Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Biomol Ther (Seoul) ; 32(1): 123-135, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38148558

ABSTRACT

Although gemcitabine-based regimens are widely used as an effective treatment for pancreatic cancer, acquired resistance to gemcitabine has become an increasingly common problem. Therefore, a novel therapeutic strategy to treat gemcitabine-resistant pancreatic cancer is urgently required. Piceamycin has been reported to exhibit antiproliferative activity against various cancer cells; however, its underlying molecular mechanism for anticancer activity in pancreatic cancer cells remains unexplored. Therefore, the present study evaluated the antiproliferation activity of piceamycin in a gemcitabine-resistant pancreatic cancer cell line and patient-derived pancreatic cancer organoids. Piceamycin effectively inhibited the proliferation and suppressed the expression of alpha-actinin-4, a gene that plays a pivotal role in tumorigenesis and metastasis of various cancers, in gemcitabine-resistant cells. Long-term exposure to piceamycin induced cell cycle arrest at the G0/G1 phase and caused apoptosis. Piceamycin also inhibited the invasion and migration of gemcitabine-resistant cells by modulating focal adhesion and epithelial-mesenchymal transition biomarkers. Moreover, the combination of piceamycin and gemcitabine exhibited a synergistic antiproliferative activity in gemcitabine-resistant cells. Piceamycin also effectively inhibited patient-derived pancreatic cancer organoid growth and induced apoptosis in the organoids. Taken together, these findings demonstrate that piceamycin may be an effective agent for overcoming gemcitabine resistance in pancreatic cancer.

2.
Cancers (Basel) ; 15(9)2023 May 07.
Article in English | MEDLINE | ID: mdl-37174108

ABSTRACT

Krukovine (KV) is an alkaloid isolated from the bark of Abuta grandifolia (Mart.) Sandw. (Menispermaceae) with anticancer potential in some cancers with KRAS mutations. In this study, we explored the anticancer efficacy and mechanism of KV in oxaliplatin-resistant pancreatic cancer cells and patient-derived pancreatic cancer organoids (PDPCOs) with KRAS mutation. After treatment with KV, mRNA and protein levels were determined by RNA-seq and Western blotting, respectively. Cell proliferation, migration, and invasion were measured by MTT, scratch wound healing assay, and transwell analysis, respectively. Patient-derived pancreatic cancer organoids (PDPCOs) with KRAS mutations were treated with KV, oxaliplatin (OXA), and a combination of KV and OXA. KV suppresses tumor progression via the downregulation of the Erk-RPS6K-TMEM139 and PI3K-Akt-mTOR pathways in oxaliplatin-resistant AsPC-1 cells. Furthermore, KV showed an antiproliferative effect in PDPCOs, and the combination of OXA and KV inhibited PDPCO growth more effectively than either drug alone.

3.
Exp Mol Med ; 54(6): 739-752, 2022 06.
Article in English | MEDLINE | ID: mdl-35662287

ABSTRACT

Liver fibrosis occurs during wound healing after repeated liver injury and is characterized by extensive extracellular matrix deposition. We previously identified hyaluronan synthase 2 (HAS2) as a driver of liver fibrosis and hepatic stellate cell (HSC) activation. Developing strategies to suppress HSC activation is key to alleviating liver fibrosis, and HAS2 is an attractive candidate for intervention. To gain insight into the molecular function of HAS2, we investigated its posttranscriptional regulation. We found that miR-200c directly targets the 3' untranslated regions of HAS2. Moreover, miR-200c and HAS2 were inversely expressed in fibrotic human and mouse livers. After establishing the direct interaction between miR-200c and HAS2, we investigated the functional outcome of regulating HAS2 expression in three murine models: CCl4-induced acute liver injury, CCl4-induced chronic liver fibrosis, and bile duct ligation-induced liver fibrosis. Hepatic Has2 expression was induced by acute and chronic CCl4 treatment. In contrast, miR-200c expression was decreased after CCl4 treatment. HSC-specific Has2 deletion reduced the expression of inflammatory markers and infiltration of macrophages in the models. Importantly, hyaluronidase-2 (HYAL2) but not HYAL1 was overexpressed in fibrotic human and murine livers. HYAL2 is an enzyme that can cleave the extracellular matrix component hyaluronan. We found that low-molecular-weight hyaluronan stimulated the expression of inflammatory genes. