Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Trials ; 24(1): 346, 2023 May 22.
Article in English | MEDLINE | ID: mdl-37217960

ABSTRACT

BACKGROUND: Hearing loss is the third leading global cause of disability and is associated with poorer quality of life. Hearing aids are often recommended for hearing loss; however, hearing aid uptake and use rates are perpetually low. Motivational interviewing (MI) is a patient-centered counseling aimed at addressing the desire in the patient to change their behavior. The aim of this study is to investigate the impact of one-on-one MI sessions on hearing aid use among new adult users. METHODS: A multi-center, prospective, randomized patient-blind controlled trial with a pre- and post-tests design. New hearing aid users ≥ 18 years of age will be recruited from Vancouver, Canada. They will be randomly assigned to a treatment or control group. The treatment group will attend a one-on-one MI session hosted by a practicing MI therapist in addition to standard in-person audiological care. The control group will receive standard in-person audiological care. Data is collected at baseline and at 1, 3, 6, and 12 months' follow-ups. The primary outcomes are data-logged hearing aid use hours and patient-reported outcomes as measured by the International Outcome Inventory for Hearing Aids questionnaire. Associations between intervention and hearing aid use hours and self-reported outcome measures will be assessed. DISCUSSION: This trial is designed to evaluate the efficacy of one-on-one MI in improving hearing aid use in new adult users in the short and long terms. Results will contribute to the evidence on whether MI counseling has an effect on hearing aid use and may guide future clinical practices. TRIAL REGISTRATION: ClinicalTrials.gov NCT04673565 . Registered on 17 December 2020.


Subject(s)
Deafness , Hearing Aids , Hearing Loss , Motivational Interviewing , Adult , Humans , Quality of Life , Prospective Studies , Hearing Loss/diagnosis , Hearing Loss/therapy , Randomized Controlled Trials as Topic , Multicenter Studies as Topic
2.
Analyst ; 147(7): 1425-1439, 2022 Mar 28.
Article in English | MEDLINE | ID: mdl-35253812

ABSTRACT

Raman spectroscopy is a non-destructive analysis technique that provides detailed information about the chemical structure of tumors. Raman spectra of 52 giant cell tumors of bone (GCTB) and 21 adjacent normal tissues of formalin-fixed paraffin embedded (FFPE) and frozen specimens were obtained using a confocal Raman spectrometer and analyzed with machine learning and deep learning algorithms. We discovered characteristic Raman shifts in the GCTB specimens. They were assigned to phenylalanine and tyrosine. Based on the spectroscopic data, classification algorithms including support vector machine, k-nearest neighbors and long short-term memory (LSTM) were successfully applied to discriminate GCTB from adjacent normal tissues of both the FFPE and frozen specimens, with the accuracy ranging from 82.8% to 94.5%. Importantly, our LSTM algorithm showed the best performance in the discrimination of the frozen specimens, with a sensitivity and specificity of 93.9% and 95.1% respectively, and the AUC was 0.97. The results of our study suggest that confocal Raman spectroscopy accomplished by the LSTM network could non-destructively evaluate a tumor margin by its inherent biochemical specificity which may allow intraoperative assessment of the adequacy of tumor clearance.


Subject(s)
Deep Learning , Giant Cell Tumors , Algorithms , Humans , Spectrum Analysis, Raman/methods , Support Vector Machine
3.
Front Immunol ; 11: 2131, 2020.
Article in English | MEDLINE | ID: mdl-33013899

ABSTRACT

Background and methods: Host genomic alterations are closely related to dysfunction of CD4+ T lymphocytes in the HIV-host interplay. However, the roles of aberrant DNA methylation and gene expression in the response to HIV infection are not fully understood. We investigated the genome-wide DNA methylation and transcriptomic profiles in two HIV-infected T lymphocyte cell lines using high-throughput sequencing. Results: Based on DNA methylation data, we identified 3,060 hypomethylated differentially methylated regions (DMRs) and 2,659 hypermethylated DMRs in HIV-infected cells. Transcription-factor-binding motifs were significantly associated with methylation alterations, suggesting that DNA methylation modulates gene expression by affecting the binding to transcription factors during HIV infection. In support of this hypothesis, genes with promoters overlapping with DMRs were enriched in the biological function related to transcription factor activities. Furthermore, the analysis of gene expression data identified 1,633 upregulated genes and 2,142 downregulated genes on average in HIV-infected cells. These differentially expressed genes (DEGs) were significantly enriched in apoptosis-related pathways. Our results suggest alternative splicing as an additional mechanism that may contribute to T-cell apoptosis during HIV infection. We also demonstrated a genome-scale correlation between DNA methylation and gene expression in HIV-infected cells. We identified 831 genes with alterations in both DNA methylation and gene expression, which were enriched in apoptosis. Our results were validated using various experimental methods. In addition, consistent with our in silico results, a luciferase assay showed that the activity of the PDX1 and SMAD3 promoters was significantly decreased in the presence of HIV proteins, indicating the potential of these genes as genetic markers of HIV infection. Conclusions: Our results suggest important roles for DNA methylation and gene expression regulation in T-cell apoptosis during HIV infection. We propose a list of novel genes related to these processes for further investigation. This study also provides a comprehensive characterization of changes occurring at the transcriptional and epigenetic levels in T cells in response to HIV infection.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Genome/genetics , HIV Infections/genetics , HIV-1/physiology , Promoter Regions, Genetic/genetics , Apoptosis , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation , HEK293 Cells , Homeodomain Proteins/genetics , Humans , Smad3 Protein/genetics , Trans-Activators/genetics , Transcriptional Activation
4.
J Orthop Res ; 38(2): 297-310, 2020 02.
Article in English | MEDLINE | ID: mdl-31471919

