Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
1.
Int J Mol Sci ; 22(19)2021 Sep 22.
Article in English | MEDLINE | ID: mdl-34638548

ABSTRACT

CLEC12A is a myeloid inhibitory receptor that negatively regulates inflammation in mouse models of autoimmune and autoinflammatory arthritis. Reduced CLEC12A expression enhances myeloid cell activation and inflammation in CLEC12A knock-out mice with collagen antibody-induced or gout-like arthritis. Similarly to other C-type lectin receptors, CLEC12A harbours a stalk domain between its ligand binding and transmembrane domains. While it is presumed that the cysteines in the stalk domain have multimerisation properties, their role in CLEC12A expression and/or signaling remain unknown. We thus used site-directed mutagenesis to determine whether the stalk domain cysteines play a role in CLEC12A expression, internalisation, oligomerisation, and/or signaling. Mutation of C118 blocks CLEC12A transport through the secretory pathway diminishing its cell-surface expression. In contrast, mutating C130 does not affect CLEC12A cell-surface expression but increases its oligomerisation, inducing ligand-independent phosphorylation of the receptor. Moreover, we provide evidence that CLEC12A dimerisation is regulated in a redox-dependent manner. We also show that antibody-induced CLEC12A cross-linking induces flotillin oligomerisation in insoluble membrane domains in which CLEC12A signals. Taken together, these data indicate that the stalk cysteines in CLEC12A differentially modulate this inhibitory receptor's expression, oligomerisation and signaling, suggestive of the regulation of CLEC12A in a redox-dependent manner during inflammation.


Subject(s)
Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Membrane Proteins/metabolism , Myeloid Cells/metabolism , Protein Multimerization/genetics , Receptors, Mitogen/genetics , Receptors, Mitogen/metabolism , Cell Line, Tumor , Cysteine/metabolism , HEK293 Cells , HeLa Cells , Humans , Inflammation/genetics , Lectins, C-Type/biosynthesis , Membrane Proteins/genetics , Mutagenesis, Site-Directed , Phosphorylation , Protein Domains/genetics , Protein Transport/genetics , Receptors, Mitogen/biosynthesis , Signal Transduction/immunology
2.
Am J Physiol Lung Cell Mol Physiol ; 315(3): L360-L370, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29792348

ABSTRACT

Although extensive work has delineated many of the mechanisms of extracellular matrix (ECM) production, far less is known about pathways that regulate ECM degradation. This is particularly true of cellular internalization and degradation of matrix, which play an underappreciated role in ECM metabolism and lung fibrosis. For example, genetic perturbation of this pathway leads to exacerbated fibrosis in experimental animal models. In this work, we present the results of an unbiased screen of Drosophila phagocytes that yielded multiple genes that, when silenced, led to increased collagen uptake. We further describe the function of cell division cycle 7 kinase (CDC7) as a specific suppressor of collagen uptake. We show that the genetic or pharmacological inhibition of CDC7 results in increased expression of the collagen endocytic receptor Endo180. Chromobox 5 (CBX5) is a putative target of CDC7, and genetic silencing of CBX5 also results in increased Endo180 and collagen uptake. Finally, CRISPR-mediated activation of Endo180 expression results in increased collagen uptake, suggesting that CDC7 regulates collagen internalization through increased Endo180 expression. Targeting the regulatory elements of the collagen degradative machinery may be a useful therapeutic approach in diseases of fibrosis or malignancy.


Subject(s)
Collagen/metabolism , Drosophila Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proteolysis , Animals , Cell Line , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Collagen/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fibrosis , Gene Expression Regulation, Enzymologic , Protein Serine-Threonine Kinases/genetics , Receptors, Mitogen/biosynthesis , Receptors, Mitogen/genetics
3.
Microbiol Spectr ; 4(5)2016 10.
Article in English | MEDLINE | ID: mdl-27780012

ABSTRACT

Lectins recognize a diverse array of carbohydrate structures and perform numerous essential biological functions. Here we focus on only two families of lectins, the Siglecs and C-type lectins. Triggering of intracellular signaling cascades following ligand recognition by these receptors can have profound effects on the induction and modulation of immunity. In this chapter, we provide a brief overview of each family and then focus on selected examples that highlight how these lectins can influence myeloid cell functioning in health and disease. Receptors that are discussed include Sn (Siglec-1), CD33 (Siglec-3), and Siglec-5, -7, -8, -9, -10, -11, -14, -15, -E, -F, and -G as well as Dectin-1, MICL, Dectin-2, Mincle/MCL, and the macrophage mannose receptor.


