Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Korean J Physiol Pharmacol ; 28(1): 83-91, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38154967

ABSTRACT

Hypoxia-inducible factor-1 alpha (HIF-1α) is a transcription factor activated under hypoxic conditions, and it plays a crucial role in cellular stress regulation. While HIF-1α activity is essential in normal tissues, its presence in the tumor microenvironment represents a significant risk factor as it can induce angiogenesis and confer resistance to anti-cancer drugs, thereby contributing to poor prognoses. Typically, HIF-1α undergoes rapid degradation in normoxic conditions via oxygen-dependent degradation mechanisms. However, certain cancer cells can express HIF-1α even under normoxia. In this study, we observed an inclination toward increased normoxic HIF-1α expression in cancer cell lines exhibiting increased HDAC6 expression, which prompted the hypothesis that HDAC6 may modulate HIF-1α stability in normoxic conditions. To prove this hypothesis, several cancer cells with relatively higher HIF-1α levels under normoxic conditions were treated with ACY-241, a selective HDAC6 inhibitor, and small interfering RNAs for HDAC6 knockdown. Our data revealed a significant reduction in HIF-1α expression upon HDAC6 inhibition. Moreover, the downregulation of HIF-1α under normoxic conditions decreased zinc finger E-box-binding homeobox 1 expression and increased E-cadherin levels in lung cancer H1975 cells, consequently suppressing cell invasion and migration. ACY-241 treatment also demonstrated an inhibitory effect on cell invasion and migration by reducing HIF-1α level. This study confirms that HDAC6 knockdown and ACY-241 treatment effectively decrease HIF-1α expression under normoxia, thereby suppressing the epithelial-mesenchymal transition. These findings highlight the potential of selective HDAC6 inhibition as an innovative therapeutic strategy for lung cancer.

2.
Int J Mol Sci ; 24(10)2023 May 12.
Article in English | MEDLINE | ID: mdl-37240016

ABSTRACT

The current method for diagnosing methamphetamine use disorder (MUD) relies on self-reports and interviews with psychiatrists, which lack scientific rigor. This highlights the need for novel biomarkers to accurately diagnose MUD. In this study, we identified transcriptome biomarkers using hair follicles and proposed a diagnostic model for monitoring the MUD treatment process. We performed RNA sequencing analysis on hair follicle cells from healthy controls and former and current MUD patients who had been detained in the past for illegal use of methamphetamine (MA). We selected candidate genes for monitoring MUD patients by performing multivariate analysis methods, such as PCA and PLS-DA, and PPI network analysis. We developed a two-stage diagnostic model using multivariate ROC analysis based on the PLS-DA method. We constructed a two-step prediction model for MUD diagnosis using multivariate ROC analysis, including 10 biomarkers. The first step model, which distinguishes non-recovered patients from others, showed very high accuracy (prediction accuracy, 98.7%). The second step model, which distinguishes almost-recovered patients from healthy controls, showed high accuracy (prediction accuracy, 81.3%). This study is the first report to use hair follicles of MUD patients and to develop a MUD prediction model based on transcriptomic biomarkers, which offers a potential solution to improve the accuracy of MUD diagnosis and may lead to the development of better pharmacological treatments for the disorder in the future.


Subject(s)
Amphetamine-Related Disorders , Methamphetamine , Humans , Methamphetamine/adverse effects , Amphetamine-Related Disorders/diagnosis , Amphetamine-Related Disorders/genetics , Hair Follicle , ROC Curve , Biomarkers
3.
J Cell Mol Med ; 24(12): 7055-7066, 2020 06.
Article in English | MEDLINE | ID: mdl-32406610

ABSTRACT

Statins are a class of lipid-lowering drugs that have recently been used in drug repositioning in the treatment of human cancer. However, the underlying mechanism of statin-induced cancer cell death has not been clearly defined. In the present study, we evaluated the anticancer effect of pitavastatin on oral squamous cell carcinoma (OSCC), SCC15 and SCC4 cells and found that FOXO3a might be a direct target in pitavastatin-induced cancer cell death. Our data revealed that pitavastatin selectively suppressed cell viability and induced intrinsic apoptosis in a FOXO3a-dependent manner in SCC15 cells while no effect was observed in SCC4 cells. Notably, treatment with pitavastatin in SCC15 cells induced the nuclear translocation of FOXO3a via dual regulation of two upstream kinases, AMPK and Akt, resulting in the up-regulation of PUMA, a transcriptional target gene of FOXO3a. Furthermore, our data revealed that FOXO3a-mediated PUMA induction plays a role in pitavastatin-induced intrinsic apoptosis in SCC15 cells. Taken together, our findings suggest that pitavastatin activates the FOXO3a/PUMA apoptotic axis by regulation of nuclear translocation of FOXO3a via Akt/FOXO3a or AMPK/FOXO3a signalling. Therefore, these findings might help to elucidate the underlying mechanism of the anticancer effects of pitavastatin on OSCC.