ABSTRACT

Giant cell tumor of bone (GCTB) is a locally aggressive destructive bone lesion. The management of pulmonary metastasis and local recurrence after the surgical treatment of GCTB remains a challenge. Pathologically, stromal cells in GCTB are known as primary neoplastic cells and are recognized as incompletely differentiated preosteoblasts. Therefore, inducing GCTB stromal cells to differentiate into cells with a mature osteoblastic phenotype may stop tumor growth and recurrence. In this study, we aimed to investigate how simvastatin, a clinically approved and commonly used statin that has been known to promote the maturation of cells of the osteogenic lineage, affects GCTB stromal cells. We found that simvastatin effectively inhibited cell viability by suppressing proliferation and by inducing apoptosis in GCTB stromal cells. Moreover, simvastatin treatment upregulated the expression of genes related to osteogenic maturation, such as runt-related transcription factor 2, osteopontin, and osteocalcin, and increased the mineralization of the extracellular matrix in GCTB stromal cells. Ingenuity pathway analysis was used to discover that the vitamin D receptor pathway was involved in the simvastatin-induced osteogenic differentiation of GCTB stromal cells by upregulating the 1,25-dihydroxyvitamin D metabolism. Taken together, this in vitro study demonstrates the antitumor and differentiation-promoting effects of simvastatin on GCTB stromal cells and suggests the possibility of using simvastatin as an adjuvant therapy for GCTB. These findings support further clinical investigation of the efficacy of using simvastatin as an adjuvant therapy for GCTB to reduce recurrence and distant metastasis after surgical treatment. © 2019 Orthopedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:297-310, 2020.


Subject(s)
Giant Cell Tumor of Bone/drug therapy , Hypolipidemic Agents/therapeutic use , Simvastatin/therapeutic use , Cell Differentiation/drug effects , Cell Line, Tumor , Drug Screening Assays, Antitumor , Giant Cell Tumor of Bone/metabolism , Humans , Hypolipidemic Agents/pharmacology , Simvastatin/pharmacology , Stromal Cells/drug effects , Stromal Cells/metabolism , Vitamin D/analogs & derivatives , Vitamin D/metabolism
7.
Dig Dis Sci ; 63(3): 676-686, 2018 03.
Article in English | MEDLINE | ID: mdl-29353445

ABSTRACT

BACKGROUND AND AIM: Hepatocellular carcinoma (HCC) is a complicated disease with low survival rate partially due to frequent recurrence and no efficient therapy. Promoter hypermethylation of tumor suppressor genes has been demonstrated as one of the molecular mechanisms contributing to tumorigenesis and progression in HCC. This study aims to investigate regulation of NKAPL expression by promoter methylation and its clinical relevance as a biomarker for HCC. METHODS: We measured mRNA expression of NKAPL in 5 HCC cell lines and a cohort of 62 pairs of primary HCC tumor and their adjacent non-cancer liver tissues. NKAPL protein expression on HCC cell lines and clinical samples was assessed by Western blot and immunohistochemistry, respectively. Association analyses between NKAPL expression and clinicopathologic characteristics in the cohort were conducted. Methylation statuses of NKAPL promoter in 18 pairs of tumor and adjacent non-tumor HCC samples were studied using methylation-specific PCR. Biological functions of NKAPL in HCC were investigated by ectopic expression of NKAPL in HCC cells, and cell viability and cell cycle analyses were performed. RESULTS: Our present study showed suppressed expression and promoter hypermethylation are common events in HCC. Demethylation experiment in HCC cells demonstrated that the NKAPL expression was regulated by promoter methylation. In addition, high methylation level of NKAPL and its low expression predict poor outcome. Furthermore, ectopic expression of NKAPL in the HCC cells inhibited cell growth. CONCLUSIONS: Our findings suggest that methylation of NKAPL is a frequent event and is a potential prognosis biomarker in HCC.