Subject(s)
Myeloid Cells/metabolism , Receptors, Mitogen/biosynthesis , Animals , Humans , Myeloid Cells/immunology , Receptors, Mitogen/immunology
4.
Cell Rep ; 11(12): 1905-18, 2015 Jun 30.
Article in English | MEDLINE | ID: mdl-26095365

ABSTRACT

The polymorphism ATG16L1 T300A, associated with increased risk of Crohn's disease, impairs pathogen defense mechanisms including selective autophagy, but specific pathway interactions altered by the risk allele remain unknown. Here, we use perturbational profiling of human peripheral blood cells to reveal that CLEC12A is regulated in an ATG16L1-T300A-dependent manner. Antibacterial autophagy is impaired in CLEC12A-deficient cells, and this effect is exacerbated in the presence of the ATG16L1(∗)300A risk allele. Clec12a(-/-) mice are more susceptible to Salmonella infection, supporting a role for CLEC12A in antibacterial defense pathways in vivo. CLEC12A is recruited to sites of bacterial entry, bacteria-autophagosome complexes, and sites of sterile membrane damage. Integrated genomics identified a functional interaction between CLEC12A and an E3-ubiquitin ligase complex that functions in antibacterial autophagy. These data identify CLEC12A as early adaptor molecule for antibacterial autophagy and highlight perturbational profiling as a method to elucidate defense pathways in complex genetic disease.


Subject(s)
Carrier Proteins/genetics , Crohn Disease/genetics , Lectins, C-Type/genetics , Receptors, Mitogen/genetics , Salmonella Infections/genetics , Alleles , Animals , Autophagy/genetics , Autophagy-Related Proteins , Crohn Disease/microbiology , Crohn Disease/pathology , Genetic Predisposition to Disease , Genomics , Humans , Lectins, C-Type/biosynthesis , Mice , Receptors, Mitogen/biosynthesis , Risk Factors , Salmonella/pathogenicity , Salmonella Infections/microbiology
5.
J Cancer Res Clin Oncol ; 141(11): 1973-83, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25842034

ABSTRACT

PURPOSE: Discoidin domain receptors (DDRs) have been identified as tyrosine kinase receptors for collagen, and the overexpression of DDR1 was correlated with hepatocellular carcinoma (HCC) progression in vitro. Little is known about DDR2 on HCC cells, and we investigated the expression and function of DDR2 in human HCC cells. METHODS: Expression of DDR2 in human HCC cell lines and patient HCC tissues was observed. The suppression of DDR2 by siRNA against DDR2 was performed in vitro and in vivo study. RESULTS: All of HCC cell lines expressed DDR2 mRNA, and all HCC tissues from the ten patients with HCC demonstrated DDR2 mRNA expression. Transfection of DDR2 siRNA significantly inhibits cell growth compared to cells with nontarget siRNA transfection in vitro (P < 0.001). In SNU182, Hep3B, and HeLa cell xenograft models, there was a significant difference in average tumor volumes after 12 days of the DDR2 siRNA injection (P < 0.05) in SNU182 xenograft mice. DDR2 siRNA injection decreased the mean tumor volume by 65.6 % compared to that of the control. The apoptosis analysis demonstrated that DDR2 siRNA treatment significantly increased apoptotic cells (P < 0.01). Cell migration (P < 0.05) and cell invasion (P < 0.01) were significantly decreased by DDR2 siRNA treatment. CONCLUSIONS: The inhibition of DDR2 by RNA interference suppressed in vivo and in vitro growth of human HCC cells. Our results may support that the use of DDR2 as a novel target of HCC treatment through control of tumor apoptosis, migration, and invasion.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Proliferation/genetics , Liver Neoplasms/pathology , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Mitogen/genetics , 3T3 Cells , Animals , Apoptosis/genetics , Carcinoma, Hepatocellular/genetics , Cell Adhesion/genetics , Cell Line, Tumor , Cell Movement/genetics , Discoidin Domain Receptors , Disease Progression , HeLa Cells , Humans , Liver Neoplasms/genetics , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , RNA Interference , RNA, Small Interfering/genetics , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptors, Mitogen/biosynthesis , Transfection , Tumor Burden/genetics
6.
BMC Cancer ; 14: 369, 2014 May 24.
Article in English | MEDLINE | ID: mdl-24885564

