Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
2.
Osteoarthritis Cartilage ; 30(8): 1050-1061, 2022 08.
Article in English | MEDLINE | ID: mdl-35460872

ABSTRACT

Joint-on-a-chip (JOC) models are powerful tools that aid in osteoarthritis (OA) research. These microfluidic devices apply emerging organ-on-a-chip technology to recapitulate a multifaceted joint tissue microenvironment. JOCs address the need for advanced, dynamic in vitro models that can mimic the in vivo tissue environment through joint-relevant biomechanical or fluidic integration, an aspect that existing in vitro OA models lack. There are existing review articles on OA models that focus on animal, tissue explant, and two-dimensional and three-dimensional (3D) culture systems, including microbioreactors and 3D printing technology, but there has been limited discussion of JOC models. The aim of this article is to review recent developments in human JOC technology and identify gaps for future advancements. Specifically, mechanical stimulation systems that mimic articular movement, multi-joint tissue cultures that enable crosstalk, and systems that aim to capture aspects of OA inflammation by incorporating immune cells are covered. The development of an advanced JOC model that captures the dynamic joint microenvironment will improve testing and translation of potential OA therapeutics.


Subject(s)
Lab-On-A-Chip Devices , Osteoarthritis , Animals , Humans , Tissue Engineering/methods
3.
Osteoarthritis Cartilage ; 30(1): 147-159, 2022 01.
Article in English | MEDLINE | ID: mdl-34547432

ABSTRACT

OBJECTIVE: There is a need to incorporate multiple tissues into in vitro OA models to evaluate novel therapeutics. This approach is limited by inherent donor variability. We present an optimized research tool: a human OA cartilage-synovium explant co-culture model (OA-EXM) that employs donor-matched lower and upper limit response controls combined with statistical approaches to address variability. Multiple rapid read-outs allow for evaluation of therapeutics while cataloguing cartilage-synovium interactions. DESIGN: 48-h human explant cultures were sourced from OA knee arthroplasties. An OA-like cartilage-synovium co-culture baseline was established relative to donor-matched upper limit supraphysiological pro-inflammatory cytokine and lower limit OA cartilage or synovium alone controls. 100 nM dexamethasone treatment validated possible "rescue effects" within the OA-EXM dual tissue environment. Gene expression, proteoglycan loss, MMP activity, and soluble protein concentrations were analyzed using blocking and clustering methods. RESULTS: The OA-EXM demonstrates the value of the co-culture approach as the addition of OA synovium increases OA cartilage proteoglycan loss and expression of MMP1, MMP3, MMP13, CXCL8, CCL2, IL6, and PTGS2, but not to the extent of supraphysiological stimulation. Conversely, OA cartilage does not affect gene expression or MMP activity of OA synovium. Dexamethasone shows dual treatment effects on synovium (pro-resolving macrophage upregulation, protease downregulation) and cartilage (pro-inflammatory, catabolic, and anabolic downregulation), and decreases soluble CCL2 levels in co-culture, thereby validating OA-EXM utility. CONCLUSIONS: The OA-EXM is representative of late-stage OA pathology, captures dual interactions between cartilage and synovium, and combined with statistical strategies provides a rapid, sensitive research tool for evaluating OA therapeutics.


Subject(s)
Cartilage, Articular/pathology , Osteoarthritis/pathology , Synovial Membrane/pathology , Aged , Aged, 80 and over , Coculture Techniques , Female , Humans , Male , Middle Aged , Multivariate Analysis
4.
Oncogene ; 37(5): 673-686, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29035390

ABSTRACT

Urothelial carcinoma (UC) carcinogenesis has been hypothesized to occur through epigenetic repression of tumor-suppressor genes (TSGs). By quantitative real-time polymerase chain reaction array, we found that one potential TSG, angiopoietin-like 4 (ANGPTL4), was expressed at very low levels in all bladder cancer cell lines we examined. Previous studies had demonstrated that ANGPTL4 is highly expressed in some cancers, but downregulated, by DNA methylation, in others. Consequently, owing to these seemingly conflicting functions in distinct cancers, the precise role of ANGPTL4 in the etiology of UC remains unclear. In this study, using methylation-specific PCR and bisulfite pyrosequencing, we show that ANGPTL4 is transcriptionally repressed by DNA methylation in UC cell lines and primary tumor samples, as compared with adjacent noncancerous bladder epithelium. Functional studies further demonstrated that ectopic expression of ANGPTL4 potently suppressed UC cell proliferation, monolayer colony formation in vitro, and invasion, migration, and xenograft formation in vivo. Surprisingly, circulating ANGPTL4 was significantly higher in plasma samples from UC patients than normal control, suggesting it might be secreted from other cell types. Interestingly, our data also indicated that exogenous cANGPTL4 could promote cell proliferation and cell migration via activation of signaling through the Erk/focal adhesion kinase axis. We further confirmed that mouse xenograft tumor growth could be promoted by administration of exogenous cANGPTL4. Finally, immunohistochemistry demonstrated that ANGPTL4 was downregulated in tumor cells but overexpressed in tumor adjacent stromal tissues of muscle-invasive UC tissue samples. In conclusion, our data support dual roles for ANGPTL4 in UC progression, either as a tumor suppressor or oncogene, in response to microenvironmental context.