Subject(s)
Apoptosis/drug effects , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Forkhead Box Protein O3/metabolism , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Quinolines/pharmacology , Adenylate Kinase/metabolism , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Humans , Models, Biological , Neoplasm Metastasis , Phosphorylation/drug effects , Protein Transport/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
4.
Int J Mol Sci ; 21(22)2020 Nov 23.
Article in English | MEDLINE | ID: mdl-33238484

ABSTRACT

Methamphetamine (MA) use disorder is a chronic neuropsychiatric disease characterized by recurrent binge episodes, intervals of abstinence, and relapses to MA use. Therefore, identification of the key genes and pathways involved is important for improving the diagnosis and treatment of this disorder. In this study, high-throughput RNA sequencing was performed to find the key genes and examine the comparability of gene expression between whisker follicles and the striatum of rats following MA self-administration. A total of 253 and 87 differentially expressed genes (DEGs) were identified in whisker follicles and the striatum, respectively. Multivariate and network analyses were performed on these DEGs to find hub genes and key pathways within the constructed network. A total of 129 and 49 genes were finally selected from the DEG sets of whisker follicles and of the striatum. Statistically significant DEGs were found to belong to the classes of genes involved in nicotine addiction, cocaine addiction, and amphetamine addiction in the striatum as well as in Parkinson's, Huntington's, and Alzheimer's diseases in whisker follicles. Of note, several genes and pathways including retrograde endocannabinoid signaling and the synaptic vesicle cycle pathway were common between the two tissues. Therefore, this study provides the first data on gene expression levels in whisker follicles and in the striatum in relation to MA reward and thereby may accelerate the research on the whisker follicle as an alternative source of biomarkers for the diagnosis of MA use disorder.


Subject(s)
Amphetamine-Related Disorders/genetics , Hair Follicle/drug effects , Methamphetamine/pharmacology , Transcriptome/genetics , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amphetamine-Related Disorders/pathology , Animals , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , Hair Follicle/metabolism , High-Throughput Nucleotide Sequencing , Humans , Huntington Disease/genetics , Huntington Disease/pathology , Neostriatum/drug effects , Neostriatum/metabolism , Parkinson Disease/genetics , Parkinson Disease/pathology , Rats , Self Administration , Signal Transduction/drug effects , Vibrissae/drug effects , Vibrissae/metabolism
5.
Int J Mol Sci ; 21(17)2020 Aug 21.
Article in English | MEDLINE | ID: mdl-32839415

ABSTRACT

Methamphetamine (MA) is a highly addictive central nervous system stimulant. Drug addiction is not a static condition but rather a chronically relapsing disorder. Hair is a valuable and stable specimen for chronic toxicological monitoring as it retains toxicants and metabolites. The primary focus of this study was to discover the metabolic effects encompassing diverse pathological symptoms of MA addiction. Therefore, metabolic alterations were investigated in human hair following heavy MA abuse using both targeted and untargeted mass spectrometry and through integrated network analysis. The statistical analyses (t-test, variable importance on projection score, and receiver-operator characteristic curve) demonstrated that 32 metabolites (in targeted metabolomics) as well as 417 and 224 ion features (in positive and negative ionization modes of untargeted metabolomics, respectively) were critically dysregulated. The network analysis showed that the biosynthesis or metabolism of lipids, such as glycosphingolipids, sphingolipids, glycerophospholipids, and ether lipids, as well as the metabolism of amino acids (glycine, serine and threonine; cysteine and methionine) is affected by heavy MA abuse. These findings reveal crucial metabolic effects caused by MA addiction, with emphasis on the value of human hair as a diagnostic specimen for determining drug addiction, and will aid in identifying robust diagnostic markers and therapeutic targets.


Subject(s)
Amphetamine/analysis , Central Nervous System Stimulants/analysis , Hair/chemistry , Methamphetamine/analysis , Substance-Related Disorders/diagnosis , Adult , Amino Acids/chemistry , Amino Acids/classification , Amino Acids/isolation & purification , Amino Acids/metabolism , Amphetamine/administration & dosage , Amphetamine/metabolism , Case-Control Studies , Central Nervous System Stimulants/administration & dosage , Central Nervous System Stimulants/metabolism , Glycerophospholipids/chemistry , Glycerophospholipids/classification , Glycerophospholipids/isolation & purification , Glycerophospholipids/metabolism , Glycosphingolipids/chemistry , Glycosphingolipids/classification , Glycosphingolipids/isolation & purification , Glycosphingolipids/metabolism , Humans , Lipid Metabolism/physiology , Male , Metabolomics/methods , Methamphetamine/administration & dosage , Methamphetamine/metabolism , Middle Aged , Principal Component Analysis , Sphingolipids/chemistry , Sphingolipids/classification , Sphingolipids/isolation & purification , Sphingolipids/metabolism , Substance Abuse Detection/methods , Substance-Related Disorders/metabolism , Tandem Mass Spectrometry
6.
Molecules ; 25(3)2020 Jan 29.
Article in English | MEDLINE | ID: mdl-32013195