Subject(s)
Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/metabolism , Co-Repressor Proteins/metabolism , Liver Neoplasms/diagnosis , Liver Neoplasms/metabolism , Nuclear Proteins/metabolism , Biomarkers/metabolism , Case-Control Studies , Co-Repressor Proteins/genetics , Female , Humans , Male , Methylation , Nuclear Proteins/genetics , Prognosis , Promoter Regions, Genetic , RNA, Messenger/metabolism
8.
J Cell Biochem ; 118(6): 1349-1360, 2017 06.
Article in English | MEDLINE | ID: mdl-27862217

ABSTRACT

Giant cell tumor of bone (GCTB) is the most common non-malignant primary bone tumor reported in Hong Kong. Failure of treatment in advanced GCTB with aggressive local recurrence remains a clinical challenge. In order to reveal the molecular mechanism underlying the pathogenesis of this tumor, we aimed to examine the transcriptome profiling of the neoplastic stromal cells of GCTB in this study. RNA-sequencing was performed on three GCTB stromal cell samples and one bone marrow-derived MSC sample and 174 differentially expressed genes (DEGs) were identified between these two cell types. The top five up-regulated genes are SPP1, F3, TSPAN12, MMP13, and LGALS3BP and further validated by qPCR and Western Blotting. Knockdown of SPP1 was found to induce RUNX2 and OPG expression in GCTB stromal cells but not the MSCs. Ingenuity pathway analysis (IPA) of the 174 DEGs revealed significant alternations in 23 pathways; variant calling analysis revealed 1915 somatic variants of 384 genes with high or moderate impacts. Interestingly, four canonical pathways were found overlapping in both analyses; from which VEGFA, CSF1, PLAUR, and F3 genes with somatic mutation were found up-regulated in GCTB stromal cells. The STRING diagram showed two main clusters of the DEGs; one cluster of histone genes that are down-regulated in GCTB samples and another related to osteoblast differentiation, angiogenesis, cell cycle progression, and tumor growth. The DEGs and somatic mutations found in our study warrant further investigation and validation, nevertheless, our study add new insights in the search for new therapeutic targets in treating GCTB. J. Cell. Biochem. 118: 1349-1360, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Bone Neoplasms/genetics , Gene Expression Profiling/methods , Giant Cell Tumor of Bone/genetics , Sequence Analysis, RNA/methods , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Mutation
9.
Oncol Rep ; 36(6): 3181-3187, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27779699

ABSTRACT

Giant cell tumor of bone (GCT) is the most commonly reported non-malignant bone tumor in Hong Kong. This kind of tumor usually affects people aged 20-40 years. Also, it is well known for recurrence locally, especially when the tumor cannot be removed completely. Filamins are actin-binding proteins which contain three family members, filamin A, B and C. They are the products of three different genes, FLNA, FLNB and FLNC, which can generate various transcript variants in different cell types. In this study, we focused on the effects of FLNBv2 and FLNBv4 toward GCT cells. The only difference between FLNBv2 and FLNBv4 is that FLNBv4 does not contain hinge 1 region. We found that the relative abundance of FLNBv4 varies among different GCT cell lines while the expression level of FLNBv4 in normal osteoblasts was only marginally detectable. In the functional aspect, overexpression of FLNBv4 led to upregulation of RANKL, OCN, OPG and RUNX2, which are closely related to GCT cell survival and differentiation. Moreover, FLNBv4 can have a negative effect on cell viability of GCT cells when compare with FLNBv2. In conclusion, splicing variants of FLNB are differentially expressed in GCT cells and may play a role in the proliferation and differentiation of tumor cells.


Subject(s)
Bone Neoplasms/metabolism , Filamins/genetics , Gene Expression Regulation, Neoplastic , Gene Expression , Giant Cell Tumor of Bone/metabolism , Adolescent , Adult , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Bone Neoplasms/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Female , Filamins/metabolism , Giant Cell Tumor of Bone/genetics , Humans , Male , Middle Aged , Protein Isoforms/genetics , Protein Isoforms/metabolism , Tumor Cells, Cultured , Young Adult
10.
Anesth Analg ; 122(5): 1567-77, 2016 May.
Article in English | MEDLINE | ID: mdl-27101500

