Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 28
1.
Br J Cancer ; 128(9): 1753-1764, 2023 05.
Article En | MEDLINE | ID: mdl-36810912

BACKGROUND: Although trimodality therapy resecting tumours followed by chemoradiotherapy is emerged for muscle-invasive bladder cancer (MIBC), chemotherapy produces toxicities. Histone deacetylase inhibitors have been identified as an effective strategy to enhance cancer radiotherapy (RT). METHODS: We examined the role of HDAC6 and specific inhibition of HDAC6 on BC radiosensitivity by performing transcriptomic analysis and mechanism study. RESULTS: HDAC6 knockdown or HDAC6 inhibitor (HDAC6i) tubacin exerted a radiosensitizing effect, including decreased clonogenic survival, increased H3K9ac and α-tubulin acetylation, and accumulated γH2AX, which are similar to the effect of panobinostat, a pan-HDACi, on irradiated BC cells. Transcriptomics of shHDAC6-transduced T24 under irradiation showed that shHDAC6 counteracted RT-induced mRNA expression of CXCL1, SERPINE1, SDC1 and SDC2, which are linked to cell migration, angiogenesis and metastasis. Moreover, tubacin significantly suppressed RT-induced CXCL1 and radiation-enhanced invasion/migration, whereas panobinostat elevated RT-induced CXCL1 expression and invasion/migration abilities. This phenotype was significantly abrogated by anti-CXCL1 antibody, indicating the key regulator of CXCL1 contributing to BC malignancy. Immunohistochemical evaluation of tumours from urothelial carcinoma patients supported the correlation between high CXCL1 expression and reduced survival. CONCLUSION: Unlike pan-HDACi, the selective HDAC6i can enhance BC radiosensitization and effectively inhibit RT-induced oncogenic CXCL1-Snail-signalling, thus further advancing its therapeutic potential with RT.


Carcinoma, Transitional Cell , Histone Deacetylase 6 , Radiation Tolerance , Urinary Bladder Neoplasms , Humans , Acetylation , Cell Line, Tumor , Histone Deacetylase 6/genetics , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Panobinostat/pharmacology , Tubulin/metabolism , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/radiotherapy
2.
Clin Transl Sci ; 15(1): 255-266, 2022 01.
Article En | MEDLINE | ID: mdl-34423904

N-methyl-D-aspartate ionotropic glutamatergic receptor (NMDAR) modulators, including rapastinel and ketamine, elicit rapid and sustained antidepressant responses in patients with treatment-resistant major depressive disorder. This phase I, randomized, multicenter, placebo-controlled, five-period, crossover, single-dose study evaluated simulated driving performance of healthy participants (N = 107) after single doses of rapastinel slow intravenous (i.v.) bolus 900 and 1800 mg, alprazolam oral 0.75 mg (positive control), ketamine i.v. infusion 0.5 mg/kg (clinical comparator), and placebo ~ 45 min before driving. The primary end point was SD of lateral position (SDLP) during the 60-min 100-km simulated driving scenario. Additional measures of driving performance, sleepiness, and cognition were also evaluated. To assess effects over time, mean SDLP was calculated for each 10-min interval of driving. Sensitivity of the assays was confirmed with alprazolam (all placebo comparisons p < 0.02). Rapastinel 900 and 1800 mg did not significantly affect simulated driving performance compared to placebo (both p > 0.5). Both rapastinel doses resulted in significantly less impaired driving compared to alprazolam or ketamine (all p < 0.002); ketamine significantly impaired driving compared to placebo (p = 0.0001). Results for the additional measures were similar to the primary end point. No new safety signals were observed for any study interventions. This first study of rapastinel effects on simulated driving found that rapastinel 900 and 1800 mg did not impair driving performance, but ketamine 0.5 mg/kg resulted in significantly impaired driving performance. Ketamine's effects on driving were maintained for at least 105 min, indicating that clinicians should be vigilant to prevent or postpone driving in patients after ketamine treatment.