ABSTRACT

Arrest defective 1 (ARD1), also known as N(alpha)-acetyltransferase 10 (NAA10) was originally identified as an N-terminal acetyltransferase (NAT) that catalyzes the acetylation of N-termini of newly synthesized peptides. After that, mammalian ARD1/NAA10 expanded its' role to lysine acetyltransferase (KAT) that post-translationally acetylates internal lysine residues of proteins. ARD1/NAA10 is the only enzyme with both NAT and KAT activities. However, recent studies on the role of human ARD1/NAA10 (hARD1/NAA10) in lysine acetylation are contradictory, as crystal structure and in vitro acetylation assay results revealed the lack of KAT activity. Thus, the role of hARD1/NAA10 in lysine acetylation is still debating. Here, we found a clue that possibly explains these complicated and controversial results on KAT activity of hARD1/NAA10. Recombinant hARD1/NAA10 exhibited KAT activity, which disappeared soon in vitro. Size-exclusion analysis revealed that most recombinant hARD1/NAA10 formed oligomers over time, resulting in the loss of KAT activity. While oligomeric recombinant hARD1/NAA10 lost its ability for lysine acetylation, its monomeric form clearly exhibited lysine acetylation activity in vitro. We also characterized the KAT activity of hARD1/NAA10 that was influenced by several experimental conditions, including concentration of reactants and reaction time. Taken together, our study proves that recombinant hARD1/NAA10 exhibits KAT activity in vitro but only under accurate conditions, including reactant concentrations and reaction duration.


Subject(s)
Lysine Acetyltransferases/metabolism , N-Terminal Acetyltransferase A/metabolism , N-Terminal Acetyltransferase E/metabolism , Acetylation , Dialysis , Escherichia coli , Humans , Lysine/metabolism , N-Terminal Acetyltransferase A/genetics , N-Terminal Acetyltransferase A/isolation & purification , N-Terminal Acetyltransferase E/genetics , N-Terminal Acetyltransferase E/isolation & purification , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
7.
J Proteome Res ; 18(11): 3913-3925, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31525931

ABSTRACT

Persistent neurochemical disturbances by repeating drug reward and withdrawal lead to addiction. Particularly, drug withdrawal, usually starting within hours of the last dose, is considered as a critical step in the transition to addiction and a treatment clue. The aim of this study was to uncover metabolic effects associated with methamphetamine (MA) short-term abstinence using both non-targeted and targeted metabolomics. Metabolic alterations were investigated in rat plasma collected immediately after 16 days of MA self-administration and after 12 and 24 h of abstinence. Principal component analysis revealed that the highest level of separation occurred between the 24 h and saline (control) groups based on the significantly changed ion features, 257/320/333 and 331/409/388, in the SA/12 h/24 h groups in positive and negative modes of UPLC-QTOF-ESI-MS, respectively. Targeted metabolomics revealed dynamic changes in the biosynthesis/metabolism of amino acids, including the phenylalanine, tyrosine, and tryptophan biosynthesis and the valine, leucine, and isoleucine biosynthesis. Integrating non-targeted and targeted metabolomics data uncovered rapid and distinct changes in the metabolic pathways involved in energy metabolism, the nervous system, and membrane lipid metabolism. These findings provide essential knowledge of the dynamic metabolic effects associated with short-term MA abstinence and may help identify early warning signs of MA dependence.


Subject(s)
Energy Metabolism , Metabolomics/methods , Methamphetamine/administration & dosage , Substance Withdrawal Syndrome/metabolism , Amino Acids/biosynthesis , Amino Acids/blood , Amino Acids/metabolism , Animals , Chromatography, Liquid/methods , Male , Mass Spectrometry/methods , Metabolic Networks and Pathways , Principal Component Analysis , Rats, Sprague-Dawley , Self Administration , Substance Withdrawal Syndrome/blood , Substance Withdrawal Syndrome/physiopathology , Time Factors
8.
J Cell Mol Med ; 23(2): 1106-1115, 2019 02.
Article in English | MEDLINE | ID: mdl-30421568

