Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Med Oncol ; 40(4): 120, 2023 Mar 19.
Article in English | MEDLINE | ID: mdl-36934368

ABSTRACT

Targeting the extracellular matrix (ECM) is considered as a promising strategy in cancer therapeutics. This study was designed to identify the potential ECM modulators for gastric cancer therapeutics. Exploration of the expression profiles of gastric tumors revealed the elevated expression of ECM genes in gastric tumor tissues compared to the adjacent normal tissues with increased expression in diffuse subtype gastric tumors and specifically in epithelial to mesenchymal transition (EMT) molecular subtype tumors. Consensus ECM gene set was derived from the expression profiles of gastric tumors. The correlative analysis was performed between the expression pattern of the ECM gene set and the drug sensitivity pattern of a panel of drugs across gastric cancer cell lines. Negative correlation between the expression of ECM genes and sensitivity of a number of drugs targeting PI3K/mTOR signaling, chromatin histone acetylation and ABL signaling was observed. These pathways are known for their role in cell-mediated adhesion, differentiation and epithelial to mesenchymal transition. The current results reveal the possibility of using PI3K/AKT/mTOR modulators for targeted gastric cancer therapy in patients with dysregulated ECM.


Subject(s)
Stomach Neoplasms , Humans , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , MTOR Inhibitors , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Epithelial-Mesenchymal Transition , Cell Line, Tumor , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Extracellular Matrix/metabolism
2.
Funct Integr Genomics ; 22(6): 1345-1360, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35987846

ABSTRACT

Deregulated transcription programs and signaling pathways are the critical factors involved in the process of carcinogenesis. Signaling pathway-based classification of tumors is expected to pave the way for the development of targeted therapeutics. We investigated the OCT4-mediated transcription program in the gene expression profiles of 939 gastric tumor samples. A set of 84 genes showing positive correlation with the activation pattern of the available OCT4 gene sets were found to consistently express in diffuse, poorly differentiated, and stage-III gastric tumors with poor prognosis. We also developed stable OCT4-silenced gastric cancer cells and the resultant gene expression changes were investigated by genome-wide mRNA profiling. Functional genomic investigation of the genes downregulated in OCT4-silenced cells and the pathways co-activated with OCT4 gene set across gastric tumors revealed the positive association of dysregulated OCT4 with TGF-ß, GLI, PRC2/EzH2, Wnt, KRAS, STK33, and YAP signaling pathways in diffuse subtype gastric tumors. Elevated expression of OCT4 gene set was identified to represent the previously described EMT_UP as well as the GENOMICALLY STABLE subtypes of gastric tumors. Integrative genomic screening of the drug sensitivity of gastric cancer cells in correlation with the expression of OCT4 gene set across drug sensitivity databases revealed the inhibitors of tyrosine kinases, HDAC, and HSP90 to have a negative correlation and needs to be investigated for their potential therapeutic features for the subset of OCT4-activated gastric tumors. Thus, the subset of gastric tumors with OCT4 activation, the associated oncogenic signaling pathways, and potential therapeutic candidates were identified for the development of targeted therapeutic strategies.


Subject(s)
Octamer Transcription Factor-3 , Stomach Neoplasms , Humans , Carcinogenesis/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Signal Transduction , Stomach Neoplasms/metabolism , Transforming Growth Factor beta/genetics , Octamer Transcription Factor-3/genetics
3.
Med Oncol ; 39(9): 132, 2022 Jun 20.
Article in English | MEDLINE | ID: mdl-35723749

ABSTRACT

Metabolic dysregulations have been identified as intrinsic hallmarks of cancer cells. Investigations of altered metabolic processes, in the context of the associated oncogenic signaling pathways are expected to pave way for the development of targeted cancer therapeutics. We have recently identified the enrichment of glucose and glutamine metabolism in a subset of intestinal subtype gastric tumors at the level of expression of genes, gene sets and the occurrence of metabolites. On the other hand, glucose transport, glucan and fatty acid metabolism were enriched in a subset of diffuse subtype gastric tumors. In the current study, along with glucose metabolism, mTOR, HSP90, MYC, E2F, P53 and proteasome pathways were found enriched in a subset of intestinal subtype and a part of MSI subtype gastric tumors. On the other hand, along with fatty acid metabolism, the oncogenic pathway KRAS was found to be enriched in a subset of GS tumors among diffuse subtype gastric tumors. Thus, oncogenic signaling pathways associated with two distinct metabolic rewiring which differentially occurs between major gastric cancer subtypes were identified. These pathways seem the potential targets to differentially target these gastric cancer subtypes. Exploratory integrative genomic analyses reveal HSP90 inhibitors, AKT/mTOR inhibitors, and cell cycle inhibitors as potential agents to target the gastric tumors with the rewired glucose metabolism and MEK/MAPK inhibitors as suitable drug candidates to target the diffuse subtype tumors with the dysregulated fatty acid metabolism. This observation would pave way for the selective and targeted use of signaling pathway modulators for targeted and stratified gastric cancer therapeutics.