Alprazolam/administration & dosage , Analgesics/administration & dosage , Anti-Anxiety Agents/administration & dosage , Antidepressive Agents/administration & dosage , Automobile Driving , Ketamine/administration & dosage , Oligopeptides/administration & dosage , Adult , Aged , Female , Humans , Male , Middle Aged , Young Adult
3.
Theranostics ; 11(19): 9667-9686, 2021.
Article En | MEDLINE | ID: mdl-34646392

The tumorous niche may drive the plasticity of heterogeneity and cancer stemness, leading to drug resistance and metastasis, which is the main reason of treatment failure in most cancer patients. The aim of this study was to establish a tumor microenvironment (TME)-based screening to identify drugs that can specifically target cancer stem cells (CSCs) and cancer-associated fibroblasts (CAFs) in the TME. Methods: Lung cancer patient-derived cancer cell and CAFs were utilized to mimic the TME and reproduce the stemness properties of CSCs in vitro and develop a high-throughput drug screening platform with phenotypical parameters. Limiting dilution assay, sphere-forming and ALDH activity assay were utilized to measure the cancer stemness characteristics. In vivo patient-derived xenograft (PDX) models and single-cell RNA sequencing were used to evaluate the mechanisms of the compounds in CSCs and CAFs. Results: The TME-based drug screening platform could comprehensively evaluate the response of cancer cells, CSCs and CAFs to different treatments. Among the 1,524 compounds tested, several drugs were identified to have anti-CAFs, anticancer and anti-CSCs activities. Aloe-emodin and digoxin both show anticancer and anti-CSCs activity in vitro and in vivo, which was further confirmed in the lung cancer PDX model. The combination of digoxin and chemotherapy improved therapeutic efficacy. The single-cell transcriptomics analysis revealed that digoxin could suppress the CSCs subpopulation in CAFs-cocultured cancer cells and cytokine production in CAFs. Conclusions: The TME-based drug screening platform provides a tool to identify and repurpose compounds targeting cancer cells, CSCs and CAFs, which may accelerate drug development and therapeutic application for lung cancer patients.


Drug Repositioning/methods , Neoplastic Stem Cells/drug effects , Tumor Microenvironment/physiology , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Cell Proliferation , Drug Evaluation, Preclinical , Drug Screening Assays, Antitumor/methods , Early Detection of Cancer , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Lung Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Pharmaceutical Preparations
4.
J Clin Invest ; 131(16)2021 08 16.
Article En | MEDLINE | ID: mdl-34228648

Unlike the better-studied aberrant epigenome in the tumor, the clinicopathologic impact of DNA methylation in the tumor microenvironment (TME), especially the contribution from cancer-associated fibroblasts (CAFs), remains elusive. CAFs exhibit profound patient-to-patient tumorigenic heterogeneity. We asked whether such heterogeneity may be exploited to quantify the level of TME malignancy. We developed a robust and efficient methylome/transcriptome co-analytical system for CAFs and paired normal fibroblasts (NFs) from non-small-cell lung cancer patients. We found 14,781 CpG sites of CAF/NF differential methylation, of which 3,707 sites showed higher methylation changes in ever-smokers than in nonsmokers. Concomitant CAF/NF differential gene expression analysis pointed to a subset of 54 smoking-associated CpG sites with strong methylation-regulated gene expression. A methylation index that summarizes the ß values of these CpGs was built for NF/CAF discrimination (MIND) with high sensitivity and specificity. The potential of MIND in detecting premalignancy across individual patients was shown. MIND succeeded in predicting tumor recurrence in multiple lung cancer cohorts without reliance on patient survival data, suggesting that the malignancy level of TME may be effectively graded by this index. Precision TME grading may provide additional pathological information to guide cancer prognosis and open up more options in personalized medicine.


Cancer-Associated Fibroblasts/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Epigenome , Lung Neoplasms/genetics , Smoking/adverse effects , Transcriptome , Adult , Aged , Aged, 80 and over , Cancer-Associated Fibroblasts/pathology , Carcinoma, Non-Small-Cell Lung/pathology , CpG Islands , DNA Methylation , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Prognosis , Smoking/genetics , Smoking/metabolism , Tumor Cells, Cultured , Tumor Microenvironment/genetics
5.
Article En | MEDLINE | ID: mdl-34036228