ABSTRACT

BACKGROUND: Although many of the recently approved genomically targeted therapies have improved outcomes for patients in non-small-cell lung cancer (NSCLC) with lung adenocarcinoma, little is known about the genomic alterations that drive lung squamous cell cancer (SCC) and development of effective targeted therapies in lung SCC is a promising area to be further investigated. Discoidin domain receptor 2 (DDR2), is a novel receptor tyrosine kinases that respond to several collagens and involved in tissue repair, primary and metastatic cancer progression. METHODS: Expression of DDR2 mRNA was analyzed in 54 lung SCC tissues by qRT-PCR. Over-expression approaches were used to investigate the biological functions of DDR2 and its' mutations in lung SCC cells. Conventional Sanger sequencing was used to investigate the mutations of DDR2 gene in 86 samples. The effect of DDR2 and its' mutations on proliferation was evaluated by MTT and colony formation assays; cell migration and invasion was evaluated by trasnwell assays. Lung SCC cells stably transfected with pEGFP-DDR2 WT, pEGFP-DDR2-S131C or empty vector were injection into nude mice to study the effect of DDR2 and its' mutation on tumorigenesis in vivo. Protein and mRNA expression levels of E-cadherin and MMP2 were determined by qRT-PCR and western blot analysis. Differences between groups were tested for significance using Student's t-test (two-tailed). RESULTS: In this study, we found that DDR2 mRNA levels were significantly decreased in 54 lung SCC tissues compared with normal lung tissues. Moreover, there were 3 novel DDR2 mutations (G531V, S131C, T681I) in 4 patients and provide the mutation rate of 4.6% in the 86 patients with lung SCC. The mutation of S131C in DDR2 could promote lung SCC cells proliferation, migration and invasion via inducing MMP-2, but reducing E-cadherin expression. CONCLUSIONS: These data indicated that the novel DDR2 mutation may contribute to the development and progression of lung SCC and this effect may be associated with increased proliferation and invasiveness, at least in part, via regulating E-cadherin expression.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Mitogen/genetics , Animals , Cadherins/biosynthesis , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Movement/genetics , China , Discoidin Domain Receptors , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mutation , Phosphorylation , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptors, Mitogen/biosynthesis
7.
Matrix Biol ; 34: 13-21, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24480069

ABSTRACT

Maturation of the glomerular basement membrane (GBM) is essential for maintaining the integrity of the renal filtration barrier. Impaired maturation causes proteinuria and renal fibrosis in the type IV collagen disease Alport syndrome. This study evaluates the role of collagen receptors in maturation of the GBM, matrix accumulation and renal fibrosis by using mice deficient for discoidin domain receptor 1 (DDR1), integrin subunit α2 (ITGA2), and type IV collagen α3 (COL4A3). Loss of both collagen receptors DDR1 and integrin α2ß1 delays maturation of the GBM: due to a porous GBM filtration barrier high molecular weight proteinuria that more than doubles between day 60 and day 100. Thereafter, maturation of the GBM causes proteinuria to drop down to one tenth until day 200. Proteinuria and the porous GBM cause accumulation of glomerular and tubulointerstitial matrix, which both decrease significantly after GBM-maturation until day 250. In parallel, in a disease with impaired GBM-maturation such as Alport syndrome, loss of integrin α2ß1 positively delays renal fibrosis: COL4A3(-/-)/ITGA2(-/-) double knockouts exhibited reduced proteinuria and urea nitrogen compared to COL4A3(-/-)/ITGA2(+/-) and COL4A3(-/-)/ITGA2(+/+) mice. The double knockouts lived 20% longer and showed less glomerular and tubulointerstitial extracellular matrix deposition than the COL4A3(-/-) Alport mice with normal integrin α2ß1 expression. Electron microscopy illustrated improvements in the glomerular basement membrane structure. MMP2, MMP9, MMP12 and TIMP1 were expressed at significantly higher levels (compared to wild-type mice) in COL4A3(-/-)/ITGA2(+/+) Alport mice, but not in COL4A3(+/+)/ITGA2(-/-) mice. In conclusion, the collagen receptors DDR1 and integrin α2ß1 contribute to regulate GBM-maturation and to control matrix accumulation. As demonstrated in the type IV collagen disease Alport syndrome, glomerular cell-matrix interactions via collagen receptors play an important role in the progression of renal fibrosis.


Subject(s)
Fibrosis/genetics , Integrin alpha2beta1/genetics , Kidney Glomerulus/growth & development , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptors, Mitogen/biosynthesis , Animals , Discoidin Domain Receptors , Extracellular Matrix/genetics , Extracellular Matrix/pathology , Fibrosis/pathology , Glomerular Basement Membrane/growth & development , Glomerular Basement Membrane/metabolism , Humans , Integrin alpha2beta1/metabolism , Kidney/pathology , Kidney Glomerulus/pathology , Mice , Mice, Knockout , Nephritis, Hereditary/genetics , Nephritis, Hereditary/pathology , Receptors, Collagen/genetics , Receptors, Collagen/metabolism
8.
Med Oncol ; 30(1): 397, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23307244

ABSTRACT

The discoidin domain receptors, DDR1 and DDR2, have been linked with numerous human cancers. We sought to determine expression level and distribution of DDRs in human breast cancer, and investigate prognostic determinates to determine whether levels of DDRs could predict survival. Tumor samples from 122 breast cancer patients were analyzed for relative expression of DDRs. An additional 24 matched tumor and normal tissues were tested for differential expression of DDR1 and DDR2. DDR2 was found to be significantly increased by 6-fold (P = 0.0005) and DDR1 decreased (P = 0.0001) in tumor vs. normal breast tissue. DDR1 expression was not predictive for patient survival; however, DDR2 expression was significantly associated with disease-free (HR = 0.55, 95 % CI = 0.24-0.78, P = 0.026) and overall survival (HR = 0.46, 95 % CI = 0.35-0.84, P = 0.019). Multivariate analysis revealed DDR2 is an independent favorable predictor for prognosis independent of tumor stage, histology, and patient age. The present research provided the first evidence that increased DDR2 mRNA expression in primary human breast cancer might be a powerful, independent predictor of recurrence and outcome.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/metabolism , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptors, Mitogen/biosynthesis , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Discoidin Domain Receptor 1 , Discoidin Domain Receptors , Disease-Free Survival , Female , Humans , Immunohistochemistry , Middle Aged , Neoplasm Staging , Prognosis , Proportional Hazards Models , Real-Time Polymerase Chain Reaction
9.
Med Oncol ; 29(5): 3077-82, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22752569