ABSTRACT

BACKGROUND: Red blood cell (RBC) transfusion is common during infant cardiac surgery. A previous report of pediatric heart transplant recipients showed that increased RBC transfusion volume was independently associated with increased length of intensive care unit stay. It is unclear whether transfusion to infants as a subgroup carries similar risks. This study investigated relationships between intraoperative RBC transfusion during heart transplantation and postoperative length of stay (LOS), morbidity, and mortality in infants. METHODS: Retrospective analysis of medical records from infants <1 year old undergoing primary heart transplantation at Loma Linda University Medical Center from 1985 to 2012 was conducted. Exclusion criteria included preoperative exchange transfusion or extracorporeal membrane oxygenation. Data sought included patient characteristics; intraoperative RBC transfusion volume and cardiopulmonary bypass details; and postoperative vasoactive support, ventilator support, morbidity, LOS, and 30-day mortality. The relationship of RBC transfusion volume (mL/kg) to these postoperative variables was assessed by univariate analysis. Multiple regression analysis of postoperative LOS included variables that were independent predictors of LOS or associated with ≥10% change in the ß-estimate for RBC effect. RESULTS: Data from 307 infants showed that most (66.8%) had single-ventricle physiology. Median age at transplant was 50 days, weight 3.95 kg, and intraoperative transfusion volume 109 mL/kg. Transfusion volume was inversely related to age and weight. Median postoperative LOS was 18.2 days. Univariate linear regression analysis of transfused volume showed no relationship to log-transformed postoperative LOS (F(1,305) = 0.00; P = 0.960; R = 0.000; ß-coefficient = 0.004; 95% confidence interval = -0.1542 to 0.1623). Transfused volume was not related to 30-day mortality (difference -0.162; -0.048 to 0.371 mL/kg; P = 0.112) or to postoperative ventilator support (R = 0.047), but was greater in patients who required reoperation (difference -0.246; -0.494 to -0.025; P = 0.004). Multiple regression analysis for all patients revealed age, preoperative ventilator support, prolonged postoperative ventilatory or vasoactive support, transplant year, and 30-day mortality, but not major adverse events, to be significant confounding variables. Adjusting for these variables, transfused volume was not associated with prolonged postoperative LOS. CONCLUSIONS: In contrast to a prior report, we found no correlation between intraoperative RBC transfusion and postoperative LOS when studying only infants. Infants have maturing organ systems, less physiologic reserve, and increased surgical blood loss (evaluated as mL/kg) during cardiac surgery than their larger, older counterparts, distinguishing them from the general pediatric population. These differences require additional studies to determine the outcome impact of transfusion strategies in the infant subgroup.


Subject(s)
Blood Loss, Surgical/prevention & control , Erythrocyte Transfusion , Heart Defects, Congenital/surgery , Heart Transplantation , Intraoperative Care/methods , Academic Medical Centers , Age Factors , Blood Loss, Surgical/mortality , California , Erythrocyte Transfusion/adverse effects , Erythrocyte Transfusion/mortality , Female , Heart Defects, Congenital/diagnosis , Heart Defects, Congenital/mortality , Heart Transplantation/adverse effects , Heart Transplantation/mortality , Hospital Mortality , Humans , Infant , Infant Mortality , Infant, Newborn , Intraoperative Care/adverse effects , Intraoperative Care/mortality , Length of Stay , Linear Models , Male , Multivariate Analysis , Postoperative Complications/mortality , Postoperative Complications/therapy , Reoperation , Respiration, Artificial , Retrospective Studies , Risk Assessment , Risk Factors , Time Factors , Treatment Outcome
11.
Connect Tissue Res ; 56(6): 493-503, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26327464

ABSTRACT

A major barrier towards the study of the effects of drugs on Giant Cell Tumor of Bone (GCT) has been the lack of an animal model. In this study, we created an animal model in which GCT stromal cells survived and functioned as proliferating neoplastic cells. A proliferative cell line of GCT stromal cells was used to create a stable and luciferase-transduced cell line, Luc-G33. The cell line was characterized and was found that there were no significant differences on cell proliferation rate and recruitment of monocytes when compared with the wild type GCT stromal cells. We delivered the Luc-G33 cells either subcutaneously on the back or to the tibiae of the nude mice. The presence of viable Luc-G33 cells was assessed using real-time live imaging by the IVIS 200 bioluminescent imaging (BLI) system. The tumor cells initially propagated and remained viable on site for 7 weeks in the subcutaneous tumor model. We also tested in vivo antitumor effects of Zoledronate (ZOL) and Geranylgeranyl transferase-I inhibitor (GGTI-298) alone or their combinations in Luc-G33-transplanted nude mice. ZOL alone at 400 µg/kg and the co-treatment of ZOL at 400 µg/kg and GGTI-298 at 1.16 mg/kg reduced tumor cell viability in the model. Furthermore, the anti-tumor effects by ZOL, GGTI-298 and the co-treatment in subcutaneous tumor model were also confirmed by immunohistochemical (IHC) staining. In conclusion, we established a nude mice model of GCT stromal cells which allows non-invasive, real-time assessments of tumor development and testing the in vivo effects of different adjuvants for treating GCT.