Subject(s)
Stomach Neoplasms , Carcinogenesis , Fatty Acids , Glucose , Humans , Signal Transduction , Stomach Neoplasms/pathology
4.
Exp Cell Res ; 410(1): 112956, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34864005

ABSTRACT

The ERK/MAPK signaling pathway is activated in various cancers including gastric cancer. Targeting the ERK/MAPK/MEK pathway has been considered as a promising strategy for cancer therapy. However, MEK inhibition leads to a series of resistance mechanisms due to mutations in MEK, elevated expression of RAS or RAF proteins and activation of the associated signaling pathways. In the present study, ERK/MAPK pathway specific gene signatures were identified to be highly activated in intestinal subtype gastric tumors. Inhibition of ERK/MAPK pathway with the inhibitor PD98059 in gastric cancer cell lines by in vitro signaling pathway and genome-wide expression profiling revealed the associated signaling pathways. Functional genomic investigation of the ERK/MAPK regulated genes reveals the association of ERK/MAPK pathway with E2F, Myc, SOX-2, TGF-ß, OCT4 and Notch pathways in gastric cancer cells. Of these, E2F, Myc and SOX-2 pathways are activated in intestinal subtype gastric tumors and TGF-ß, OCT4, Notch pathways are activated in diffuse subtype gastric tumors. Further, the mutational load of gastric tumors was found to have association and correlation with the activation pattern of ERK/MAPK pathways across gastric tumors. ERK/MAPK activation was also found to represent the EBV and MSI activated subtypes of gastric tumors. Identification of potent drug candidates inhibiting the ERK/MAPK and associated pathways would pave a way for developing the targeted therapeutics for a subset of gastric tumors with activated ERK/MAPK signaling cascade.


Subject(s)
MAP Kinase Signaling System/genetics , Molecular Targeted Therapy , Stomach Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Mitogen-Activated Protein Kinase Kinases/genetics , Neoplasm Proteins/genetics , Protein Kinase Inhibitors/therapeutic use , Signal Transduction/genetics , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology
5.
Oncogene ; 40(34): 5327-5341, 2021 08.
Article in English | MEDLINE | ID: mdl-34253860

ABSTRACT

P21 Activated Kinase 1 (PAK1) is an oncogenic serine/threonine kinase known to play a significant role in the regulation of cytoskeleton and cell morphology. Runt-related transcription factor 3 (RUNX3) was initially known for its tumor suppressor function, but recent studies have reported the oncogenic role of RUNX3 in various cancers. Previous findings from our laboratory provided evidence that Threonine 209 phosphorylation of RUNX3 acts as a molecular switch in dictating the tissue-specific dualistic functions of RUNX3 for the first time. Based on these proofs and to explore the translational significance of these findings, we designed a small peptide (RMR) from the protein sequence of RUNX3 flanking the Threonine 209 phosphorylation site. The selection of this specific peptide from multiple possible peptides was based on their binding energies, hydrogen bonding, docking efficiency with the active site of PAK1 and their ability to displace PAK1-RUNX3 interaction in our prediction models. We found that this peptide is stable both in in vitro and in vivo conditions, not toxic to normal cells and inhibits the Threonine 209 phosphorylation in RUNX3 by PAK1. We also tested the efficacy of this peptide to block the RUNX3 Threonine 209 phosphorylation mediated tumorigenic functions in in vitro cell culture models, patient-derived explant (PDE) models and in in vivo tumor xenograft models. These results proved that this peptide has the potential to be developed as an efficient therapeutic molecule for targeting RUNX3 Threonine 209 phosphorylation-dependent tumor phenotypes.


Subject(s)
p21-Activated Kinases , Carcinogenesis , Humans , Oncogenes , Phosphorylation , Protein Serine-Threonine Kinases , Threonine
6.
Drug Dev Res ; 82(8): 1182-1192, 2021 12.
Article in English | MEDLINE | ID: mdl-34002415

ABSTRACT

The oncogenic signaling pathway Wnt is often activated in many cancers including gastric cancer. Wnt signaling pathway is considered as a potential target for developing new targeted therapeutics. Kinase inhibitors are the promising class of drugs for many diseases including cancers. Toward identifying the potent inhibitors targeting Wnt signaling pathway, a kinase inhibitor library with 82 inhibitors were screened using Wnt pathway reporter assay in gastric cancer cells. Notably, 34 kinase inhibitors were identified to inhibit Wnt mediated reporter activity to the extent of more than 50%. The corresponding kinase genes, which are known targets of these kinase inhibitors, were investigated for their expression in the available mRNA profiles of gastric tumors. A major group of the kinase genes showed higher expression in intestinal subtype gastric tumors. Another group of kinase genes were found expressed in diffuse type gastric tumors. The kinase genes expressed in intestinal type gastric tumors were found associated with varying survival of gastric cancer patients whereas those expressed in diffuse type tumors were found associated with the poor survival. Thus, the kinase genes specifically expressed in intestinal and diffuse type gastric tumors and the kinase inhibitors to target Wnt signaling pathway in gastric cancer subtypes have been identified.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Protein Kinases/genetics , Stomach Neoplasms/drug therapy , Wnt Signaling Pathway/drug effects , Cell Line, Tumor , Humans , Protein Kinase Inhibitors/therapeutic use , Stomach Neoplasms/enzymology , Stomach Neoplasms/mortality
7.
Toxicol In Vitro ; 74: 105152, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33771646

ABSTRACT

Elevated expression of YY1 is known to confer anti-apoptotic phenotype and hence is an attractive target for cancer therapeutics. In a repurpose screening, towards the identification of the inhibitors of YY1 regulated transcription in gastric cancer cells, the calcium channel blockers lercanidipine and amlodipine have been identified to inhibit YY1 more efficiently. We further probed these calcium channel blockers for their potential feature of alleviating the drug resistance in gastric cancer cells. Lercanidipine and amlodipine were found to show an enhanced effect with doxorubicin in inhibiting the growth of gastric cancer cells. While doxorubicin was identified to activate the pathways TGF-ß and ERK/MAPK, lercanidipine was found to inhibit these pathways. This being the molecular mechanism behind the identified advantage of lercanidipine and amlodipine in sensitizing gastric cancer cells to doxorubicin. In multiple cellular models from different lineages, the cells with less sensitivity to doxorubicin were found to have the inherent activation of ERK/MAPK and TGF-ß pathways. Also, we have identified that doxorubicin, in combination with any of the calcium channel blockers, could inhibit the potential of cellular proliferation and spheroid formation in gastric cancer cells. The current study shows the usefulness of lercanidipine and amlodipine for the targeted and combinatorial therapeutics of gastric cancer and specifically to improve the efficiency of doxorubicin.


Subject(s)
Amlodipine/pharmacology , Antibiotics, Antineoplastic/pharmacology , Calcium Channel Blockers/pharmacology , Dihydropyridines/pharmacology , Doxorubicin/pharmacology , Stomach Neoplasms/drug therapy , Cell Line , Cell Survival/drug effects , Drug Synergism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Humans , Stomach Neoplasms/genetics , Transcription, Genetic , Transcriptome/drug effects , Transforming Growth Factor beta/antagonists & inhibitors , YY1 Transcription Factor/antagonists & inhibitors
8.
J Cell Commun Signal ; 15(2): 251-267, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33620645

ABSTRACT

Gastric cancer is one of the leading causes of cancer-related death worldwide. The transcription factor YY1 regulates diverse biological processes, including cell proliferation, development, DNA damage responses, and carcinogenesis. This study was designed to explore the role of YY1 regulated transcription in gastric cancer. YY1 silencing in gastric cancer cells has resulted in the inhibition of Wnt/ß-catenin, JNK/MAPK, ERK/MAPK, ER, and HIF-1α signaling pathways. Genome-wide mRNA profiling upon silencing the expression YY1 gene in gastric cancer cells and comparison with the previously identified YY1 regulated genes from other lineages revealed a moderate overlap among the YY1 regulated genes. Despite the differing genes, all the YY1 regulated gene sets were expressed in most of the intestinal subtype gastric tumors and a subset of diffuse subtype gastric tumors. Integrative functional genomic analysis of the YY1 gene sets revealed an association among the pathways Wnt/ß-catenin, Rapamycin, Cyclin-D1, Myc, E2F, PDGF, and AKT. Further, the drugs capable of inhibiting YY1 mediated transcription were identified as suitable targeted therapeutic candidates for gastric tumors with activated YY1. The data emerging from the investigation would pave the way for the development of YY1-based targeted therapeutics for gastric cancer.