ABSTRACT

The concept of drug repositioning has recently received considerable attention in the field of oncology. In the present study, we propose that paroxetine can be used as a potent anticancer drug. Paroxetine, one of the selective serotonin reuptake inhibitors (SSRIs), has been widely prescribed for the treatment of depression and anxiety disorders. Recently, SSRIs have been reported to have anticancer activity in various types of cancer cells; however, the underlying mechanisms of their action are not yet known. In this study, we investigated the potential anticancer effect of paroxetine in human colorectal cancer cells, HCT116 and HT-29. Treatment with paroxetine reduced cell viability, which was associated with marked increase in apoptosis, in both the cell lines. Also, paroxetine effectively inhibited colony formation and 3D spheroid formation. We speculated that the mode of action of paroxetine might be through the inhibition of two major receptor tyrosine kinases - MET and ERBB3 - leading to the suppression of AKT, ERK and p38 activation and induction of JNK and caspase-3 pathways. Moreover, in vivo experiments revealed that treatment of athymic nude mice bearing HT-29 cells with paroxetine remarkably suppressed tumour growth. In conclusion, paroxetine is a potential therapeutic option for patients with colorectal cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , Paroxetine/pharmacology , Proto-Oncogene Proteins c-met/metabolism , Receptor, ErbB-3/metabolism , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Colonic Neoplasms/metabolism , HCT116 Cells , HT29 Cells , Humans , Male , Mice , Mice, Nude , Selective Serotonin Reuptake Inhibitors/pharmacology
9.
Int J Mol Sci ; 20(24)2019 Dec 09.
Article in English | MEDLINE | ID: mdl-31835389

ABSTRACT

Thiazolidinedione is a five-membered heterocycle that is widely used in drug discovery endeavors. In this study, we report the design, synthesis, and biological evaluation of a series of thiazolidinedione-based HDAC6 inhibitors. In particular, compound 6b exerts an excellent inhibitory activity against HDAC6 with an IC50 value of 21 nM, displaying a good HDAC6 selectivity over HDAC1. Compound 6b dose-dependently induces the acetylation level of α-tubulin via inhibition of HDAC6 in human neuroblastoma SH-SY5Y cell line. Moreover, compound 6b efficiently reverses methamphetamine-induced morphology changes of SH-SY5Y cells via regulating acetylation landscape of α-tubulin. Collectively, compound 6b represents a novel HDAC6-isoform selective inhibitor and demonstrates promising therapeutic potential for the treatment of methamphetamine addiction.


Subject(s)
Drug Discovery , Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase Inhibitors , Thiazolidinediones , Amphetamine-Related Disorders/drug therapy , Amphetamine-Related Disorders/enzymology , Cell Line, Tumor , Histone Deacetylase 6/chemistry , Histone Deacetylase 6/metabolism , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/metabolism , Thiazolidinediones/chemical synthesis , Thiazolidinediones/chemistry , Thiazolidinediones/pharmacology
10.
Molecules ; 24(12)2019 Jun 12.
Article in English | MEDLINE | ID: mdl-31212725

ABSTRACT

Metabolomics is a powerful tool used to understand comprehensive changes in the metabolic response and to study the phenotype of an organism by instrumental analysis. It most commonly involves mass spectrometry followed by data mining and metabolite assignment. For the last few decades, hair has been used as a valuable analytical sample to investigate retrospective xenobiotic exposure as it provides a wider window of detection than other biological samples such as saliva, plasma, and urine. Hair contains functional metabolomes such as amino acids and lipids. Moreover, segmental analysis of hair based on its growth rate can provide information on metabolic changes over time. Therefore, it has great potential as a metabolomics sample to monitor chronic diseases, including drug addiction or abnormal conditions. In the current review, the latest applications of hair metabolomics in animal studies and clinical settings are highlighted. For this purpose, we review and discuss the characteristics of hair as a metabolomics sample, the analytical techniques employed in hair metabolomics and the consequence of hair metabolome alterations in recent studies. Through this, the value of hair as an alternative biological sample in metabolomics is highlighted.


Subject(s)
Hair/metabolism , Metabolome , Metabolomics , Animals , Chromatography, High Pressure Liquid , Humans , Mass Spectrometry , Metabolomics/methods
11.
J Neuroinflammation ; 14(1): 240, 2017 Dec 11.
Article in English | MEDLINE | ID: mdl-29228978