Subject(s)
Benzamides/pharmacology , Bone Neoplasms , Diphosphonates/pharmacology , Giant Cell Tumor of Bone , Imidazoles/pharmacology , Neoplasms, Experimental , Animals , Bone Neoplasms/drug therapy , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Line, Tumor , Female , Giant Cell Tumor of Bone/drug therapy , Giant Cell Tumor of Bone/genetics , Giant Cell Tumor of Bone/metabolism , Giant Cell Tumor of Bone/pathology , Heterografts , Humans , Luciferases/biosynthesis , Luciferases/genetics , Male , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Stromal Cells/metabolism , Stromal Cells/pathology , Transduction, Genetic , Xenograft Model Antitumor Assays , Zoledronic Acid
12.
Int J Clin Pharmacol Ther ; 52(1): 27-38, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24161161

ABSTRACT

OBJECTIVES: The objective of this study was to evaluate the effect of a single intravenous dose of tocilizumab (TCZ) on pharmacokinetics (PK) of oral contraceptive (OC; norethindrone (NE) and ethinyl estradiol (EE)) and on sex hormone levels (progesterone (PG), luteinizing hormone (LH), and follicle-stimulating hormone (FSH)) in subjects with active rheumatoid arthritis (RA) who were on stable doses of methotrexate. METHODS: This was an open-label, nonrandomized, multicenter, two-parallel group, one-sequence crossover study. In Group 1, Cycle 1 was a baseline cycle to determine the PK of OC and levels of sex hormones. At the start of Cycle 2, patients continued to receive OC and single TCZ dosing on Day 1. In Cycle 2, we determined the PK of OC and levels of sex hormones when OC and TCZ were combined. In Cycle 3, we determined the PK of OC and the levels of sex hormones after TCZ treatment was stopped. PK for EE and NE were analyzed serially on Day 7 when maximum TCZ effect on inflammation as indicated by C-reactiv protein (CRP) was expected. Hormone levels (PG, LH and FSH) were measured mid-cycle (cycle Days 12 - 16 and Day 21) during each cycle. Group 2 (healthy subjects) was studied to compare the levels of OC PK exposures with those in each cycle of Group 1 (RA subjects). RESULTS: Levels of PG, LH and FSH were not affected by the combination of TCZ/OC treatment in RA patients studied. No breakthrough bleeding was attributed to the initiation of TCZ treatment in subjects receiving OCs. PK exposures of EE and NE were similar between RA and healthy subjects at baseline and were not affected by single-dose TCZ. Administration of OC with or without a single dose of TCZ was well tolerated. CONCLUSIONS: Data from this study indicated that the PK and sex hormone levels were not affected in RA subjects who had active disease and were on a stable regimen of methotrexate.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Arthritis, Rheumatoid/metabolism , Contraceptives, Oral/pharmacokinetics , Ethinyl Estradiol/pharmacokinetics , Norethindrone/pharmacokinetics , Adolescent , Adult , Antibodies, Monoclonal, Humanized/pharmacokinetics , C-Reactive Protein/analysis , Contraceptives, Oral/adverse effects , Cross-Over Studies , Drug Interactions , Female , Follicle Stimulating Hormone/blood , Humans , Luteinizing Hormone/blood , Progesterone/blood , Receptors, Interleukin-6/blood
13.
Cytotherapy ; 15(12): 1484-97, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24199592

ABSTRACT

BACKGROUND AIMS: Cancer is one of the greatest health challenges facing the world today with >10 million new cases of cancer every year. The self-renewal, tumor-homing ability and low immunogenicity of mesenchymal stromal cells (MSCs) make them potential delivery candidates for suicide genes for anti-tumor therapy. However, unstable supply and short life span of adult MSCs in vitro have limited this therapeutic potential. In this study, we aimed to evaluate if immortalization of human fetal bone marrow-derived mesenchymal stromal cells by simian virus 40 (SV40-hfBMSCs) could be a stable source of MSCs for clinical application of suicide gene therapy. METHODS AND RESULTS: Transduction of SV40 and herpes simplex virus thymidine kinase-IRES-green fluorescent protein (TK-GFP) did not cause significant change in the stem cell properties of hfBMSCs. The anti-tumor effect of SV40-TK-hfBMSCs in the presence of the prodrug ganciclovir was demonstrated in vitro and in nude mice bearing human prostate cancer cells, DU145 and PC3, which had been transduced with luciferase and GFP for imaging evaluation by an in vivo live imaging system (IVIS 200 imaging system; Caliper Life Sciences). Repeated injection of low doses (1 × 10(6) cells/kg) of SV40-TK-hfBMSCs was as effective as previously reported and did not cause observable harmful side effects in multiple organs. Mixed lymphocyte reaction showed that SV40-TK-hfBMSCs did not induce significant proliferation of lymphocytes isolated from healthy adults. CONCLUSIONS: Taken together, immortalized hfBMSCs represent a reliable and safe source of MSCs for further clinical translational study.