PURPOSE: Epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs) show efficacy in treating patients with lung adenocarcinoma with EGFR-activating mutations. However, a significant subset of targeted patients fail to respond. Unlike acquired resistance (AR), intrinsic resistance (IR) remains poorly understood. We investigated whether epigenomic factors contribute to patient-to-patient heterogeneity in the EGFR-TKI response and aimed to characterize the IR subpopulation that obtains no benefit from EGFR-TKIs. PATIENTS AND METHODS: We conducted genome-wide DNA methylation profiling of 79 tumors sampled from patients with advanced lung adenocarcinoma before they received EGFR-TKI treatment and analyzed the patient responses. Pyrosequencing was performed in a validation cohort of 163 patients with EGFR-activating mutations. RESULTS: A DNA methylation landscape of 216 CpG sites with differential methylation was established to elucidate the association of DNA methylation with the characteristics and EGFR-TKI response status of the patients. Functional analysis of 37 transcription-repressive sites identified the enrichment of transcription factors, notably homeobox (HOX) genes. DNA methylation of HOXB9 (cg13643585) in the enhancer region yielded 88% sensitivity for predicting drug response (odds ratio [OR], 6.64; 95% CI, 1.98 to 25.23; P = .0009). Pyrosequencing validated that HOXB9 gained methylation in patients with a poor EGFR-TKI response (OR, 3.06; 95% CI, 1.13 to 8.19; P = .019). CONCLUSION: Our data suggest that homeobox DNA methylation could be a novel tumor cellular state that can aid the precise categorization of tumor heterogeneity in the study of IR to EGFR-TKIs. We identified, for the first time, an epigenomic factor that can potentially complement DNA mutation status in discriminating patients with lung adenocarcinoma who are less likely to benefit from EGFR-TKI treatment, thereby leading to improved patient management in precision medicine.


Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/genetics , DNA Methylation , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic , Genome-Wide Association Study , Homeodomain Proteins/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Protein Kinase Inhibitors/therapeutic use , Adult , Aged , ErbB Receptors/genetics , Female , Humans , Male , Middle Aged
6.
Cancer Immunol Immunother ; 70(5): 1435-1450, 2021 May.
Article En | MEDLINE | ID: mdl-33175182

BACKGROUND: Malignant pleural effusion (MPE)-macrophage (Mφ) of lung cancer patients within unique M1/M2 spectrum showed plasticity in M1-M2 transition. The M1/M2 features of MPE-Mφ and their significance to patient outcomes need to be clarified; furthermore, whether M1-repolarization could benefit treatment remains unclear. METHODS: Total 147 stage-IV lung adenocarcinoma patients undergoing MPE drainage were enrolled for profiling and validation of their M1/M2 spectrum. In addition, the MPE-Mφ signature on overall patient survival was analyzed. The impact of the M1-polarization strategy of patient-derived MPE-Mφ on anti-cancer activity was examined. RESULTS: We found that MPE-Mφ expressed both traditional M1 (HLA-DRA) and M2 (CD163) markers and showed a wide range of M1/M2 spectrum. Most of the MPE-Mφ displayed diverse PD-L1 expression patterns, while the low PD-L1 expression group was correlated with higher levels of IL-10. Among these markers, we identified a novel two-gene MPE-Mφ signature, IL-1ß and TGF-ß1, representing the M1/M2 tendency, which showed a strong predictive power in patient outcomes in our MPE-Mφ patient cohort (N = 60, p = 0.013) and The Cancer Genome Atlas Lung Adenocarcinoma dataset (N = 478, p < 0.0001). Significantly, ß-glucan worked synergistically with IFN-γ to reverse the risk signature by repolarizing the MPE-Mφ toward the M1 pattern, enhancing anti-cancer activity. CONCLUSIONS: We identified MPE-Mφ on the M1/M2 spectrum and plasticity and described a two-gene M1/M2 signature that could predict the outcome of late-stage lung cancer patients. In addition, we found that "re-education" of these MPE-Mφ toward anti-cancer M1 macrophages using clinically applicable strategies may overcome tumor immune escape and benefit anti-cancer therapies.


Lung Neoplasms/immunology , Macrophages/physiology , Pleural Effusion, Malignant/immunology , Biomarkers, Tumor/metabolism , Cell Differentiation , Cell Plasticity , Cells, Cultured , Gene Expression Regulation, Neoplastic , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Neoplasm Staging , Th1 Cells/immunology , Th2 Cells/immunology , Transcriptome , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
8.
Cancers (Basel) ; 12(3)2020 Mar 23.
Article En | MEDLINE | ID: mdl-32210009