ABSTRACT

Hepatocellular carcinoma (HCC) is highly invasive with a high frequency of recurrence following surgery and poor prognosis. The underlying molecular mechanisms for HCC recurrence are not well understood. Here, we used microarray technology for genome-wide analysis to identify genes who may be involved in tumor recurrence. cDNA from HCC tumor tissues of patients with early recurrence (ER; n = 10) and patients whose HCC had not recurred ≥ 2 years postsurgery (nER; n = 10) was hybridized to the Affymetrix Human Geome U133 plus 2.0 whole-genome microarray. Gene clusters were identified and used for hierarchial clustering and principal component analysis. Genes with more than twofold change in expression between ER and nER groups were further analyzed. Expression levels of a subset of genes were validated using RT-PCR and immunohistochemistry. A total of 1,646 genes had significantly different expression between the ER and nER groups (P < 0.05) with 61 and 49 genes in the ER upregulated and downregulated for more than twofold in comparison with the nER group, respectively. The cellular functions of differentially expressed genes included cell adhesion, motility, cytoskeleton, transcription, metabolism, signal transduction, and apoptosis. The discoidin domain receptor 1 (DDR1) mRNA expression was significantly higher in the ER (3.36 ± 0.39) compared with the nER group (3.01 ± 0.49; P = 0.020). A greater proportion of liver tissue samples from ER versus nER patients had DDR1 protein expression (80.0 vs. 40.0 %, P = 0.022). Using microarray technology, we identified a number of genes whose expression differed between patients with recurrent HCC compared to those without. DD1 mRNA and protein levels were higher in patients with recurrent HCC, suggesting this gene maybe involved in tumor invasion and metastasis.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Neoplasm Recurrence, Local/genetics , Receptor Protein-Tyrosine Kinases/analysis , Receptors, Mitogen/analysis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cluster Analysis , Discoidin Domain Receptors , Female , Genome-Wide Association Study , Humans , Immunohistochemistry , In Situ Hybridization , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Oligonucleotide Array Sequence Analysis , Principal Component Analysis , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Mitogen/biosynthesis , Receptors, Mitogen/genetics , Reverse Transcriptase Polymerase Chain Reaction
10.
J Cancer Res Clin Oncol ; 138(1): 73-84, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22009181

ABSTRACT

PURPOSE: Vascular endothelial growth factor (VEGF) greatly contributes to the progression of hepatocellular carcinoma (HCC). It is reported that a selective cyclooxygenase-2 (COX-2) inhibitor inhibits cellular proliferation and may attenuate VEGF expression in HCC. We propose that different cascades in the VEGF pathway respond to COX-2 inhibition, depending on the cell types. METHODS: The six human HCC cell lines--Hep3B, SNU387, SNU182, SNU423, SNU449, and PLC/PRF5--were cultured under normoxic and hypoxic conditions. Cells were treated with a selective COX-2 inhibitor (NS-398) and discoidin domain receptor 2 (DDR2) siRNA, and microarray analysis was performed. RESULTS: NS-398 inhibited HCC proliferation and decreased the expression level of VEGF in HCC cells only under normoxia conditions. In hypoxia conditions, VEGF expression level in Hep3B cell was suppressed, while that in SNU387 cell was increased by NS-398 (P < 0.001). The NS-398-induced increase in VEGF expression in SNU387 cell was associated with the up-regulation of the DDR2 gene. NS-398-treated SNU series cells and PLC/PRF5 cells displayed a robust increase in DDR2 mRNA expression. Also, transfection with DDR2 siRNA decreased the VEGF expression level of SNU387, 423, 449 cells under hypoxia conditions (P < 0.05). In vivo chromatin immunoprecipitation assay demonstrated that NS-398 induces the enhancement of HIF-1α binding on VEGF promoter, leading to the increase in VEGF gene expression in hypoxic conditions. There is strong evidence that it is related to the DDR2 gene expression in SNU387 cells. CONCLUSION: These findings disclose a novel cell-dependent regulatory mechanism of VEGF involving DDR2 gene in HCC cells.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Cyclooxygenase 2 Inhibitors/pharmacology , Liver Neoplasms/drug therapy , Nitrobenzenes/pharmacology , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Mitogen/metabolism , Sulfonamides/pharmacology , Vascular Endothelial Growth Factor A/biosynthesis , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Cell Hypoxia/genetics , Cell Hypoxia/physiology , Cell Line, Tumor , Discoidin Domain Receptors , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Promoter Regions, Genetic , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Mitogen/biosynthesis , Receptors, Mitogen/genetics , Transfection , Up-Regulation , Vascular Endothelial Growth Factor A/genetics
11.
Oncol Rep ; 26(4): 971-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21701781

