma and the number of c-kit(+)/CD44(+) cells in cancer foci. Finally, we found that syndecan-1 immunopositivity in prostate cancer cells was significantly associated with biochemical recurrence after radical prostatectomy. Taken together, our results show that syndecan-1 contributes to prostatic carcinogenesis by maintaining TICs and may be a target molecule for therapy.


Subject(s)
Neoplasm Proteins/physiology , Neoplastic Stem Cells/pathology , Prostatic Neoplasms/pathology , Syndecan-1/physiology , Aged , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Combined Modality Therapy , Disease Progression , Docetaxel , Gene Knockdown Techniques , Gene Silencing , Genetic Therapy/methods , Heterografts , Humans , Male , Mice , Mice, SCID , Mice, Transgenic , NADPH Oxidases/metabolism , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Transplantation , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Prostatectomy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/therapy , Reactive Oxygen Species/metabolism , Recurrence , Syndecan-1/biosynthesis , Syndecan-1/genetics , Taxoids/pharmacology , Taxoids/therapeutic use , Tumor Cells, Cultured
18.
J Trauma Acute Care Surg ; 73(1): 60-6, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22743373

ABSTRACT

BACKGROUND: There is emerging evidence that early trauma-induced coagulopathy (TIC) is mechanistically linked to disruption of the vascular endothelium and its glycocalyx, assessed by thrombomodulin and syndecan 1, respectively. This study evaluated if degradation of the endothelial glycocalyx and ensuing release of its heparin-like substances induce autoheparinization and thereby contributes to TIC. METHODS: Prospective observational study of 77 trauma patients admitted to a Level I trauma center having blood sampled at admission. Data on demography, hematology, Injury Severity Score, transfusion requirements, 30-day mortality, and thrombelastography (TEG, concurrent kaolin-activated/kaolin-heparinase-activated) were recorded. Retrospective analysis of plasma/serum for biomarkers reflecting endothelial glycocalyx and cell damage (syndecan 1, thrombomodulin), tissue injury (histone-complexed DNA fragments), sympathoadrenal activation (adrenaline, noradrenaline), coagulation activation/anticoagulation (prothrombin fragment 1+2, fibrinogen, von Willebrand factor, factor XIII, antithrombin, protein C, activated protein C, tissue factor pathway inhibitor), fibrinolysis (tissue-type plasminogen activator, plasminogen activator inhibitor 1) and inflammation (interleukin 6, terminal complement complex). Stratification of patients was according to the degree of TEG-measured heparinization. RESULTS: Four patients (5.2%) displayed evidence of high-degree autoheparinization, and these patients had higher Injury Severity Score (median [interquartile range], 31 [26-37] vs. 17 [10-26]), increased glucose (median, 13.6 vs. 8.0 mmol/L), and lower hemoglobin level (median, 5.8 vs. 8.4 mmol/L) and received more transfusions during the first 1 hour (median, 5 vs. 0) and 24 hours (median, 10 vs. 0) (all p < 0.05). Importantly, patients with autoheparinization had fourfold higher syndecan 1 levels (median [interquartile range], 116 ng/mL [78-140 ng/mL] vs. 31 ng/mL [18-49 ng/mL]), and they had higher international normalized ratio (median, 1.4 vs. 1.1), thrombomodulin (median, 4.1 vs. 1.7 ng/mL) and interleukin 6 (median, 129 vs. 71 pg/mL) but lower protein C (85% vs. 109%) (all p < 0.05), indicating profound endothelial damage, coagulopathy and inflammation. CONCLUSION: Five percent of the patients with trauma in the present study had evidence of acute endogenous coagulopathy with autoheparinization by TEG, which appeared mechanistically linked to endothelial glycocalyx degradation. Acute endogenous autoheparinization may contribute to TIC. LEVEL OF EVIDENCE: Prognostic study, level III.


Subject(s)
Blood Coagulation Disorders/etiology , Endothelium, Vascular/physiopathology , Glycocalyx/physiology , Heparin/physiology , Wounds and Injuries/complications , Adult , Blood Coagulation Disorders/blood , Blood Coagulation Disorders/physiopathology , Blood Transfusion , Female , Humans , Injury Severity Score , Male , Middle Aged , Prospective Studies , Syndecan-1/blood , Syndecan-1/physiology , Thrombelastography , Thrombomodulin/blood , Thrombomodulin/physiology , Wounds and Injuries/blood , Wounds and Injuries/physiopathology
19.
Shock ; 38(1): 57-62, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22706022