Risk factors including genetic effects are still being investigated in lung adenocarcinoma (LUAD). Mitochondria play an important role in controlling imperative cellular parameters, and anomalies in mitochondrial function might be crucial for cancer development. The mitochondrial genomic aberrations found in lung adenocarcinoma and their associations with cancer development and progression are not yet clearly characterized. Here, we identified a spectrum of mitochondrial genome mutations in early-stage lung adenocarcinoma and explored their association with prognosis and clinical outcomes. Next-generation sequencing was used to reveal the mitochondrial genomes of tumor and conditionally normal adjacent tissues from 61 Stage 1 LUADs. Mitochondrial somatic mutations and clinical outcomes including relapse-free survival (RFS) were analyzed. Patients with somatic mutations in the D-loop region had longer RFS (adjusted hazard ratio, adjHR = 0.18, p = 0.027), whereas somatic mutations in mitochondrial Complex IV and Complex V genes were associated with shorter RFS (adjHR = 3.69, p = 0.012, and adjHR = 6.63, p = 0.002, respectively). The risk scores derived from mitochondrial somatic mutations were predictive of RFS (adjHR = 9.10, 95%CI: 2.93-28.32, p < 0.001). Our findings demonstrated the vulnerability of the mitochondrial genome to mutations and the potential prediction ability of somatic mutations. This research may contribute to improving molecular guidance for patient treatment in precision medicine.

9.
Article En | MEDLINE | ID: mdl-31501147

Dalbavancin is a lipoglycopeptide antibiotic with a prolonged half-life. A phase 1 study assessed dalbavancin levels in epithelial lining fluid (ELF) in 35 healthy adults using ELF bronchial microsampling up to 168 h after administration of 1,500 mg dalbavancin. The penetration of dalbavancin into ELF was 36%. ELF levels of dalbavancin exceeded the MIC90s of Streptococcus pneumoniae and Staphylococcus aureus for ≥7 days.


Anti-Bacterial Agents/pharmacokinetics , Respiratory Mucosa/chemistry , Teicoplanin/analogs & derivatives , Adult , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/analysis , Anti-Bacterial Agents/blood , Humans , Infusions, Intravenous , Male , Microbial Sensitivity Tests , Middle Aged , Pneumonia, Bacterial/drug therapy , Pneumonia, Pneumococcal/drug therapy , Pneumonia, Staphylococcal/drug therapy , Staphylococcus aureus/drug effects , Streptococcus pneumoniae/drug effects , Teicoplanin/administration & dosage , Teicoplanin/analysis , Teicoplanin/blood , Teicoplanin/pharmacokinetics , Young Adult
10.
PLoS One ; 12(8): e0183507, 2017.
Article En | MEDLINE | ID: mdl-28829816

The type 2 transmembrane serine protease matriptase is involved in many pathophysiological processes probably via its enzymatic activity, which depends on the dynamic relationship between zymogen activation and protease inhibition. Matriptase shedding can prolong the life of enzymatically active matriptase and increase accessibility to substrates. We show here that matriptase shedding occurs via a de novo proteolytic cleavage at sites located between the SEA domain and the CUB domain. Point or combined mutations at the four positively charged amino acid residues in the region following the SEA domain allowed Arg-186 to be identified as the primary cleavage site responsible for matriptase shedding. Kinetic studies further demonstrate that matriptase shedding is temporally coupled with matriptase zymogen activation. The onset of matriptase shedding lags one minute behind matriptase zymogen activation. Studies with active site triad Ser-805 point mutated matriptase, which no longer undergoes zymogen activation or shedding, further suggests that matriptase shedding depends on matriptase zymogen activation, and that matriptase proteolytic activity may be involved in its own shedding. Our studies uncover an autonomous mechanism coupling matriptase zymogen activation, proteolytic activity, and shedding such that a proportion of newly generated active matriptase escapes HAI-1-mediated rapid inhibition by shedding into the extracellular milieu.


Enzyme Precursors/metabolism , Serine Endopeptidases/metabolism , Amino Acid Sequence , Antibodies, Monoclonal/immunology , Cell Line, Tumor , Enzyme Activation , Humans , Point Mutation , Proteolysis , Sequence Homology, Amino Acid , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Serine Endopeptidases/immunology
11.
Arterioscler Thromb Vasc Biol ; 37(7): 1402-1414, 2017 07.
Article En | MEDLINE | ID: mdl-28522697