ABSTRACT

BACKGROUND: Methamphetamine (METH) is a commonly abused drug that may result in neurotoxic effects. Recent studies have suggested that involvement of neuroinflammatory processes in brain dysfunction is induced by misuse of this drug. However, the mechanism underlying METH-induced inflammation and neurotoxicity in neurons is still unclear. In this study, we investigated whether asiatic acid (AA) effected METH-mediated neuroinflammation and neurotoxicity in dopaminergic neuronal cells. And we further determined whether the effect involved in the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and signal transducer and activator of transcription (STAT)3 and extracellular signal-regulated kinase (ERK) pathway. METHODS: We used the human dopaminergic neuroblastoma SH-SY5Y cell line, murine microglial BV2 cell line, and primary culture of rat embryo mesencephalic neurons. Pro-inflammatory cytokine production was monitored by ELISA and RT/real-time PCR. The cell cycle distribution and mitochondrial membrane integrity was analyzed by flow cytometry. We used immunoblotting, DNA-binding activity, and immunofluorescence staining to analyze the effect of AA on activation of the NF-κB, STAT3, MAPK-ERK, and apoptosis signaling pathways. RESULTS: METH induced TNF receptor (TNFR) expression and led to morphological changes of cells. Additionally, this drug increased pro-inflammatory cytokine (TNFα and IL-6) expression. AA significantly suppressed METH-induced TNFR expression in concentration dependent. Increased secretion of TNFα and IL-6 was inhibited in METH-stimulated neuronal cells by AA administration. AA showed significant protection against METH-induced translocation of NF-κB/STAT3 and ERK phosphorylation. AA inhibited METH-induced proteolytic fragmentation of caspase-3 and PARP. The pro-apoptotic protein Bax was significantly decreased, while the anti-apoptotic protein Bcl-xL was increased by AA treatment in METH-stimulated cells. A similar protective effect of AA on mitochondrial membrane integrity was also confirmed by flow cytometry and immunofluorescence staining. CONCLUSIONS: Based on the literatures and our findings, AA is a promising candidate for an anti-neurotoxic agent, and it can potentially be used for the prevention and treatment of various neurological disorders.


Subject(s)
Central Nervous System Stimulants/toxicity , Methamphetamine/toxicity , Neurons/drug effects , Neuroprotective Agents/pharmacology , Pentacyclic Triterpenes/pharmacology , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Cell Line , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Inflammation/chemically induced , Mice , Mitochondria/drug effects , NF-kappa B/metabolism , Neurons/metabolism , Rats , STAT3 Transcription Factor/metabolism
12.
J Cell Physiol ; 231(2): 357-69, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26089158

ABSTRACT

Adiponectin predominantly secreted from adipose tissue has exhibited potent anti-proliferative properties in cancer cells via modulating cell cycle and apoptosis. FoxO3A, a Forkhead box O member of the transcription factor, plays a critical role in modulating expression of genes involved in cell death and/or survival. In this study, we investigated the role of FoxO3A signaling in anti-cancer activities of adiponectin. Herein, we have shown that treatment with globular adiponectin (gAcrp) increases p27 but decreases cyclinD1 expression in human hepatoma (HepG2) and breast (MCF-7) cancer cells. Gene ablation of FoxO3A prevented gAcrp-induced increase in p27 and decreased in cyclin D1 expression, and further ameliorated cell cycle arrest by gAcrp, indicating a critical role of FoxO3A in gAcrp-induced cell cycle arrest of cancer cells. Moreover, treatment with gAcrp also induced caspase-3/7 activation and increased Fas ligand (FasL) expression in both HepG2 and MCF-7 cells. Transfection with FoxO3A siRNA inhibited gAcrp-induced caspase-3/7 activation and FasL expression, suggesting that FoxO3A signaling also plays an important role in gAcrp-induced apoptosis of cancer cells. We also found that gene silencing of AMPK prevented gAcrp-induced nuclear translocation of FoxO3A in HepG2 and MCF-7 cells. In addition, suppression of AMPK also blocked gAcrp-induced cell cycle arrest and further attenuated gAcrp-induced caspase-3/7 activation, indicating that AMPK signaling plays a pivotal role in both gAcrp-induced cell cycle arrest and apoptosis via acting as an upstream signaling of FoxO3A. Taken together, our findings demonstrated that AMPK/FoxO3A axis plays a cardinal role in anti-proliferative effect of adiponectin in cancer cells.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adiponectin/metabolism , Forkhead Transcription Factors/metabolism , Neoplasms/metabolism , Neoplasms/pathology , AMP-Activated Protein Kinase Kinases , Apoptosis/physiology , Caspase 3/metabolism , Caspase 7/metabolism , Cell Cycle Checkpoints/physiology , Fas Ligand Protein/metabolism , Forkhead Box Protein O3 , Forkhead Transcription Factors/antagonists & inhibitors , Forkhead Transcription Factors/genetics , Gene Knockout Techniques , Hep G2 Cells , Humans , MCF-7 Cells , Models, Biological , Neoplasms/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/genetics , Signal Transduction
13.
Biol Pharm Bull ; 39(9): 1488-95, 2016.
Article in English | MEDLINE | ID: mdl-27582330