Subject(s)
Angiopoietin-Like Protein 4/genetics , Carcinoma, Transitional Cell/genetics , Epigenesis, Genetic/genetics , Gene Expression Regulation, Neoplastic/genetics , Tumor Microenvironment , Urinary Bladder Neoplasms/genetics , Aged , Aged, 80 and over , Angiopoietin-Like Protein 4/blood , Angiopoietin-Like Protein 4/metabolism , Animals , Carcinogenesis/genetics , Carcinoma, Transitional Cell/pathology , Carcinoma, Transitional Cell/surgery , Case-Control Studies , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cystectomy , DNA Methylation/genetics , Down-Regulation , Female , Genes, Tumor Suppressor , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Oncogenes/genetics , Promoter Regions, Genetic/genetics , Signal Transduction , Urinary Bladder/pathology , Urinary Bladder/surgery , Urinary Bladder Neoplasms/pathology , Urothelium/pathology , Xenograft Model Antitumor Assays
5.
Oncogene ; 35(16): 2133-44, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-26212015

ABSTRACT

Accumulating evidence reveals the effectiveness of epigenetic therapy in gastric cancer. However, the molecular mechanisms and targets underlying such therapeutic responses remain elusive. Herein, we report an aberrant yet therapeutically rectifiable epigenetic signaling in gastric carcinogenesis. Administration of DNA-demethylating drug 5-aza-2'-deoxycytidine (5-aza-dC) reduced gastric cancer incidence by ~74% (P < 0.05) in N-nitroso-N-methylurea-treated mice. Through genome-wide methylation scanning, novel promoter hypermethylation-silenced and drug-targeted genes were identified in the resected murine stomach tumors and tissues. We uncovered that growth/differentiation factor 1 (Gdf1), a member of the transforming growth factor-ß superfamily, was silenced by promoter hypermethylation in control tumor-bearing mice, but became reactivated in 5-aza-dC-treated mice (P < 0.05). In parallel, the downregulated SMAD2/3 phosphorylation in gastric cancer was revived by 5-aza-dC in vivo. Such hypermethylation-dependent silencing and 5-aza-dC-mediated reactivation of GDF1-SMAD2/3 activity was conserved in human gastric cancer cells (P < 0.05). Subsequent functional characterization further revealed the antiproliferative activity of GDF1, which was exerted through activation of SMAD2/3/4-mediated signaling, transcriptional controls on p15, p21 and c-Myc cell-cycle regulators and phosphorylation of retinoblastoma protein. Clinically, hypermethylation and loss of GDF1 was significantly associated with reduced phosphorylated-SMAD2/3 and poor survival in stomach cancer patients (P < 0.05). Taken together, we demonstrated a causal relationship between DNA methylation and a tumor-suppressive pathway in gastric cancer. Epigenetic silencing of GDF1 abrogates the growth-inhibitory SMAD signaling and renders proliferation advantage to gastric epithelial cells during carcinogenesis. This study lends support to epigenetic therapy for gastric cancer chemoprevention and identifies a potential biomarker for prognosis.


Subject(s)
Epigenesis, Genetic , Gene Silencing , Growth Differentiation Factor 1/genetics , Signal Transduction/genetics , Smad Proteins/metabolism , Stomach Neoplasms/pathology , Animals , DNA Methylation , Humans , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism
6.
Oncogene ; 32(1): 15-26, 2013 Jan 03.
Article in English | MEDLINE | ID: mdl-22330137

ABSTRACT

Epigenetic modifications are a driving force in carcinogenesis. However, their role in cancer metastasis remains poorly understood. The present study investigated the role of DNA methylation in the cervical cancer metastasis. Here, we report evidence of the overexpression of DNA methyltransferases 3B (DNMT3B) in invasive cervical cancer and of the inhibition of metastasis by DNMT3B interference. Using methyl-DNA immunoprecipitation coupled with microarray analysis, we found that the protein tyrosine phosphatase receptor type R (PTPRR) was silenced through DNMT3B-mediated methylation in the cervical cancer. PTPRR inhibited p44/42 MAPK signaling, the expression of the transcription factor AP1, human papillomavirus (HPV) oncogenes E6/E7 and DNMTs. The methylation status of PTPRR increased in cervical scrapings (n=358) in accordance with disease severity, especially in invasive cancer. Methylation of the PTPRR promoter has an important role in the metastasis and may be a biomarker of invasive cervical cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Epigenesis, Genetic , Gene Silencing , MAP Kinase Signaling System , Neoplasm Metastasis , Receptor-Like Protein Tyrosine Phosphatases, Class 7/genetics , Uterine Cervical Neoplasms/pathology , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation , Down-Regulation , Female , Humans , Neoplasm Invasiveness , Promoter Regions, Genetic , Uterine Cervical Neoplasms/enzymology , Uterine Cervical Neoplasms/genetics , DNA Methyltransferase 3B
7.
Br J Cancer ; 92(12): 2190-4, 2005 Jun 20.
Article in English | MEDLINE | ID: mdl-15942635