OBJECTIVE: Vascular calcification (VC) is a major cause of mortality in patients with end-stage renal diseases. Biomarkers to predict the progression of VC early are in urgent demand. APPROACH AND RESULTS: We identified circulating, cell-free microRNAs as potential biomarkers using in vitro VC models in which both rat and human aortic vascular smooth muscle cells were treated with high levels of phosphate to mimic uremic hyperphosphatemia. Using an Affymetrix microRNA array, we found that miR-125b and miR-382 expression levels declined significantly as biomineralization progressed, but this decline was only observed for miR-125b in the culture medium. A time-dependent decrease in aortic tissue and serum miR-125b levels was also found in both ex vivo and in vivo renal failure models. We examined the levels of circulating, cell-free miR-125b in sera from patients with end-stage renal diseases (n=88) and found an inverse association between the severity of VC and the circulating miR-125b level, irrespective of age or mineral-related hormones (odds ratio, 0.71; P=0.03). Furthermore, serum miR-125b levels on enrollment can predict VC progression years later (for high versus low, odds ratio, 0.14; P<0.01; for the highest versus lowest tertile and middle versus lowest tertile, odds ratio, 0.55 and 0.13; P=0.3 and <0.01, respectively). The uremic VC prediction efficacy using circulating miR-125b levels was also observed in an independent cohort (n=135). CONCLUSIONS: The results suggest that serum miR-125b levels are associated with VC severity and serve as a novel predictive marker for the risk of uremia-associated calcification progression.


Aortic Diseases/etiology , MicroRNAs/blood , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Uremia/etiology , Vascular Calcification/etiology , Aged , Aged, 80 and over , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Aortic Diseases/blood , Aortic Diseases/genetics , Aortic Diseases/pathology , Apoptosis , Cells, Cultured , Chi-Square Distribution , Disease Models, Animal , Disease Progression , Down-Regulation , Female , Genetic Markers , Humans , Hyperphosphatemia/blood , Hyperphosphatemia/etiology , Hyperphosphatemia/genetics , Kaplan-Meier Estimate , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/etiology , Kidney Failure, Chronic/genetics , Logistic Models , Male , MicroRNAs/genetics , Middle Aged , Multivariate Analysis , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Odds Ratio , Predictive Value of Tests , Rats, Sprague-Dawley , Risk Factors , Severity of Illness Index , Time Factors , Transfection , Uremia/blood , Uremia/complications , Uremia/genetics , Vascular Calcification/blood , Vascular Calcification/genetics , Vascular Calcification/pathology
12.
Clin Cancer Res ; 20(7): 1978-89, 2014 Apr 01.
Article En | MEDLINE | ID: mdl-24691641

PURPOSE: The high risk of recurrence after transurethral resection of bladder tumor of nonmuscle invasive disease requires lifelong treatment and surveillance. Changes in DNA methylation are chemically stable, occur early during tumorigenesis, and can be quantified in bladder tumors and in cells shed into the urine. Some urine markers have been used to help detect bladder tumors; however, their use in longitudinal tumor recurrence surveillance has yet to be established. EXPERIMENTAL DESIGN: We analyzed the DNA methylation levels of six markers in 368 urine sediment samples serially collected from 90 patients with noninvasive urothelial carcinoma (Tis, Ta, T1; grade low-high). The optimum marker combination was identified using logistic regression with 5-fold cross-validation, and validated in separate samples. RESULTS: A panel of three markers discriminated between patients with and without recurrence with the area under the curve of 0.90 [95% confidence interval (CI), 0.86-0.92] and 0.95 (95% CI, 0.90-1.00), sensitivity and specificity of 86%/89% (95% CI, 74%-99% and 81%-97%) and 80%/97% (95% CI, 60%-96% and 91%-100%) in the testing and validation sets, respectively. The three-marker DNA methylation test reliably predicted tumor recurrence in 80% of patients superior to cytology (35%) and cystoscopy (15%) while accurately forecasting no recurrence in 74% of patients that scored negative in the test. CONCLUSIONS: Given their superior sensitivity and specificity in urine sediments, a combination of hyper- and hypomethylated markers may help avoid unnecessary invasive exams and reveal the importance of DNA methylation in bladder tumorigenesis.


Biomarkers, Tumor/urine , DNA Methylation/genetics , Neoplasm Recurrence, Local/diagnosis , Urinary Bladder Neoplasms/urine , Adult , Aged , Carcinogenesis , Female , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/urine , Neoplasm Staging , Prognosis , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology
13.
Genome Res ; 24(4): 545-53, 2014 Apr.
Article En | MEDLINE | ID: mdl-24515120