Subject(s)
Bone Marrow Cells/cytology , Genes, Transgenic, Suicide/genetics , Mesenchymal Stem Cells/cytology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/therapy , Animals , Cell Line, Tumor , Drug Delivery Systems , Fetus/cytology , Ganciclovir/administration & dosage , Gene Expression Regulation/drug effects , Genetic Therapy , Humans , Male , Mesenchymal Stem Cells/chemistry , Mice , Prostatic Neoplasms/pathology , Simian virus 40
14.
Connect Tissue Res ; 54(6): 439-49, 2013.
Article in English | MEDLINE | ID: mdl-24060052

ABSTRACT

Denosumab and Zoledronic acid (ZOL) are two antiresorptive drugs currently in use for treating osteoporosis. They have different mechanisms of action but both have been shown to delay the onset of skeletal-related events in patients with giant cell tumor of bone (GCT). However, the anti-tumor mechanisms of denosumab on the neoplastic GCT stromal cells remain unknown. In this study, we focused on the direct effects of denosumab on the neoplastic GCT stromal cells and compared with ZOL. The microscopic view demonstrated a reduced cell growth in ZOL-treated but not in denosumab-treated GCT stromal cells. ZOL was found to exhibit a dose-dependent inhibition in cell growth in all GCT stromal cell lines tested and cause apoptosis in two out of three cell lines. In contrast, denosumab only exerted a minimal inhibitory effect in one cell line and did not induce any apoptosis. ZOL significantly inhibited the mRNA expression of receptor activator of nuclear factor kappa-B ligand (RANKL) and osteoprotegerin (OPG) in two GCT stromal cell lines whereas their protein levels remained unchanged. On the contrary, denosumab did not regulate RANKL and OPG expression at both mRNA and protein levels. Moreover, the protein expression of Macrophage Colony-Stimulating Factor (M-CSF), Alkaline Phosphatase (ALP), and Collagen α1 Type I were not regulated by denosumab and ZOL either. Our findings provide new insights in the anti-tumor effect of denosumab on GCT stromal cells and raise a concern that tumor recurrence may occur after the withdrawal of the drug.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Diphosphonates/therapeutic use , Giant Cell Tumor of Bone/drug therapy , Giant Cell Tumor of Bone/pathology , Imidazoles/therapeutic use , Alkaline Phosphatase/metabolism , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Count , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Collagen Type I/genetics , Collagen Type I/metabolism , Denosumab , Diphosphonates/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Giant Cell Tumor of Bone/genetics , Humans , Imidazoles/pharmacology , Macrophage Colony-Stimulating Factor/metabolism , Osteoprotegerin/genetics , Osteoprotegerin/metabolism , RANK Ligand/genetics , RANK Ligand/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Solubility , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/pathology , Zoledronic Acid
15.
Int J Oncol ; 42(2): 437-43, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23229819

ABSTRACT

Giant cell tumor of bone (GCT) is a destructive neoplasm of uncertain etiology that affects the epiphyseal ends of long bones in young adults. GCT stromal cells (GCTSCs) are the primary neoplastic cells of this tumor and are the only proliferating cell component in long-term culture, which recruits osteoclast-like giant cells that eventually mediate bone destruction. The oncogenesis of GCT and factors driving the neoplastic stromal cells to proliferate extensively and pause at an early differentiation stage of pre-osteoblast lineage remain unknown. Overexpression of p63 was observed in GCTSCs and there is growing evidence that p63 is involved in oncogenesis through different mechanisms. This study aimed to understand the specific role of p63 in cell proliferation and oncogenesis of GCTSCs. We confirmed p63 expression in the mononuclear cells in GCT by immunohistochemical staining. By real-time PCR analysis, we showed a higher level (>15­fold) of TAp63 expression in GCTSCs compared to that in mesenchymal stem cells. Furthermore, we observed that knockdown of the p63 gene by siRNA transfection greatly reduced cell proliferation and induced cell cycle arrest at S phase in GCTSCs. We found that the mRNA expression of CDC2 and CDC25C was substantially suppressed by p63 knockdown at 24-72 h. Moreover, p63 was found to be recruited on the regulatory regions of CDC2 and CDC25C, which contain p53-responsive elements. In summary, our data suggest that p63 regulates GCTSC proliferation by binding to the CDC2 and CDC25C p53-REs, which may inhibit the p53 tumor suppressor activity and contribute to GCT tumorigenesis.


Subject(s)
Bone Neoplasms/genetics , Genes, cdc , Giant Cell Tumors/genetics , Membrane Proteins/biosynthesis , Bone Neoplasms/pathology , CDC2-CDC28 Kinases/biosynthesis , Carcinogenesis , Cell Differentiation , Cell Proliferation , Gene Expression Regulation, Neoplastic , Giant Cell Tumors/metabolism , Humans , Membrane Proteins/genetics , Stromal Cells/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , cdc25 Phosphatases/biosynthesis
16.
Biomed Mater ; 6(1): 015004, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21205994

ABSTRACT

Water-soluble phosphorylated chitosan (P-chitosan) and disodium (1 → 4)-2-deoxy-2-sulfoamino-ß-D-glucopyranuronan (S-chitosan) are two chemically modified chitosans. In this study, we found that P-chitosan significantly promotes cell proliferation of both human primary osteoblasts (OBs) and the OB like stromal cell component of the giant cell tumor of bone (GCTB) cells at the concentration from 125 to 1000 µg ml⁻¹ at all time points of 1, 3, 5 and 7 days after treatment. Further investigation of the osteogenic effect of the P-chitosan suggested that it regulates the levels of osteoclastogenic factors, receptor activator of nuclear factor kappa B ligand and osteoprotegerin expression. An interesting finding is that S-chitosan at lower concentration (100 µg ml⁻¹) stimulates cell proliferation while a higher dose (1000 µg ml⁻¹) of S-chitosan inhibits it. The inhibitory effect of S-chitosan on human primary GCT stromal cells was greater than that of OBs (p < 0.05). Taken together, our findings elucidated the osteogenic effect of P-chitosan and the varying effects of S-chitosan on the proliferation of human primary OBs and GCT stromal cells and provided us the rationale for the construction of novel bone repair biomaterials with the dual properties of bone induction and bone tumor inhibition.


Subject(s)
Chitosan/analogs & derivatives , Giant Cell Tumor of Bone/drug therapy , Osteoblasts/drug effects , Base Sequence , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Chitosan/chemistry , Chitosan/pharmacology , DNA Primers/genetics , Gene Expression/drug effects , Giant Cell Tumor of Bone/genetics , Giant Cell Tumor of Bone/metabolism , Giant Cell Tumor of Bone/pathology , Humans , Materials Testing , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis/drug effects , Osteoprotegerin/genetics , Osteoprotegerin/metabolism , Phosphorylation , RANK Ligand/genetics , RANK Ligand/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Solubility , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/pathology , Tumor Cells, Cultured , Water
17.
J Orthop Res ; 29(3): 403-13, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20886653

ABSTRACT

Giant cell tumor (GCT) is the most common nonmalignant primary bone tumor reported in Hong Kong. It usually affects young adults between the ages of 20 and 40. This tumor is well known for its potential to recur following treatment. To date no effective adjuvant therapy exists for GCT. Our project aimed to study the effects of pamidronate (PAM), farnesyl transferase inhibitor (FTI-277), geranylgeranyl transferase inhibitor (GGTI-298), and their combinations on GCT stromal cells (SC). Individual treatment with PAM, FTI-277, and GGTI-298, inhibited the cell viability and proliferation of GCT SC in a dose-dependent way. Combination of FTI-277 with GGTI-298 caused synergistic effects in reducing cell viability, and its combination index was 0.49, indicating a strong synergism. Moreover, the combination of FTI-277 with GGTI-298 synergistically enhanced cell apoptosis and activated caspase-3/7, -8, and -9 activities. PAM induced cell-cycle arrest at the S-phase. The combination of PAM with GGTI-298 significantly increased OPG/RANKL mRNA ratio and activated caspase-3/7 activity. Our findings support that the combination of bisphosphonates with GGTIs or FTIs with GGTIs may be used as potential adjuvants in the treatment of GCT of bone.


Subject(s)
Bone Neoplasms , Diphosphonates/pharmacology , Enzyme Inhibitors/pharmacology , Giant Cell Tumor of Bone , Osteoprotegerin/genetics , RANK Ligand/genetics , Alkyl and Aryl Transferases/antagonists & inhibitors , Apoptosis/drug effects , Benzamides/pharmacology , Bone Density Conservation Agents/pharmacology , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Bone Neoplasms/physiopathology , Caspases/metabolism , Cell Division/drug effects , Cell Survival/drug effects , Drug Synergism , Farnesyltranstransferase/antagonists & inhibitors , Gene Expression/drug effects , Giant Cell Tumor of Bone/drug therapy , Giant Cell Tumor of Bone/pathology , Giant Cell Tumor of Bone/physiopathology , Humans , Methionine/analogs & derivatives , Methionine/pharmacology , Pamidronate , Prenylation/drug effects , RNA, Messenger/metabolism , S Phase/drug effects , Tumor Cells, Cultured
18.
J Cell Biochem ; 110(2): 438-46, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20225273