ABSTRACT

Discoidin domain receptors (DDR1 and DDR2) are tyrosine kinase receptors for fibrillar collagen implicated in postnatal development, tissue repair, and primary and metastatic cancer progression. While DDR1 has been described in tumor cells, DDR2 has been localized in the tumor stroma, but its presence in the tumor cells remains unknown. The aim of this study was to elucidate the role of DDR2 signaling in tumor cells during hepatic metastasis progression. DDR2 expression and phosphorylation in cultured human A375 melanoma cells was documented by Western blot analysis. A375 cells were stably transfected with a small interfering RNA (siRNA) against DDR2 and two clones were selected: A375R2-70 and A375R2-40, with 70 and 40% of the DDR2 protein expression respectively, compared to mock-transfected cells (A375R2-100). Development of experimental liver metastasis by intrasplenic inoculation of A375R2-70 and A37R2-40 clones was reduced by 60 and 75%, respectively, measured as tumor volume, compared to livers injected with A375R2-100 cells. Accordingly, A375R2-70 and A37R2-40 clones showed reduced in vitro gelatinase activity and JNK phosphorylation, compared to mock transfected cells, with maximal inhibition in A375R2-40. Additionally, A375 melanoma, SK-HEP hepatoma and HT-29 colon carcinoma human cell lines transiently transfected with siRNA against DDR2 also showed reduced proliferation and migration rates compared to mock-transfected ones. In conclusion, DDR2 promotes A375 melanoma metastasis to the liver and the underlying mechanism implicates regulation of metalloproteinase release, cell growth and chemotactic invasion of the host tissue.


Subject(s)
Liver Neoplasms/enzymology , Liver Neoplasms/secondary , Melanoma/enzymology , Melanoma/secondary , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Mitogen/metabolism , Animals , Cell Growth Processes/physiology , Cell Line, Tumor , Discoidin Domain Receptors , Down-Regulation , Female , HT29 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Melanoma/genetics , Melanoma/pathology , Metalloproteases/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Phosphorylation , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Mitogen/biosynthesis , Receptors, Mitogen/genetics , Signal Transduction , Transfection
12.
J Biol Chem ; 286(15): 12912-23, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21335558

ABSTRACT

Discoidin domain receptors (DDRs) DDR1 and DDR2 are receptor tyrosine kinases with the unique ability among receptor tyrosine kinases to respond to collagen. Several signaling molecules have been implicated in DDR signaling, including Shp-2, Src, and MAPK pathways, but a detailed understanding of these pathways and their transcriptional targets is still lacking. Similarly, the regulation of the expression of DDRs is poorly characterized with only a few inflammatory mediators, such as lipopolysaccharide and interleukin-1ß identified as playing a role in DDR1 expression. DDRs have been reported to induce the expression of various genes including matrix metalloproteinases and bone morphogenetic proteins, but the regulatory mechanisms underlying DDR-induced gene expression remain to be determined. The aim of the present work was to elucidate the molecular mechanisms implicated in the expression of DDRs and to identify DDR-induced signaling pathways and target genes. Our data show that collagen I induces the expression of DDR1 in a dose- and time-dependent manner in primary human lung fibroblasts. Furthermore, activation of DDR2, JAK2, and ERK1/2 MAPK signaling pathways was essential for collagen I-induced DDR1 and matrix metalloproteinase 10 expression. Finally, inhibition of the ERK1/2 pathway abrogated DDR1 expression by blocking the recruitment of the transcription factor polyoma enhancer A-binding protein 3 to the DDR1 promoter. Our data provide new insights into the molecular mechanisms of collagen I-induced DDR1 expression and demonstrate an important role for ERK1/2 activation and the recruitment of polyoma enhancer-A binding protein 3 to the DDR1 promoter.


Subject(s)
Collagen Type I/metabolism , Fibroblasts/metabolism , Gene Expression Regulation/physiology , Janus Kinase 2/metabolism , Lung/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptors, Mitogen/biosynthesis , Collagen Type I/genetics , Discoidin Domain Receptors , Fibroblasts/cytology , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Janus Kinase 2/genetics , Lipopolysaccharides/pharmacology , Lung/cytology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Matrix Metalloproteinase 10/biosynthesis , Matrix Metalloproteinase 10/genetics , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Mitogen/genetics
13.
J Physiol Biochem ; 65(3): 235-41, 2009 Sep.
Article in English | MEDLINE | ID: mdl-20119818

ABSTRACT

Carbohydrates are thought to function as tags that mark circulatory glycoproteins for rapid clearance. Scavenger endothelial cells (SECs) play the primary role in clearing glycoproteins via receptor-mediated endocytosis in adult animals. We found that horseradish peroxidase (HRP), a glycoprotein, was removed quickly, mostly by receptor mediation from the chicken embryo circulation, but bovine serum albumin was not. The half-life of HRP in the circulation varied with the embryo stage and fell rapidly from 0.73 h at embryonic day 4 (E4) to 0.23 h at E5, with no great difference among stages after E5. HRP clearance was far slower at E3.5 than at E5, but was obviously suppressed by mannan. These results imply that the function of clearing glycoprotein or waste macromolecules from the circulation via receptor-mediated endocytosis appears early in the embryo.


Subject(s)
Glycoproteins/metabolism , Mannans/pharmacology , Animals , Chick Embryo/embryology , Chickens , Endocytosis , Endothelial Cells/metabolism , Horseradish Peroxidase/metabolism , Ovalbumin/metabolism , Receptors, Mitogen/biosynthesis , Serum Albumin, Bovine , Time Factors
14.
J Mol Neurosci ; 38(1): 2-11, 2009 May.
Article in English | MEDLINE | ID: mdl-18836851

ABSTRACT

The discoidin domain receptor (DDR1) is highly expressed in oligodendrocytes during the neurodevelopmental myelination process and is genetically associated to schizophrenia. In this study, we aimed to further assess the involvement of DDR1 in both remyelination and oligodendrocyte differentiation. In the mouse model of demyelination-remyelination induced by oral administration of cuprizone, in situ hybridization showed an upregulation of the DDR1 gene in three different white matter areas (corpus callosum, dorsal fornix, and external capsule) during the remyelination period. Moreover, real time reverse transcriptase polymerase chain reaction showed that the increase in DDR1 messenger RNA (mRNA) was strongly correlated with the number of DDR1-positive cells in the corpus callosum (Spearman coefficient = 0.987, P = 0.013). Cells positive for DDR1 mRNA were also positive for oligodendrocyte markers (OLIG2, carnosine, and APC) but not for markers of oligodendrocyte precursors (NG2), myelin markers (CNPase), microglia (CD11b), or reactive glia (GFAP). Differentiation of a human oligodendroglial cell line, HOG16, was associated with an increase in mRNA expression of DDR1 and several myelin proteins (MBP and MOBP) but not other proteins (APC and CNPase). Here, we demonstrate that DDR1 is upregulated in vitro and in vivo when oligodendrocyte myelinating machinery is activated. Further studies are needed to identify the specific molecular pathway.


Subject(s)
Cell Differentiation , Myelin Sheath/physiology , Oligodendroglia/cytology , Oligodendroglia/metabolism , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptors, Mitogen/biosynthesis , Schizophrenia/metabolism , Animals , Cell Line , Corpus Callosum/cytology , Corpus Callosum/metabolism , Cuprizone/administration & dosage , Discoidin Domain Receptors , Humans , Male , Mice , Models, Animal , Monoamine Oxidase Inhibitors/administration & dosage , Myelin Proteins/biosynthesis , Myelin Proteins/genetics , Myelin Sheath/drug effects , Oligodendroglia/drug effects , RNA, Messenger/analysis , RNA, Messenger/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Mitogen/genetics , Up-Regulation
15.
Cancer ; 113(10): 2823-31, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18853422

ABSTRACT

BACKGROUND: Acquired features of cells under epithelial-mesenchymal transition (EMT) have not yet been fully identified. The current study was conducted to assess alterations in both the proliferative potential and the responsiveness to extracellular matrices (ECMs) in EMT. METHODS: MDCK cells and SLUG-transfected MDCK clones (SLUG-MDCK) were used in this study. The cell cycle was analyzed by using flow cytometry and Western blotting. ECM-stimulated cell proliferation was examined by using the following ECMs, type I collagen, type IV collagen, fibronectin, and laminin. Protein phosphorylation was detected by immunoprecipitation-Western by using the 4G10 antibody. RESULTS: Both G1 and G2/M arrest were found in the SLUG-MDCK cells, and the responsible molecules for the cell-cycle arrests were, at least in part, p21WAF1/Cip1 and Wee1. Once in contact with type I collagen, the SLUG-MDCK cells, showing the Wee1 degradation, dramatically started to proliferate up to 6-fold in cell number at Day 5, in contrast to only a 2-fold increase in the control. The analysis of the collagen receptors in the SLUG-MDCK cells disclosed a striking increase in the discoid domain receptor (DDR) 2 expression and a clear decrease in the DDR1 expression. The immunoprecipitated DDR2 protein extracted from SLUG-MDCK cells, which were cultured on collagen for 30 minutes, was tyrosine-phosphorylated, indicating valid functionality of the up-regulated receptor. The altered expression from DDR1 to DDR2 was also found in the naturally dedifferentiated sister cell lines of human liver cancer. CONCLUSIONS: Collectively, SLUG-induced EMT may alter the expression profile of receptor tyrosine kinases, including DDRs.


Subject(s)
Cell Differentiation/physiology , Cell Transformation, Neoplastic/metabolism , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation/physiology , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptors, Mitogen/biosynthesis , Transcription Factors/metabolism , Animals , Blotting, Western , Cell Cycle/physiology , Cell Line , Cell Proliferation , Discoidin Domain Receptors , Dogs , Epithelium/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression , Humans , Immunoprecipitation , Mesoderm/metabolism , Microscopy, Confocal , Snail Family Transcription Factors , Transfection
16.
Biochem Biophys Res Commun ; 374(4): 662-7, 2008 Oct 03.
Article in English | MEDLINE | ID: mdl-18664364

ABSTRACT

Discoidin domain receptor-2 (DDR2) is a receptor tyrosine kinase that binds to the extracellular matrix. We investigated the role of hypoxia in DDR2 expression in vascular smooth muscle cells (VSMCs) and the underlying mechanism. Subjecting VSMCs to hypoxia (2.5% O(2)) induced DDR2 expression; treatments with a specific inhibitor (SB203580) of p38 mitogen-activated protein kinase (MAPK) or p38-specific small interference RNA (siRNA) abolished this hypoxia-induced DDR2 expression. Gel shifting assays showed that hypoxia increased the Myc-Max-DNA binding activity in the promoter region of DDR2; inhibition of p38 MAPK activation by SB203580 and p38-specific siRNA blocked hypoxia-induced DDR2 promoter activity. Hypoxia also induced matrix metalloproteinase-2 (MMP-2) activity in VSMCs and increased their migration. These VSMC responses to hypoxia were inhibited by DDR2- and p38-specific siRNAs. Our results suggested that hypoxia induces DDR2 expression in VSMCs at the transcriptional level, which is mediated by the p38 MAPK pathway and contributes to VSMC migration.


Subject(s)
Cell Movement , Hypoxia/enzymology , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptors, Mitogen/biosynthesis , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , DNA/metabolism , Discoidin Domain Receptors , Electrophoretic Mobility Shift Assay , Hypoxia/genetics , Imidazoles/pharmacology , Male , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Promoter Regions, Genetic , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , Pyridines/pharmacology , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Mitogen/genetics , p38 Mitogen-Activated Protein Kinases/genetics
17.
Cancer Sci ; 99(1): 39-45, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17970783

ABSTRACT

A novel gene, prostate cancer antigen (PCA)-1, was recently reported to be expressed in the prostate; however, its biological roles remain unclear. Knockdown of the PCA-1 gene by small interfering RNA transfection induced apoptosis through reducing the expression of the anti-apoptotic molecule Bcl-xl and cytoplasmic release of cytochrome c in the androgen-independent prostate cancer cell line PC3. Moreover, in vitro matrigel and in vivo chorioallantoic membrane assays showed that silencing of PCA-1 significantly downregulated discoidin receptor (DDR)-1 expression, resulting in suppression of cancer-cell invasion. Transfection with PCA-1 increased the levels of both Bcl-xl and DDR1, which made the cells more invasive through the upregulation of matrix metalloproteinase 9 in DU145. Interestingly, long-term culture using androgen-free medium increased the level of PCA-1 and the related expression of Bcl-xl and DDR-1 in the androgen-sensitive cancer cell line LNCaP, suggesting that PCA-1 signaling is associated with androgen independence. Immunohistochemical analysis in a series of 169 prostate carcinomas showed that PCA-1 and DDR1 were strongly expressed in prostate cancer cells, including preneoplastic lesions, but there was little or no expression in normal epithelium. Moreover, the expression of PCA-1 and DDR-1 was associated with a hormone-independent state of prostate cancer. Taken together, we propose that PCA-1-DDR-1 signaling is a new important axis involved in malignant potential prostate cancer associated with hormone-refractory status.


Subject(s)
Antigens, Neoplasm/physiology , Prostatic Neoplasms/pathology , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Mitogen/physiology , Animals , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/genetics , Apoptosis/physiology , Cell Line, Tumor , Cell Survival , Chick Embryo , Discoidin Domain Receptors , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Immunohistochemistry , Male , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Neoplasm Invasiveness , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Mitogen/biosynthesis , Receptors, Mitogen/genetics , Signal Transduction , Transfection , bcl-X Protein/biosynthesis , bcl-X Protein/genetics
18.
Br J Cancer ; 96(5): 808-14, 2007 Mar 12.
Article in English | MEDLINE | ID: mdl-17299390