Extracellular cues play critical roles in the establishment of the epigenome during development and may also contribute to epigenetic perturbations found in disease states. The direct role of the local tissue environment on the post-development human epigenome, however, remains unclear due to limitations in studies of human subjects. Here, we use an isogenic human ileal neobladder surgical model and compare global DNA methylation levels of intestinal epithelial cells pre- and post-neobladder construction using the Infinium HumanMethylation450 BeadChip. Our study is the first to quantify the effect of environmental cues on the human epigenome and show that the local tissue environment directly modulates DNA methylation patterns in normal differentiated cells in vivo. In the neobladder, the intestinal epithelial cells lose their tissue-specific epigenetic landscape in a time-dependent manner following the tissue's exposure to a bladder environment. We find that de novo methylation of many intestine-specific enhancers occurs at the rate of 0.41% per month (P < 0.01, Pearson = 0.71), while demethylation of primarily non-intestine-specific transcribed regions occurs at the rate of -0.37% per month (P < 0.01, Pearson = -0.57). The dynamic resetting of the DNA methylome in the neobladder not only implicates local environmental cues in the shaping and maintenance of the epigenome but also illustrates an unexpected cross-talk between the epigenome and the cellular environment.


Cell Differentiation/genetics , DNA Methylation/genetics , Epigenesis, Genetic , Intestines/growth & development , Aged , CpG Islands , Genome, Human , Humans , Intestines/surgery , Intestines/transplantation , Middle Aged , Tissue Transplantation
14.
Cancer Cell ; 21(5): 655-667, 2012 May 15.
Article En | MEDLINE | ID: mdl-22624715

Cancer cells typically exhibit aberrant DNA methylation patterns that can drive malignant transformation. Whether cancer cells are dependent on these abnormal epigenetic modifications remains elusive. We used experimental and bioinformatic approaches to unveil genomic regions that require DNA methylation for survival of cancer cells. First, we surveyed the residual DNA methylation profiles in cancer cells with highly impaired DNA methyltransferases. Then, we clustered these profiles according to their DNA methylation status in primary normal and tumor tissues. Finally, we used gene expression meta-analysis to identify regions that are dependent on DNA methylation-mediated gene silencing. We further showed experimentally that these genes must be silenced by DNA methylation for cancer cell survival, suggesting these are key epigenetic events associated with tumorigenesis.


Cell Transformation, Neoplastic/genetics , DNA Methylation , Epigenesis, Genetic , Neoplasms/genetics , Cell Death , Cell Survival , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cluster Analysis , Computational Biology , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , HCT116 Cells , Humans , Neoplasms/enzymology , Neoplasms/pathology , Phenotype , RNA Interference , Reproducibility of Results , Time Factors , Transfection
15.
Cancer Res ; 68(18): 7428-38, 2008 Sep 15.
Article En | MEDLINE | ID: mdl-18794131

Curcumin (diferuloylmethane) is an active component of the spice turmeric and has a diversity of antitumor activities. In this study, we found that curcumin can inhibit cancer cell invasion and metastasis through activation of the tumor suppressor DnaJ-like heat shock protein 40 (HLJ1). Human lung adenocarcinoma cells (CL1-5) treated with curcumin (1-20 mumol/L) showed a concentration-dependent reduction in cell migration, invasion, and metastatic ability, and this was associated with increased HLJ1 expression. Knockdown of HLJ1 expression by siRNA was able to reverse the curcumin-induced anti-invasive and antimetastasis effects in vitro and in vivo. The HLJ1 promoter and enhancer in a luciferase reporter assay revealed that curcumin transcriptionally up-regulates HLJ1 expression through an activator protein (AP-1) site within the HLJ1 enhancer. JunD, one of the AP-1 components, was significantly up-regulated by curcumin (1-20 mumol/L) in a concentration- and time-dependent manner. Knockdown of JunD expression could partially reduce the curcumin-induced HLJ1 activation and diminish the anti-invasive effect of curcumin, indicating that JunD would seem to be involved in curcumin-induced HLJ1 expression. Curcumin was able to induce c-Jun NH(2)-kinase (JNK) phosphorylation, whereas the JNK inhibitor (SP-600125) could attenuate curcumin-induced JunD and HLJ1 expression. Activation of HLJ1 by curcumin further leads to up-regulation of E-cadherin and a suppression of cancer cell invasion. Our results show that curcumin induces HLJ1, through activation of the JNK/JunD pathway, and inhibits lung cancer cell invasion and metastasis by modulating E-cadherin expression. This is a novel mechanism and supports the application of curcumin in anti-cancer metastasis therapy.


Adenocarcinoma/drug therapy , Curcumin/pharmacology , HSP40 Heat-Shock Proteins/biosynthesis , Lung Neoplasms/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Cadherins/biosynthesis , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , HSP40 Heat-Shock Proteins/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , MAP Kinase Kinase 4/metabolism , Mice , Mice, SCID , Neoplasm Invasiveness , Neoplasm Metastasis , Proto-Oncogene Proteins c-jun/metabolism , Random Allocation , Signal Transduction , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transfection , Up-Regulation
16.
J Clin Pharmacol ; 48(1): 108-16, 2008 Jan.
Article En | MEDLINE | ID: mdl-18094223

The systemic exposure of fluticasone propionate with hydrofluoroalkane propellant compared with chlorofluoro-carbon propellant and the effect of fluticasone propionate hydrofluoroalkane on 24-hour urinary cortisol in children aged 4 to 11 years with asthma were evaluated. Study 1 was an open-label, 2-way crossover study in which 16 subjects were randomized to 7.5 days each of fluticasone propionate hydrofluoroalkane 88 mug twice a day or fluticasone propionate chlorofluorocarbon 88 mug twice a day. In study 2, 63 subjects received 13.5 days of placebo followed by 27.5 days of fluticasone propionate hydrofluoroalkane 88 mug twice a day. The main outcome measure for study 1 was the difference between fluticasone propionate hydrofluoroalkane and fluticasone propionate chlorofluorocarbon in fluticasone propionate AUC(last) (area under the plasma fluticasone propionate concentration-time curve from zero up to the last quantifiable plasma concentration), and for study 2, 24-hour overnight urinary cortisol excretion. In study 1, fluticasone propionate systemic exposure was significantly lower (55%) with hydrofluoroalkane metered dose inhaler compared with chlorofluorocarbon metered dose inhaler. Study 2 showed no statistically significant changes in 24-hour overnight urinary cortisol excretion and no relationship to fluticasone propionate systemic exposure at this dose. The results of these 2 studies showed that in children aged 4 to 11 years with asthma, fluticasone propionate hydrofluoroalkane has lower systemic exposure compared with chlorofluorocarbon and no hypothalamic-pituitary-adrenal axis effects as measured by 24-hour urinary cortisol excretion.


Androstadienes/therapeutic use , Asthma/drug therapy , Hydrocarbons, Fluorinated/chemistry , Hydrocortisone/urine , Administration, Inhalation , Aerosol Propellants/chemistry , Androstadienes/blood , Androstadienes/pharmacokinetics , Anti-Asthmatic Agents/administration & dosage , Anti-Asthmatic Agents/adverse effects , Anti-Asthmatic Agents/chemistry , Area Under Curve , Asthma/metabolism , Child , Child, Preschool , Chlorofluorocarbons/chemistry , Cough/chemically induced , Cross-Over Studies , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Fever/chemically induced , Fluticasone , Half-Life , Headache/chemically induced , Humans , Hydrocortisone/analogs & derivatives , Male , Metered Dose Inhalers , Nausea/chemically induced , Respiratory Tract Infections/chemically induced
17.
Front Biosci ; 13: 621-35, 2008 Jan 01.
Article En | MEDLINE | ID: mdl-17981575

Matriptase is a member of an expanding group of type II transmembrane serine proteases. Recently, much has been learned about the biochemistry, cellular biology, normal tissue physiology, and human pathology of this protease, and of its inhibitor, termed the hepatocyte growth factor inhibitor-1 (HAI-1). This review examines the recent literature that has characterized the regulation of matriptase and HAI-1 with an emphasis on the molecular mechanisms governing its zymogen activation, inhibition by HAI-1, and ectodomain shedding.


Proteinase Inhibitory Proteins, Secretory/chemistry , Serine Endopeptidases/chemistry , Amino Acid Motifs , Animals , Antibodies, Monoclonal/chemistry , Cytosol/metabolism , Gene Expression Regulation, Enzymologic , Humans , Lysophospholipids/chemistry , Membrane Glycoproteins/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Protein Conformation , Proteinase Inhibitory Proteins, Secretory/metabolism , Suramin/chemistry
18.
FASEB J ; 20(13): 2393-5, 2006 Nov.
Article En | MEDLINE | ID: mdl-17023518