9.
Hum Cell ; 34(2): 445-456, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33405176

ABSTRACT

To overcome the issues of poor prognosis and to tackle the non-responsiveness to various chemotherapeutics; it is necessary to develop targeted cancer therapeutic agents. Also, it is being necessary to understand the molecular targets of the drug candidates and drugs in the context of cellular signaling pathways, to make progress towards the development of targeted cancer therapeutics. Towards addressing these, we have established a cell-based and pathway-focused drug screening system for the pathways such as MYC, E2F, WNT, ERK, NRF1/2, HIF1α, p53, YY1 and NFκB. These signaling pathways are highly dysregulated in many cancers, including gastric cancer. The developed firefly luciferase assay-based screening system in gastric cancer lineage is suitable for the screening of the massive panel of drugs, drug candidates, small molecule inhibitors, chemicals and alternate drug formulations. The developed stable cell lines have been demonstrated for their pathway activity reporting features using the corresponding pathway-specific modulators. A proof-of-concept medium throughput screening focusing on YY1 signaling pathway also revealed the connection between calcium channel blockers and YY1 signaling. The developed signaling pathway screening assay cells are valuable resource and will serve as the screening platform for screening the drug libraries towards the development of targeted cancer therapeutics.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor/methods , High-Throughput Screening Assays/methods , Neoplasms/genetics , Neoplasms/pathology , Signal Transduction/genetics , Cell Line, Tumor , E2F Transcription Factors , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , Mitogen-Activated Protein Kinases , NF-kappa B , Nuclear Respiratory Factor 1 , Proto-Oncogene Proteins c-myc , Tumor Suppressor Protein p53 , YY1 Transcription Factor
10.
Funct Integr Genomics ; 20(6): 813-824, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32949316

ABSTRACT

Gastric cancer is one of the most common cancers and ranks third in cancer-related deaths across globe. Cancer cells are known to take advantage of the altered metabolic processes to sustain their survival, proliferation, and cancer progression. In this investigation, we explored the available genome-wide expression profiles of few hundreds of gastric tumors and non-cancerous gastric tissues and analyzed in the context of metabolic pathways. Gastric tumors were investigated for the metabolic processes related to glucose metabolism, glucose transport, glutamine metabolism, and fatty acid metabolism, by metabolic pathway-focused gene set enrichment analysis. Notably, all glucose metabolism and glutamine metabolism-related gene sets were found enriched in intestinal subtype gastric tumors. On the other hand, the gene sets related to glucose transport and glucan (glycan) metabolisms are enriched in diffuse subtype gastric tumors. Strikingly, fatty acid metabolisms, fatty acid transport, and fat differentiation-related signatures are also highly activated in diffuse subtype gastric tumors. Exploration of the recently established metabolome profile of the massive panel of cell lines also revealed the metabolites of glucose and fatty acid metabolic pathways to show the differing abundance across gastric cancer subtypes. The subtype-specific metabolic rewiring and the existence of two distinct metabolic dysregulations involving glucose and fatty acid metabolism in gastric cancer subtypes have been identified. The identified differing metabolic dysregulations would pave way for the development of targeted therapeutic strategies for the gastric cancer subtypes.


Subject(s)
Fatty Acids/metabolism , Glucose/metabolism , Metabolome/genetics , Stomach Neoplasms/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Metabolic Networks and Pathways/genetics , Stomach Neoplasms/classification , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
11.
Ann N Y Acad Sci ; 1467(1): 94-111, 2020 05.
Article in English | MEDLINE | ID: mdl-31944316