ABSTRACT

The discoidin domain receptors, (DDR)1 and DDR2, have been linked to numerous human cancers. We sought to determine expression levels of DDRs in human lung cancer, investigate prognostic determinates, and determine the prevalence of recently reported mutations in these receptor tyrosine kinases. Tumour samples from 146 non-small cell lung carcinoma (NSCLC) patients were analysed for relative expression of DDR1 and DDR2 using quantitative real-time PCR (qRT-PCR). An additional 23 matched tumour and normal tissues were tested for differential expression of DDR1 and DDR2, and previously reported somatic mutations. Discoidin domain receptor 1 was found to be significantly upregulated by 2.15-fold (P=0.0005) and DDR2 significantly downregulated to an equivalent extent (P=0.0001) in tumour vs normal lung tissue. Discoidin domain receptor 2 expression was not predictive for patient survival; however, DDR1 expression was significantly associated with overall (hazard ratio (HR) 0.43, 95% CI=0.22-0.83, P=0.014) and disease-free survival (HR=0.56, 95% CI=0.33-0.94, P=0.029). Multivariate analysis revealed DDR1 is an independent favourable predictor for prognosis independent of tumour differentiation, stage, histology, and patient age. However, contrary to previous work, we did not observe DDR mutations. We conclude that whereas altered expression of DDRs may contribute to malignant progression of NSCLC, it is unlikely that this results from mutations in the DDR1 and DDR2 genes that we investigated.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptors, Mitogen/biosynthesis , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , DNA Mutational Analysis , Discoidin Domain Receptors , Humans , Lung Neoplasms/genetics , Male , Mice , Mutation , Polymorphism, Genetic , Prognosis , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Mitogen/genetics , Reverse Transcriptase Polymerase Chain Reaction
19.
Am J Physiol Regul Integr Comp Physiol ; 292(1): R415-22, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16917016

ABSTRACT

High-potassium diets can improve vascular function, yet the effects of potassium supplementation on ischemic stroke have not been studied. We hypothesized that dietary potassium supplementation would reduce ischemic cerebral infarct size by reversing cerebral artery hypertrophy. Six-week-old male stroke-prone spontaneously hypertensive rats (SHRSP) were fed diets containing 0.79% potassium (LK) or 2.11% potassium (HK) for 6 wk; Wistar-Kyoto (WKY) rats were fed the LK diet. The HK diet did not reduce blood pressure, as measured by telemetry, in the SHRSP. Cerebral ischemia was induced by middle cerebral artery (MCA) occlusion. The resultant infarct was smaller in the HK-SHRSP than in the LK-SHRSP: 55.1 +/- 6.3 vs. 71.4 +/- 2.4% of the hemisphere infarcted (P < 0.05). Infarcts were smaller in WKY rats (33.5 +/- 4.8%) than in LK-SHRSP or HK-SHRSP. The vessel wall of MCAs from LK-SHRSP was hypertrophied compared with WKY rats; this was reversed in HK-SHRSP. RT-PCR analysis of the cerebral vessels showed that expression of platelet-derived growth factor receptors-alpha and -beta, epidermal growth factor receptor, and collagen I and III was increased in the vessels from LK-SHRSP compared with WKY rats and reduced in HK-SHRSP. These results suggest that potassium supplementation provides neuroprotection in a model of ischemic stroke independent of blood pressure and possibly through changes in vascular structure.


Subject(s)
Blood Vessels/pathology , Hypertension/pathology , Infarction, Middle Cerebral Artery/pathology , Potassium, Dietary/pharmacology , Animals , Blood Pressure/drug effects , Body Weight/drug effects , Cell Proliferation/drug effects , Collagen/biosynthesis , Diet , Hypertension/complications , Infarction, Middle Cerebral Artery/etiology , Male , Muscle Contraction/physiology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Potassium/blood , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptors, Mitogen/biosynthesis , Receptors, Vascular Endothelial Growth Factor/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Telemetry
20.
J Histochem Cytochem ; 55(4): 347-53, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17189524

ABSTRACT

Local growth, invasion, and metastasis of malignancies of the head and neck involve extensive degradation and remodeling of the underlying, collagen-rich connective tissue. Urokinase plasminogen activator receptor-associated protein (uPARAP)/Endo180 is an endocytic receptor recently shown to play a critical role in the uptake and intracellular degradation of collagen by mesenchymal cells. As a step toward determining the putative function of uPARAP/Endo180 in head and neck cancer progression, we used immunohistochemistry to determine the expression of this collagen internalization receptor in 112 human squamous cell carcinomas and 19 normal or tumor-adjacent head and neck tissue samples from the tongue, gingiva, cheek, tonsils, palate, floor of mouth, larynx, maxillary sinus, upper jaw, nasopharynx/nasal cavity, and lymph nodes. Specificity of detection was verified by staining of serial sections with two different monoclonal antibodies against two non-overlapping epitopes on uPARAP/Endo180 and by the use of isotype-matched non-immune antibodies. uPARAP/Endo180 expression was observed in stromal fibroblast-like, vimentin-positive cells. Furthermore, expression of the collagen internalization receptor was increased in tumor stroma compared with tumor-adjacent connective tissue or normal submucosal connective tissue and was most prominent in poorly differentiated tumors. These data suggest that uPARAP/Endo180 participates in the connective tissue destruction during head and neck squamous cell carcinoma progression by mediating cellular uptake and lysosomal degradation of collagen.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Head and Neck Neoplasms/metabolism , Receptors, Mitogen/biosynthesis , Blotting, Western , Carcinoma, Squamous Cell/pathology , Disease Progression , Head and Neck Neoplasms/pathology , Humans , Immunohistochemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...