Titanium dioxide nanoparticles (nanoTiO2) have been widely used as a photocatalyst in air and water cleaning. However, these nanoparticles inhalation can induce pulmonary toxicity and its mechanism is not fully understood. In this study we investigated the pulmonary toxicity of nanoTiO2 and its molecular pathogenesis. The adult male ICR mice were exposed to intratracheal single dose of 0.1 or 0.5 mg nanoTiO2 (19-21 nm) and lung tissues were collected at 3rd day, 1st wk, and 2nd wk for morphometric, microarray gene expression, and pathway analyses. NanoTiO2 can induce pulmonary emphysema, macrophages accumulation, extensive disruption of alveolar septa, type II pneumocyte hyperplasia, and epithelial cell apoptosis. NanoTiO2 induced differential expression of hundreds of genes include activation of pathways involved in cell cycle, apoptosis, chemokines, and complement cascades. In particular, nanoTiO2 up-regulates placenta growth factor (PlGF) and other chemokines (CXCL1, CXCL5, and CCL3) expressions that may cause pulmonary emphysema and alveolar epithelial cell apoptosis. Cultured human THP-1 cell-derived macrophages treated with nanoTiO2 in vitro also resulted in up-regulations of PlGF, CXCL1, CXCL5, and CCL3. These results indicated that nanoTiO2 can induce severe pulmonary emphysema, which may be caused by activation of PlGF and related inflammatory pathways.


Pulmonary Emphysema/chemically induced , Pulmonary Emphysema/pathology , Titanium/toxicity , Animals , Cell Differentiation/drug effects , Cell Line , Cell Survival/drug effects , Humans , Macrophages/cytology , Macrophages/drug effects , Male , Mice , Mice, Inbred ICR , Monocytes/cytology , Monocytes/drug effects , Nanoparticles , Nanotechnology
19.
Psychiatry Clin Neurosci ; 58(2): 168-72, 2004 Apr.
Article En | MEDLINE | ID: mdl-15009822

The purpose of the present paper was to evaluate the relationship between plasma antipsychotics concentration and cognitive task performance. This may provide valuable information for rational dosage titration. Literature on the relationship between plasma risperidone (RIS) concentration and performance on the Continuous Performance Test (CPT) remains scarce. Ten patients (four male, six female) were given RIS for more than 1 year. Steady-state plasma concentrations of the parent drug RIS and its active metabolite, 9-hydroxy-risperidone (9-OH-RIS), were measured using specific liquid chromatography-tandem mass spectrometry assay. Psychopathology, side-effects of extrapyramidal symptoms (EPS) and CPT were also assessed. A negative correlation was found between CPT performance and the plasma RIS, 9-OH-RIS and its active moiety (RIS + 9-OH-RIS) concentrations. Both RIS and 9-OH-RIS have an impact on the CPT performance of schizophrenic patients. Optimal active moiety plasma concentration for best cognitive performance needs further study.


Antipsychotic Agents/administration & dosage , Antipsychotic Agents/pharmacokinetics , Attention/drug effects , Neuropsychological Tests , Psychomotor Performance/drug effects , Risperidone/administration & dosage , Risperidone/pharmacokinetics , Schizophrenia/drug therapy , Schizophrenic Psychology , Adult , Brief Psychiatric Rating Scale/statistics & numerical data , Discrimination Learning/drug effects , Dose-Response Relationship, Drug , Female , Humans , Isoxazoles/pharmacokinetics , Male , Neurologic Examination/drug effects , Paliperidone Palmitate , Pattern Recognition, Visual/drug effects , Psychometrics/statistics & numerical data , Pyrimidines/pharmacokinetics , Reaction Time/drug effects , Statistics as Topic , Treatment Outcome
20.
Int J Pharm ; 260(1): 39-46, 2003 Jul 09.
Article En | MEDLINE | ID: mdl-12818808

The aim of this study was to investigate the effect of formulation on the pharmacokinetics of diclofenac in two sustained release formulations (formulation A and Voltaren SR) after oral delivery. The dissolution of diclofenac from sustained release formulation was pH-dependent. While drug released from both formulations increased with increased pH, the release kinetics of these two formulations was different. The pharmacokinetic study was conducted in 12 healthy subjects administered with multiple doses of 100mg of diclofenac in a crossover design. There was a significant difference in area under the plasma concentration-time curve [AUC(0-24)] and C(max) observed. The formulation with a reduced diffusion exponent with increased kinetic constant results in increased absorption of diclofenac in vivo. This study demonstrated the impact of release mechanism of the formulation on the absorption in vivo.


Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Diclofenac/pharmacokinetics , Administration, Oral , Adult , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/blood , Area Under Curve , Chromatography, High Pressure Liquid , Cross-Over Studies , Delayed-Action Preparations , Diclofenac/administration & dosage , Diclofenac/blood , Humans , Hydrogen-Ion Concentration , Male , Solubility
...