ABSTRACT

The effective cure for oral squamous cell carcinoma (OSCC) patients is challenging due late diagnosis and fatal metastasis. The standard diagnosis for OSCC often depends on the subjective interpretation of conventional histopathology. Additionally, there is no standard way for OSCC prognosis. Over the past decade, nano-mechanical stiffness has been considered as a quantitative measure for cancer diagnosis. Nevertheless, its application to OSCC diagnosis and prognosis is still in a primitive stage. In this study, we investigated whether the OSCC progression can be predicted by nano-mechanical properties in combination with biochemical properties, especially the epithelial-mesenchymal transition (EMT). Atomic force microscopy-based nano-mechanical measurements of three different OSCC cell lines-SCC-4, SCC-9, and SCC-15-were conducted together with biochemical analyses. The gradual upregulation of Snail2, N-cadherin, and vimentin and the simultaneous downregulation of E-cadherin were observed, and the degree of upregulation and downregulation was stronger in the order of the cell lines mentioned above. The strength of enhancement in migration was in the same order as well. Consistently, nano-mechanical stiffness was gradually decreased as the EMT progresses. These results suggest that the nano-mechanical assay could serve as a quantitative tool to predict the OSCC progression in the context of the EMT. Furthermore, we found that the upregulated vimentin, a major filamentous component of the cytoskeleton, may contribute to mechanical softening, which can be discerned from the role of actin filaments in mechanical stiffness. In conclusion, our combinational study proposes a novel way to elucidate the mechanism of OSCC progression and its therapeutic targets.


Subject(s)
Carcinoma, Squamous Cell/pathology , Epithelial-Mesenchymal Transition , Mouth Neoplasms/pathology , Antigens, CD/metabolism , Cadherins/metabolism , Cell Line, Tumor , Cell Movement , Elastic Modulus , Humans , Microscopy, Atomic Force , Snail Family Transcription Factors/metabolism , Vimentin/metabolism
14.
J Biol Chem ; 289(52): 35839-48, 2014 Dec 26.
Article in English | MEDLINE | ID: mdl-25368326

ABSTRACT

Non-small-cell lung cancer (NSCLC) is associated with diverse genetic alterations including mutation of epidermal growth factor receptor (EGFR). Isoliquiritigenin (ILQ), a chalcone derivative, possesses anticancer activities. In the present study, we investigated the effects of ILQ on the growth of tyrosine kinase inhibitor (TKI)-sensitive and -resistant NSCLC cells and elucidated its underlying mechanisms. Treatment with ILQ inhibited growth and induced apoptosis in both TKI-sensitive and -resistant NSCLC cells. ILQ-induced apoptosis was associated with the cleavage of caspase-3 and poly-(ADP-ribose)-polymerase, increased expression of Bim, and reduced expression of Bcl-2. In vitro kinase assay results revealed that ILQ inhibited the catalytic activity of both wild type and double mutant (L858R/T790M) EGFR. Treatment with ILQ inhibited the anchorage-independent growth of NIH3T3 cells stably transfected with either wild type or double-mutant EGFR with or without EGF stimulation. ILQ also reduced the phosphorylation of Akt and ERK1/2 in both TKI-sensitive and -resistant NSCLC cells, and attenuated the kinase activity of Akt1 and ERK2 in vitro. ILQ directly interacted with both wild type and double-mutant EGFR in an ATP-competitive manner. A docking model study showed that ILQ formed two hydrogen bonds (Glu-762 and Met-793) with wild type EGFR and three hydrogen bonds (Lys-745, Met-793, and Asp-855) with mutant EGFR. ILQ attenuated the xenograft tumor growth of H1975 cells, which was associated with decreased expression of Ki-67 and diminished phosphorylation of Akt and ERK1/2. Taken together, ILQ suppresses NSCLC cell growth by directly targeting wild type or mutant EGFR.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Chalcones/pharmacology , ErbB Receptors/genetics , Lung Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cell Proliferation , ErbB Receptors/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , HEK293 Cells , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Nude , Mutation, Missense , NIH 3T3 Cells , Protein Binding , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
15.
Mol Carcinog ; 54(2): 83-92, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24038446