ABSTRACT

Syndecan 1 is the predominant heparan sulfate proteoglycan found on the surface of epithelial cells and, like glutamine, is essential in maintaining the intestinal epithelial barrier. We therefore hypothesized that loss of epithelial syndecan 1 would abrogate the gut-protective effects of enteral glutamine. Both an in vitro and in vivo model of gut ischemia-reperfusion (IR) was utilized. In vitro, intestinal epithelial cells underwent hypoxia-reoxygenation to mimic gut IR with 2 mM (physiologic) or 10 mM glutamine supplementation. Permeability, caspase activity, cell growth, and cell surface and shed syndecan 1 were assessed. In vivo, wild-type and syndecan 1 knockout (KO) mice received ± enteral glutamine followed by gut IR. Intestinal injury was assessed by fluorescent dye clearance and histopathology, permeability as mucosal-to-serosal clearance ex vivo in everted sacs, and inflammation by myeloperoxidase (MPO) activity. In an in vitro model of gut IR, glutamine supplementation reduced epithelial cell permeability and apoptosis and enhanced cell growth. Shed syndecan 1 was reduced by glutamine without an increase in syndecan 1 mRNA. In vivo, intestinal permeability, inflammation, and injury were increased after gut IR in wild-type mice and further increased in syndecan 1 KO mice. Glutamine's attenuation of IR-induced intestinal hyperpermeability, inflammation, and injury was abolished in syndecan 1 KO mice. These results suggest that syndecan 1 plays a novel role in the protective effects of enteral glutamine in the postischemic gut.


Subject(s)
Glutamine/therapeutic use , Intestinal Diseases/prevention & control , Reperfusion Injury/prevention & control , Syndecan-1/physiology , Animals , Caspases/metabolism , Cell Division/drug effects , Cell Division/physiology , Cell Hypoxia/physiology , Cells, Cultured , Epithelial Cells/drug effects , Glutamine/pharmacology , Intestinal Diseases/pathology , Intestinal Diseases/physiopathology , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestines/blood supply , Mice , Mice, Inbred C57BL , Mice, Knockout , Permeability/drug effects , Rats , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Syndecan-1/deficiency , Syndecan-1/metabolism
20.
Int J Cancer ; 131(6): E884-96, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22573479

ABSTRACT

microRNAs are small endogenous noncoding RNAs, which post-transcriptionally regulate gene expression. In breast cancer, overexpression of the transmembrane heparan sulfate proteoglycan syndecan-1, a predicted target of the oncomiR miR-10b, correlates with poor clinical outcome. To investigate the potential functional relationship of miR-10b and syndecan-1, MDA-MB-231 and MCF-7 breast cancer cells were transiently transfected with pre-miR-10b, syndecan-1 siRNA or control reagents, respectively. Altered cell behavior was monitored by proliferation, migration and invasion chamber assays, and time-lapse video microscopy. miR-10b overexpression induced post-transcriptional downregulation of syndecan-1, as demonstrated by quantitative real-time PCR (qPCR), flow cytometry, and 3'UTR luciferase assays, resulting in increased cancer cell migration and matrigel invasiveness. Syndecan-1 silencing generated a copy of this phenotype. Adhesion to fibronectin and laminin and basal cell proliferation was increased. Syndecan-1 coimmunoprecipitated with focal adhesion kinase, which showed increased activation upon syndecan-1 depletion. Affymetrix screening and confirmatory qPCR and Western blotting analysis of syndecan-1-deficient cells revealed upregulation of ATF-2, COX-2, cadherin-11, vinculin, actin γ 2, MYL9, transgelin-1, RhoA/C, matrix metalloproteinase 2 (MMP2) and heparanase, and downregulation of AML1/RUNX1, E-cadherin, CLDN1, p21WAF/CIP, cyclin-dependent kinase 6, TLR-4, PAI1/2, Collagen1alpha1, JHDM1D, Mpp4, MMP9, matrilin-2 and ANXA3/A10. Video microscopy demonstrated massively increased Rho kinase-dependent motility of syndecan-1-depleted cells, which displayed increased filopodia formation. We conclude that syndecan-1 is a novel target of the oncomiR miR-10b. Rho-GTPase-dependent modulation of cytoskeletal function and downregulation of E-cadherin expression are identified as relevant effectors of the miR-10b-syndecan-1 axis, which emerges as a promising target for the development of new therapeutic approaches for breast cancer.


Subject(s)
Breast Neoplasms/pathology , Cadherins/physiology , Cell Movement , MicroRNAs/physiology , Syndecan-1/antagonists & inhibitors , rho GTP-Binding Proteins/physiology , Activating Transcription Factor 2/genetics , Cell Communication , Cell Line, Tumor , Core Binding Factor Alpha 2 Subunit/genetics , Disease Progression , Female , Humans , Neoplasm Invasiveness , Phosphorylation , RNA, Messenger/analysis , RNA, Small Interfering/genetics , Syndecan-1/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...