ABSTRACT

Giant cell tumor (GCT) of bone is an aggressive non-cancerous tumor, which consists of multi-nucleated osteoclast-like giant cells, stromal cells, and monocytes. It is believed that stromal cells are the neoplastic component of this tumor. Expression of the receptor activator of nuclear factor kappa B ligand (RANKL) in the stromal cells stimulates the monocytes to form giant multi-nucleated osteoclast-like cells, causing bone over-resorption at the tumor site. Previously, our group has reported the up-regulation of RANKL in GCT of bone stromal cells, but the mechanism is unknown. Using stromal cell culture of GCT obtained from patients, we demonstrated the up-regulation of the transcriptional activator CCAAT/enhancer binding protein beta (C/EBPbeta). RANKL promoter studies revealed that C/EBPbeta over-expression induced RANKL promoter activity in a dose-dependent manner and a CCAAT-box within the region nt -357/-1 contributed to the basal transcription activity, with a possible C/EBPbeta binding element in the region nt -460/-358 leading to further induction. Furthermore, we also showed that C/EBPbeta bound to the RANKL promoter in GCT stromal cells in vivo by chromatin immunoprecipitation. To conclude, our study has shown that C/EBPbeta is a RANKL promoter activator in stromal cells of GCT of bone and we have proposed a model in which C/EBPbeta plays an important role in the osteolytic characteristics and pathological causes of GCT of bone.


Subject(s)
Bone Neoplasms/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , Gene Expression Regulation , Giant Cell Tumor of Bone/metabolism , RANK Ligand/genetics , Up-Regulation , Base Sequence , Bone Neoplasms/pathology , Chromatin Immunoprecipitation , DNA Primers , Giant Cell Tumor of Bone/pathology , Humans , Molecular Sequence Data , Polymerase Chain Reaction , Promoter Regions, Genetic , Stromal Cells/metabolism
19.
Am J Bot ; 92(3): 525-33, 2005 Mar.
Article in English | MEDLINE | ID: mdl-21652431

ABSTRACT

Bauhinia blakeana (Leguminosae subfam. Caesalpinioideae tribe Cercideae), or the Hong Kong Orchid Tree, is of great horticultural value. It is completely sterile and is shown here to be the result of hybridization between the largely sympatric species, B. purpurea and B. variegata. Although the analysis of patterns of morphological variation revealed only a few examples of phenotypic intermediacy, study of intersimple sequence repeat (ISSR) markers enabled unequivocal identification of the parental species due to the presence of additive inheritance of alleles and the absence of any bands that are unique to B. blakeana. Investigation of aspects of the reproductive biology of the taxa furthermore revealed that the parental species are largely xenogamous, have flowering periods that overlap seasonally and temporally, and share common pollinators. Evidence is provided to show that B. blakeana is not naturally stabilized and is only maintained horticulturally by artificial propagation. It is therefore recommended that the hybrid be regarded as a horticultural cultivar rather than a naturally occurring species; a new cultivar name, Bauhinia 'Blakeana', is accordingly validated.

20.
Biochem Biophys Res Commun ; 322(3): 827-35, 2004 Sep 24.
Article in English | MEDLINE | ID: mdl-15336538

ABSTRACT

Chronic hepatitis B virus (HBV) infection is one of the major causes of hepatocellular carcinoma. HBV encodes an oncogenic hepatitis B virus X protein (HBx), which can transactivate host cell transcriptional machinery and mediate cellular transformation. To disclose the early genetic response in HBx-mediated transformation process, we constructed a conditional HBx-expressing hepatocyte cell line, which allows us to compare the gene expression profiles under controllable HBx induction. A cDNA microarray containing more than 8700 mouse genes and ESTs was utilized to examine the gene expression profiles. We identified 260 candidate genes and 259 ESTs which have shown aberrant expression under HBx induction. Most of them are involved in signal transduction pathway, cell cycle control, metastasis, transcriptional regulation, immune response, and metabolism. These results provide additional insight into early cellular targets of HBx, which could give us a better understanding of the function of HBx and their progressive changes during HBx-mediated hepatocarcinogenesis.


Subject(s)
Carcinoma, Hepatocellular/genetics , DNA, Complementary/genetics , Gene Expression Profiling/methods , Liver Neoplasms/genetics , Oligonucleotide Array Sequence Analysis/methods , Trans-Activators/physiology , Base Sequence , Carcinoma, Hepatocellular/virology , Cell Division , Cell Line , Cell Transformation, Neoplastic , Cloning, Molecular , DNA Primers , Hepatitis B Antigens/physiology , Humans , Liver Neoplasms/virology , Recombinant Proteins/metabolism , Trans-Activators/genetics , Transcriptional Activation , Transfection , Viral Regulatory and Accessory Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...