ABSTRACT

The identification of new drugs for the targeted therapy of gastric cancer remains an important need. The RAS/RAF/MEK/ERK/ELK1 signaling cascade is activated in many cancers, including gastric cancer. To identify the targetable inhibitors of the ERK/MAPK pathway, we performed a repurposing screening of a panel of antimicrobial agents in gastric cancer cells using an ERK/MAPK-driven firefly luciferase reporter assay. Multiple antibiotics were identified to inhibit ERK-mediated transcriptional activity. Among them, doxycycline showed high inhibition of ERK/MAPK-regulated transcriptional activity and the levels of ERK proteins. Doxycycline was further identified to inhibit the proliferation and the colony- and spheroid-forming potential of gastric cancer cells. By in vitro signaling pathway and genome-wide expression profiling analyses, doxycycline was identified to inhibit signaling pathways and transcriptional activities regulated by ER, Myc, E2F1, Wnt, SMAD2/3/4, Notch, and OCT4. Doxycycline was also found to activate p53-, ATF6-, NRF1/2-, and MTF1-mediated transcription and inhibit the transcription of histones, proteasomal genes, fibroblast growth factor, and other oncogenic factors. These observations show the multitargeting and targeted therapeutic features of doxycycline for a subset of gastric tumors.


Subject(s)
Adenocarcinoma/drug therapy , Anti-Bacterial Agents/pharmacology , Cell Proliferation/drug effects , Doxycycline/pharmacology , Signal Transduction/drug effects , Stomach Neoplasms/drug therapy , Adenocarcinoma/metabolism , Anti-Bacterial Agents/therapeutic use , Doxycycline/therapeutic use , Drug Screening Assays, Antitumor , Humans , MAP Kinase Signaling System/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Stomach Neoplasms/metabolism
12.
Antiviral Res ; 174: 104695, 2020 02.
Article in English | MEDLINE | ID: mdl-31846633

ABSTRACT

Yin Yang 1 (YY1) is a ubiquitous transcription factor with both transcriptional activating and repressing functions. Targeting YY1 is considered as a potential therapeutic strategy for several malignancies. Telomerase Reverse Transcriptase (TERT) is also considered as a potential target for cancer therapeutics. To enable the large-scale screening and identification of potential YY1 targeting drugs, a gastric cancer cell line-based drug screening assay was developed. In a YY1 targeted drug repurpose screen, abacavir sulfate, a nucleoside analog reverse transcriptase inhibitor, known to target TERT was identified to show the feature of activating YY1 mediated transcription. We further explored i) the molecular targets of abacavir, ii) activation pattern of pathways regulated by abacavir in gastric tumors, and iii) therapeutic potential of abacavir for gastric cancer cells. Oncogenic signaling pathways like MYC, HIF1-α, ERK, WNT, E2F, NFκB and NRF1/2 were also found to be highly activated by abacavir. Abacavir was found to have less impact on the viability of gastric cancer cells. Across gastric tumors, we observed the co-activation of TERT, alternative lengthening of telomere (ALT), DNA repair, and the oncogenic pathways MYC, E2F/DP1, ERK, YY1, HIF1α, and NFκB specific gene-sets, in a subset of gastric tumors. The observed connectivity among TERT, DNA repair, and multiple oncogenic pathways indicate the need for the development of combinatorial therapeutics for the gastric tumors with the activated TERT.


Subject(s)
Antineoplastic Agents/pharmacology , Dideoxynucleosides/pharmacology , Signal Transduction , Stomach Neoplasms/drug therapy , YY1 Transcription Factor/genetics , Cell Death/drug effects , Cell Line, Tumor , Drug Repositioning , Humans , Reverse Transcriptase Inhibitors/pharmacology , Telomerase/antagonists & inhibitors
14.
Cancer Immunol Immunother ; 66(7): 941-954, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28405765

ABSTRACT

The existing large number of gene expression profiles of tumour samples offers a great advantage for the integrative functional genomic exploration of molecular dysregulation in cancers. The clusters of genes (modules) derived from a gastric cancer (GC) coexpression network were explored to understand their clinical and functional significance. Among the modules derived from the GC mRNA expression network, six modules were relatively highly expressed in diffuse type gastric tumours. Elevated expression of genes related to extracellular matrix (ECM), angiogenesis, collagen and intracellular cytoskeletal components and immune response were identified in these modules. ECM-related modules exhibited an inverse correlation with modules representing the expression of immune response genes. A reduced expression of immune response genes was identified as the key factor associated with the aggressive features of diffuse gastric tumours, which is indicative of tumour progression involving the escape from immune surveillance in diffuse tumours. A part of the identified aggressive factors was common between intestinal and diffuse type tumours. The coexpressed modules and their expression patterns delineate the fine transition involved in cancer progression in the later stages of tumours. The identified modules could serve as surrogate gene-sets, indicating the molecular staging of GC aggressiveness with underlying biological interaction.