ABSTRACT

The Akt, family of serine/threonine protein kinases functions as key regulators of multiple aspects of cell behavior, such as survival, proliferation, migration, and carcinogenesis. Notably, Akt exerts its anti-apoptotic effects through the phosphorylation of numerous substrates related with cell cycle, genome stability, and cancer development. In this report, nevertheless, we focused our view on the novel role of Akt which involves in a pro-apoptotic action by phosphorylating second mitochondria derived activator of caspases (Smac) protein during etoposide-induced apoptotic processes. Our data reveals that Akt could bind to and phosphorylate Smac at serine residue 67, which enhances the ability of Smac to interact with the cytosolic X-chromosome linked IAP (XIAP) protein. The cellular interaction of wild-type Smac with XIAP was enhanced with similar activation kinetics of Akt activity, while this interaction was markedly attenuated in cells expressing the phosphorylation-defective mutant S67A-Smac during etoposide-induced apoptosis. Moreover, we provide the evidence indicating that the phosphorylation of Smac at ser-67 markedly upregulates the caspase-3 activity by promoting the interaction of Smac with XIAP. Taken together, we propose that the phosphorylation of Smac by Akt might be a novel mechanism that involves in amplification of caspase cascade pathway during etoposide-induced apoptosis in HeLa cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Caspase 3/metabolism , Etoposide/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Mitochondrial Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Apoptosis Regulatory Proteins , Caspase 3/genetics , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , HeLa Cells , Humans , Phosphorylation , Serine/metabolism , Signal Transduction/drug effects , X-Linked Inhibitor of Apoptosis Protein/metabolism
16.
Mol Carcinog ; 54(4): 322-31, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24974831

ABSTRACT

Lung cancer is a leading cause of death worldwide and MET amplification is a major therapeutic limitation in acquired-resistance lung cancer. We hypothesized that butein, a phytochemical, can overcome gefitinib-induced resistance by targeting both EGFR and MET in non-small cell lung cancer (NSCLC). To investigate the ability of butein to target EGFR and MET, we used in silico docking, a library of natural compounds and kinase assays. The effects of butein on growth, induction of apoptosis and expression of EGFR/MET signaling targets were examined in HCC827 (gefitinib-sensitive) and HCC827GR (gefitinib-resistant) NSCLC cells. Results were confirmed in vivo by a HCC827 or HCC827GR cell xenograft mouse model, each treated with vehicle, butein or gefitinib. Butein inhibited phosphorylation and kinase activity of EGFR and MET as well as soft agar colony formation and decreased viability of HCC827 and HCC827GR cells. Butein increased apoptosis-related protein expression in these cells. Results were confirmed by co-treatment with inhibitors of EGFR/MET or double knock-down. Finally, xenograft study results showed that butein strongly suppressed HCC827 and HCC827GR tumor growth. Immunohistochemical data suggest that butein inhibited Ki-67 expression. These results indicate that butein has potent anticancer activity and targets both EGFR and MET in acquired-resistance NSCLC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Chalcones/pharmacology , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/drug therapy , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Quinazolines/pharmacology , Animals , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , ErbB Receptors/metabolism , Gefitinib , Humans , Lung/drug effects , Lung/metabolism , Lung/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice, Nude , Molecular Docking Simulation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/metabolism
17.
Cancer Sci ; 105(10): 1245-53, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25117641

ABSTRACT

Although epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) have been introduced for the treatment of non-small cell lung cancer (NSCLC), the emergence of secondary T790M mutation in EGFR or amplification of the Met proto-oncogene restrain the clinical success of EGFR-TKIs. Since heat shock protein-90 (Hsp90) stabilizes various oncoproteins including EGFR and c-Met, the inhibition of Hsp90 activity appears as a rational strategy to develop anticancer drugs. Despite preclinical efficacy of geldanamycin-anasamycin (GA)-derivatives containing benzoquinone moiety as Hsp90 inhibitors, the hepatotoxicity of these GA-derivatives restricts their therapeutic benefit. We have prepared WK-88 series of GA-derivatives, which lack the benzoquinone moiety. In this study, we have examined the anticancer effects of WK88-1 in Met-amplified- and gefitinib-resistant (HCC827GR) NSCLC cells and its parental HCC827 cells. Treatment with WK88-1 reduced the cell viability in both HCC827 and HCC827GR cells, which was associated with marked decrease in the constitutive expression of Hsp90 client proteins, such as EGFR, ErbB2, ErbB3, Met and Akt. Moreover, WK88-1 attenuated phosphorylation of these Hsp90 client proteins and reduced the anchorage-independent growth of HCC827GR cells. Administration of WK88-1 did not cause hepatotoxicity in animals and significantly reduced the growth of HCC827GR cells xenograft tumors in nude mice. Our study provides evidence that ErbB3 might be a client for Hsp90 in Met-amplified NSCLCs. In conclusion, we demonstrate that inhibition of Hsp90 dampens the activation of EGFR- or c-Met-mediated survival of Met-amplified NSCLCs and that WK88-1 as a Hsp90 inhibitor alleviates gefitinib resistance in HCC827GR cells.