ABSTRACT

While there is no reliable serum biomarker for the diagnosis and monitoring of patients with gastric cancer, we tested the potential diagnostic and prognostic values of detecting methylation changes in the serum of gastric cancer patients. DNA was extracted from the pretherapeutic serum of 60 patients with confirmed gastric adenocarcinoma and 22 age-matched noncancer controls. Promoter hypermethylation in 10 tumour-related genes (APC, E-cadherin, GSTP1, hMLH1, MGMT, p15, p16, SOCS1, TIMP3 and TGF-beta RII) was determined by quantitative methylation-specific PCR (MethyLight). Preferential methylation in the serum DNA of gastric cancer patients was noted in APC (17%), E-cadherin (13%), hMLH1 (41%) and TIMP3 (17%) genes. Moreover, patients with stages III/IV diseases tended to have higher concentrations of methylated APC (P = 0.08), TIMP3 (P = 0.005) and hMLH1 (P = 0.03) in the serum. In all, 33 cancers (55%) had methylation detected in the serum in at least one of these four markers, while three normal subjects had methylation detected in the serum (specificity 86%). The combined use of APC and E-cadherin methylation markers identified a subgroup of cancer patients with worse prognosis (median survival 3.3 vs 16.1 months, P = 0.006). These results suggest that the detection of DNA methylation in the serum may carry both diagnostic and therapeutic values in gastric cancer patients.


Subject(s)
Adenocarcinoma/genetics , DNA, Neoplasm/genetics , Stomach Neoplasms/genetics , Adenocarcinoma/blood , Adenocarcinoma/diagnosis , Adult , Aged , Aged, 80 and over , DNA Methylation , DNA, Neoplasm/blood , Female , Genes, Tumor Suppressor/physiology , Humans , Male , Middle Aged , Oncogenes/physiology , Prognosis , Promoter Regions, Genetic , Stomach Neoplasms/blood , Stomach Neoplasms/diagnosis
8.
Br J Cancer ; 91(7): 1335-41, 2004 Oct 04.
Article in English | MEDLINE | ID: mdl-15354212

ABSTRACT

The interleukin-mediated Janus kinase (JAK)/STAT pathway plays a crucial role in carcinogenesis. Recently, increased STAT3 activity was found in hepatocellular carcinoma and multiple myeloma in which there was silencing of SOCS-1 (suppressor of cytokine signalling-1) by gene promoter hypermethylation. We investigated the expression level of interleukin-6 (IL-6) and SOCS-1 in gastric cancer cell lines. Expression of SOCS-1 correlated with IL-6 level in most of the cell lines, except for AGS cells in which SOCS-1 was absent despite a high level of IL-6 production. Methylation analysis by methylation-specific polymerase chain reaction and bisulphite sequencing revealed that CpG island of SOCS-1 was densely methylated in AGS cells. Demethylation treatment by 5'aza-deoxycytidine restored SOCS-1 expression and also suppressed constitutive STAT3 phosphorylation in AGS cells. Moreover, methylation of SOCS-1 was detected in 27.5% (11 of 40) of primary gastric tumours samples, 10% (one of 10) of adjacent noncancer tissues but not in any (zero of nine) normal gastric mucosa. Methylation of SOCS-1 also correlated with the loss of mRNA expression in some primary gastric cancers. In conclusion, this is the first report to demonstrate that hypermethylation of SOCS-1 led to gene silencing in gastric cancer cell line and primary tumour samples. Downregulation of SOCS-1 cooperates with IL-6 in the activation of JAK/STAT pathway in gastric cancer.