Subject(s)
Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Genes, Neoplasm , Immunity, Cellular/genetics , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Disease Progression , Gene Expression Profiling , Genetic Markers , Genome-Wide Association Study , Genotyping Techniques , Humans , Immunotherapy , Kaplan-Meier Estimate , Oligonucleotide Array Sequence Analysis , RNA, Messenger/biosynthesis , Statistics as Topic , Stomach Neoplasms/genetics , Transcription, Genetic , Transcriptome
15.
Cell Oncol (Dordr) ; 40(3): 219-233, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28390038

ABSTRACT

PURPOSE: Hepatocellular carcinoma (HCC) is one of the most common human malignancies. It has frequently been associated with metabolic perturbations and liver damages. Various members of the family of acyl-CoA synthetases are known to be involved in the production of bioactive fatty acids, and altered expression of its encoding genes has been found to be involved in metabolic perturbations. For the development of novel diagnostic and therapeutic HCC options, a fundamental understanding of the mechanisms associated with the deregulation of candidate genes involved in metabolic perturbation is required. METHODS: A meta-analysis of multiple HCC mRNA profiles was performed to identify consistently deregulated genes. Expression of the acyl-CoA synthetase medium chain family member 3 (ACSM3) gene was subsequently assessed in different HCC tumor stages and correlated with various clinicopathological features. Transcription regulation, survival and pathway-associated features of the ACSM3 gene were investigated using integrative functional genomic and molecular cell biological methods. RESULTS: We found that expression of the ACSM3 gene was significantly reduced in HCC tissues and was frequently downregulated in patients exhibiting high alpha-fetoprotein (AFP) levels, high alanine aminotransferase (ALT) levels, multiple nodules and large tumors. Loss of ACSM3 expression was found to correlate with advanced HCC stages and a poor survival. In addition, HNF4α was found to positively regulate the expression of the ACSM3 gene, while PPARγ was found to transcriptionally repress it. Downregulation of ACSM3 expression was perceived upon activation of the TGFß, WNT, AKT and MYC signalling pathways. In addition, we found that ACSM3 expression correlates with fatty acid oxidation in HCC. CONCLUSION: Our data provide evidence for a differential expression and regulation of the ACSM3 gene in HCC, and may lay a foundation for therapeutically targeting fatty acid metabolism in these tumors.


Subject(s)
Carcinoma, Hepatocellular/pathology , Coenzyme A Ligases/biosynthesis , Gene Expression Regulation, Neoplastic/genetics , Liver Neoplasms/pathology , Adult , Aged , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/genetics , Coenzyme A Ligases/genetics , Fatty Acids/metabolism , Female , Gene Expression Profiling , Humans , Liver Neoplasms/genetics , Male , Middle Aged , Oxidation-Reduction , Transcriptome
16.
Mol Carcinog ; 56(6): 1590-1602, 2017 06.
Article in English | MEDLINE | ID: mdl-28059478

ABSTRACT

Frequent amplification of 7q21-22 genomic region is known in gastric cancer. Multiple genes including SHFM1, MCM7, and COL1A2 were reported to be the potential cancer candidate genes of this 20 Mb amplicon. This amplicon has two polycistrionic miRNA clusters and in the present study, miR-106b-25 cluster located in intron-13 of MCM7 was identified to express in gastric tumors. Among the 7q21-22 candidate genes, SHFM1 and MCM7 are expressed in intestinal type gastric tumors, whereas COL1A2 is expressed in diffuse type gastric tumors. Across gastric tumors, miR-25 was identified to co-express with MCM7 and SHFM1. On the other hand, negative correlation was observed between miR-25 and COL1A2 expression. miR-25 originating from MCM7 was found capable of selectively targeting the adjacent gene COL1A2. Silencing of miR-25 was found capable of elevating the expression of COL1A2 and inhibiting E-cadherin expression, revealing the diffuse type gastric cancer suppressive role conferred by miR-25. miR-25 was also found to suppress p53, and activate c-Src revealing its intestinal type gastric cancer associated oncogenic functions. Genome-wide expression profiling upon miR-25 silencing reveals that miR-25 is capable of suppressing 40 genes which are co-expressed with COL1A2, involved in epithelial to mesenchymal transition and angiogenesis which are the typical diffuse type gastric cancer features. The results clearly demonstrate 7q21-22 amplification, MCM7, and its intronic miR-25 are the major molecular switches involved in the complex oncogenic circuits of gastric cancer.