Subject(s)
Antineoplastic Agents/pharmacology , Benzoquinones/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Gene Amplification , Lactams, Macrocyclic/pharmacology , Lung Neoplasms/drug therapy , Proto-Oncogene Proteins c-met/genetics , Quinazolines/pharmacology , Animals , Apoptosis/drug effects , Benzoquinones/chemistry , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm , Gefitinib , Humans , Lactams, Macrocyclic/chemistry , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Proto-Oncogene Mas , Receptor, ErbB-3/metabolism
18.
Bioorg Med Chem Lett ; 24(1): 224-7, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24345447

ABSTRACT

Heat shock protein 90 (Hsp90) represents an attractive cancer therapeutic target due to its role in the stabilization and maturation of many oncogenic proteins. We have designed a series of hybrid Hsp90 inhibitors by connecting the resorcinol ring of VER-49009 (2) and the trimethoxyphenyl ring of PU3 (3) using structure-based approach. Subsequent testing established that compound 1f inhibited gefitinib-resistant H1975 cell proliferation, brought about the degradation of Hsp90 client proteins including EGFR, Met, Her2 and Akt and induced the expression of Hsp70. The design, synthesis, and evaluation of 1f are described herein.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Discovery , Drug Resistance, Neoplasm/drug effects , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lung Neoplasms/drug therapy , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Crystallography, X-Ray , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Lung Neoplasms/pathology , Models, Molecular , Molecular Structure , Structure-Activity Relationship
19.
Inflamm Res ; 62(2): 145-54, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23064654

ABSTRACT

OBJECTIVE: Porphyromonas gingivalis is a major periodontopathogen that plays a role in the pathogenesis of periodontal disease. In this study, we investigated the effect of 18alpha-glycyrrhetinic acid (18α-GA), a natural triterpenoid compound derived from licorice root extract, on P. gingivalis lipopolysaccharide (LPS)-induced vascular permeability, which is a hallmark of inflammatory diseases such as periodontitis. METHODS: The inhibitory effects of 18α-GA on endothelial permeability were determined by measuring in vivo and in vitro endothelial permeability. Endothelial cells were pretreated with 18α-GA before exposure to P. gingivalis LPS, and total RNA or proteins were extracted and analyzed by reverse transcription polymerase chain reaction or western blotting. RESULTS: Porphyromonas gingivalis LPS-induced endothelial permeability was significantly inhibited by 18α-GA both in vivo and in vitro. 18α-GA reduces P. gingivalis LPS-induced gap formation of endothelial cells. Importantly, 18α-GA modulated the expression and secretion of interleukin-8 (IL-8), a key inducer of vascular permeability, by downregulating nuclear factor-κB (NF-κB). 18α-GA suppressed P. gingivalis LPS-stimulated inhibitor of kappa B (IκB) kinase activation, IκBα phosphorylation, and nuclear translocation of NF-κB. CONCLUSIONS: Overall, these findings suggest that 18α-GA significantly reduces P. gingivalis LPS-induced vascular permeability by repressing NF-κB-dependent endothelial IL-8 production, suggesting its therapeutic potential in P. gingivalis-related vascular diseases.


Subject(s)
Capillary Permeability/drug effects , Endothelial Cells/drug effects , Glycyrrhetinic Acid/pharmacology , Lipopolysaccharides , Porphyromonas gingivalis , Cells, Cultured , Endothelial Cells/metabolism , Humans , Interleukin-8/metabolism , NF-kappa B/metabolism
20.
J Pineal Res ; 54(3): 264-70, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22924616

ABSTRACT

The sustained expansion of a tumor mass requires new blood vessel formation to provide rapidly proliferating tumor cells with an adequate supply of oxygen and nutrients. Hypoxia-inducible factor-1 (HIF-1) plays an essential role in tumor angiogenesis and growth by regulating the transcription of genes in response to hypoxic stress. This study was designed to investigate the effects of melatonin on tumor growth and angiogenesis, as well as the mechanism underlying the antitumor activities of melatonin. In this study, we show that the administration of melatonin inhibits tumor growth and blocks tumor angiogenesis in mice. Moreover, melatonin diminished the expression of the HIF-1α protein within the tumor mass during tumorigenesis. Our findings suggest that melatonin is a promising anti-angiogenic therapeutic agent targeting HIF-1α in cancer. Considering that HIF-1α is overexpressed in a majority of human cancers, melatonin could offer a potent therapeutic agent for cancer.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Neoplasms/blood supply , Kidney Neoplasms/drug therapy , Melatonin/pharmacology , Animals , Cell Line, Tumor , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Microvessels/drug effects , Microvessels/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology
SELECTION OF CITATIONS
SEARCH DETAIL