Subject(s)
Carrier Proteins/metabolism , DNA-Binding Proteins/pharmacology , Interleukin-6/pharmacology , Intracellular Signaling Peptides and Proteins , Protein-Tyrosine Kinases/pharmacology , Repressor Proteins/metabolism , Stomach Neoplasms/pathology , Trans-Activators/pharmacology , Carrier Proteins/biosynthesis , Cell Transformation, Neoplastic , DNA Methylation , Down-Regulation , Humans , Janus Kinase 1 , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Repressor Proteins/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins , Tumor Cells, Cultured
9.
Gut ; 53(3): 331-8, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14960510

ABSTRACT

BACKGROUND AND AIMS: Although peroxisome proliferator activated receptor gamma (PPARgamma) agonists have been implicated in differentiation and growth inhibition of cancer cells, the potential therapeutic and chemopreventive effects on gastric cancer are poorly defined. We examined the in vitro and in vivo effects of PPARgamma ligands on growth of gastric cancer, and the effect of PPARgamma activation on expression of cyclooxygenase 2 (COX-2) and cancer related genes. METHODS: Gastric cell lines (MKN28 and MKN45) were treated with two specific PPARgamma ligands: ciglitazone and 15-deoxy-Delta(12,)(14)-prostaglandin J(2). Cell growth was determined by bromodeoxyuridine incorporation assay and apoptosis was measured by DNA fragmentation. Expression of COX-2 was determined by western blot and real time quantitative polymerase chain reaction (PCR). Expression profiles of cancer related genes were screened with cDNA array. In vivo growth of implanted MKN45 cells in nude mice was monitored after oral treatment with rosiglitazone. RESULTS: PPARgamma ligands suppressed the in vitro growth of MKN45 cells in a dose dependent manner whereas prostacyclin, a PPARdelta agonist, had no growth inhibitory effect. Growth inhibition was more pronounced in MKN45 cells, which was accompanied by DNA fragmentation and downregulation of COX-2. Screening by cDNA microarray showed that PPARgamma ligand treatment was associated with upregulation of bad and p53, and downregulation of bcl-2, bcl-xl, and cyclin E1 in MKN45 cells, which was confirmed by quantitative real time PCR. In contrast, MKN28 cells with lower PPARgamma and COX-2 expression levels had lower growth inhibitory responses to PPARgamma ligands. Microarray experiments only showed induction of the bad gene in MKN28 cells. In vivo growth of MKN45 cells in nude mice was retarded by rosiglitazone. Mean tumour volume in rosiglitazone treated mice was significantly lower than controls at six weeks (p = 0.019) and seven weeks (p = 0.001) after treatment. CONCLUSIONS: PPARgamma ligands suppress both in vitro and in vivo growth of gastric cancer and may play a major role in cancer therapy and prevention.


Subject(s)
Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins , Neoplasm Proteins/physiology , Prostaglandin D2/analogs & derivatives , Stomach Neoplasms/pathology , Transcription Factors/physiology , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Division/drug effects , Cyclooxygenase 2 , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Humans , Isoenzymes/metabolism , Ligands , Male , Membrane Proteins , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Nuclear Receptor Coactivators , Prostaglandin D2/pharmacology , Prostaglandin-Endoperoxide Synthases/metabolism , Rosiglitazone , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Thiazolidinediones/pharmacology , Thiazolidinediones/therapeutic use , Transcription Factors/agonists , Tumor Cells, Cultured/drug effects
10.
Br J Cancer ; 88(10): 1560-5, 2003 May 19.
Article in English | MEDLINE | ID: mdl-12771922

ABSTRACT

Expression of cyclin D2 is absent in 30-70% of gastric cancers. We investigated the role of promoter hypermethylation in the transcriptional silencing of cyclin D2 in five gastric cell lines and 47 primary gastric carcinomas. CpG island methylation status of the cyclin D2 gene was studied by methylation-specific polymerase chain reaction and bisulphite sequencing. RNA and protein expression was analysed by reverse transcription-PCR and Western blot, respectively. Dense methylation of cyclin D2 was detected in three cell lines (KATOIII, AGS and NCI-N87), which also lacked cyclin D2 mRNA and protein expression. Bisulphite DNA sequencing revealed that loss of cyclin D2 expression was closely associated with the density of methylation in the promoter region. Treatment with DNA methyltransferase inhibitor, 5-aza-2'-deoxycytidine, restored the cyclin D2 expression level in methylated gastric cells. Among the 47 primary gastric cancers, cyclin D2 hypermethylation was detected in 23 (48.9%) cases. None of the 23 normal gastric biopsies from noncancer patients showed hypermethylation. Hypermethylation was associated with loss of mRNA (P&<0.001) and protein (P=0.006) expressions. Our study showed that cyclin D2 hypermethylation is associated with loss of cyclin D2 expression in a subset of gastric cancers, which may suggest an alternative gastric carcinogenesis pathway in the absence of cyclin D2 expression.


Subject(s)
Carcinoma/genetics , Carcinoma/pathology , Cyclins/biosynthesis , DNA Methylation , Nerve Tissue Proteins/biosynthesis , Promoter Regions, Genetic/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Blotting, Western , Cell Transformation, Neoplastic , Cyclin D2 , Humans , Polymerase Chain Reaction , RNA, Messenger/analysis , Transcription, Genetic , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...