Subject(s)
Collagen Type I/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Minichromosome Maintenance Complex Component 7/genetics , Stomach Neoplasms/genetics , Stomach/pathology , Cell Line, Tumor , Chromosomes, Human, Pair 7/genetics , Gastric Mucosa/metabolism , Gene Amplification , Humans , Introns , Stomach Neoplasms/pathology
17.
Mol Carcinog ; 56(4): 1239-1250, 2017 04.
Article in English | MEDLINE | ID: mdl-27805295

ABSTRACT

Genomic Copy Number Variations (CNV) and the associated gene signatures are useful for cancer prognosis, diagnosis, and targeted therapeutics. Earlier, 7q21-22 region was reported for frequent amplification in gastric cancer and potential candidate genes were identified. An analysis of the expression pattern of the 159 genes located in this amplicon revealed the consistent elevated expression of 21 genes in gastric tumors. These genes are closely arranged within the 20 Mb region, and they showed a bimodal expression pattern. SHFM1 and 14 other genes are expressed in intestinal type gastric tumors. COL1A2 and PCOLCE genes of this region are expressed in diffuse type gastric tumors. Similarly, genome-wide expression neighbors of SHFM1 and COL1A2 also showed mutually exclusive expression pattern, and stratify intestinal and diffuse type gastric tumors. The expression of COL1A2 gene-set is associated with poor prognosis, whereas the SHFM1 gene-set is associated with better prognosis among the gastric cancer patients. Despite being physical neighbors, the SHFM1 and COL1A2 genes express differentially in the two major clinical sub-types of gastric cancer in a mutually exclusive manner. The tight gene regulations operating between these juxtaposed genes deserve investigation to understand the molecular regulatory switch defining the determinants of the gastric cancer sub-types. © 2016 Wiley Periodicals, Inc.


Subject(s)
Chromosomes, Human, Pair 7/genetics , Gene Amplification , Gene Expression Regulation, Neoplastic , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Stomach/pathology , Collagen Type I/genetics , DNA Copy Number Variations , Gastric Mucosa/metabolism , Humans , Prognosis , Proteasome Endopeptidase Complex/genetics , Stomach Neoplasms/diagnosis , Stomach Neoplasms/epidemiology , Survival Analysis
18.
J Environ Pathol Toxicol Oncol ; 35(3): 249-262, 2016.
Article in English | MEDLINE | ID: mdl-27910780

ABSTRACT

Doxorubicin (DOX) is a well-known cytotoxic agent used extensively as a chemotherapeutic drug to eradicate a wide variety of human cancers. Reactive oxygen species (ROS)-mediated oxidative stress during DOX treatment can induce cardiac, renal, and hepatic toxicities, which can constrain its use as a potential cytotoxic agent. The present work investigates the antioxidant potential of naringenin (NAR) against DOXinduced toxicities of a Dalton's lymphoma ascites (DLA) tumor-bearing mouse model. Mice were randomized into four groups: a negative control, positive control, DOX (2.5 mg/kg) treated, and DOX (2.5 mg/kg) + NAR (50 mg/kg/d) treated. DOX administration significantly altered the levels of functional markers in blood and antioxidant enzymes in kidney, heart, lung, liver, spleen, and tumor tissues. These changes in antioxidant enzymes and successive lipid peroxidation were prevented by NAR supplementation, resulting in decreases in the risk of toxicity due to DOX therapy. Histopathology results and electron paramagnetic resonance imaging (EPRI) of the tumor microenvironment confirmed this evidence. Using EPRI, pharmacokinetics of the nitroxide, 3-carbamoyl-2,2,5,5-tetramethylpyrrolidine-1-oxyl (3-CP) was monitored intratumorally before and after chemotherapy. EPRI of the DOX + NAR-treated mouse model showed reduced tumor size with significant modification of the hypoxic condition inside the tumor microenvironment. Consequently, these findings suggest that NAR treatment significantly reduces DOX-induced toxicity and the hypoxic condition in a DLA tumor-bearing mouse model.


Subject(s)
Antioxidants/pharmacology , Carcinoma, Ehrlich Tumor/drug therapy , Doxorubicin/antagonists & inhibitors , Doxorubicin/toxicity , Flavanones/pharmacology , Hypoxia/drug therapy , Animals , Antibiotics, Antineoplastic/toxicity , Electron Spin Resonance Spectroscopy , Enzyme Activation/drug effects , Lipid Peroxidation/drug effects , Mice , Oxidoreductases/metabolism , Pyrrolidines/pharmacokinetics , Random Allocation
19.
Cell Oncol (Dordr) ; 39(6): 523-536, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27562587

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is the fifth most common cancer and the third leading cause of cancer death worldwide. It is a heterogeneous disorder and >80 % of the tumors develop in patients with liver cirrhosis, resulting from chronic inflammation and/or fibrosis. Here, we set out to identify novel targets for HCC therapy and to define a subgroup of patients that might benefit most from it. METHODS: Cellular pathway activation profiling of 45 transcription factors in a HCC-derived cell line (HEP3B), in vitro analysis of NFκB reporter activity in additional HCC-derived cell lines and pathway-focused integrative analyses of publicly available primary HCC-derived expression profiling data (GSE6764, GSE9843, E-TABM-36 and E-TABM-292) were employed to reveal a role of NFκB in HCC development. In order to identify potential targeting agents, a luciferase-based NFκB reporter screening assay was established in HEP3B cells. After screening of a drug library through this assay, a potent NFκB pathway inhibitor was identified and characterized using an array of additional in vitro assays. RESULTS: Using cellular pathway activation profiling, we found a high activation of NFκB-mediated signaling in HCC-derived cell lines and in primary HCC tumors. Through NFκB inhibitor screening we observed a highly efficacious NFκB pathway inhibitory potential of ornithogalum in HCC-derived HEP3B cells. Although its active component still remains to be defined, ornithogalum has been found to inhibit endoplasmic reticulum (ER) and oxidative stress responses. ER stress, oxidative stress and NFκB signaling were found to be enhanced in a subset of HCCs, as well as in (precancerous) liver cirrhosis tissues. CONCLUSION: From our data we conclude that NFκB signaling is activated in precancerous cirrhosis tissues and in a subset of HCCs. We found that ornithogalum exhibits NFκB targeting and stress relieving activities. NFκB inhibitors, including the active component of ornithogalum, may serve as putative preventive and targeted therapeutic agents for at least a subset of HCCs in which the NFκB pathway is activated. These latter notions require further investigation in a translational context.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Molecular Targeted Therapy/methods , NF-kappa B/metabolism , Antineoplastic Agents/pharmacology , Blotting, Western , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Gene Knockdown Techniques , Humans , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Neoplasms/pathology , Ornithogalum , Precancerous Conditions/metabolism , Precancerous Conditions/pathology
20.
Genes Cells ; 21(9): 949-65, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27465470

ABSTRACT

Dysregulated molecular processes are the major factors that drive and feed the signaling processes involved in carcinogenesis. In recent years, regulation of mRNAs by microRNAs (miRNAs) has been found to play a vital role in many cancers including hepatocellular carcinoma (HCC). However, genomewide studies defining molecular regulatory circuits at both mRNA and miRNA levels are just emerging. To uncover the molecular and functional processes involved in liver tumorigenesis at mRNA and miRNA level, a co-expression-based network of miRNAs was constructed from multiple miRNA profiles. The applicability of the network approach to microRNA expression profiles was assessed. Although the clustering consistency of miRNAs across the profiles was found moderate, miRNA networking has been found informative. Furthermore, microRNA network modules were integrated with the functionally defined mRNA modules derived from an mRNA co-expression network of an earlier study. Three highly clustered regulatory circuits of mRNA-miRNA modules have been identified as involved in hepatocyte, inflammatory-stress and proliferative process activated subcategories of HCC. A subset of the proliferative miRNA module was found clustered in the 14q32.31 chromosomal region. The current integrative network analysis of mRNA-miRNA modules shows the intricate miRNA-mRNA functional circuits and signaling interactions involved in liver tumorigenesis.


Subject(s)
Carcinoma, Hepatocellular/genetics , Gene Regulatory Networks , Liver Neoplasms/genetics , MicroRNAs/genetics , RNA, Messenger/genetics , Carcinogenesis/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Computational Biology/methods , Gene Expression Profiling , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , MicroRNAs/biosynthesis , MicroRNAs/metabolism , Neoplasm Staging , RNA, Messenger/biosynthesis , RNA, Messenger/metabolism , Signal Transduction , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL
...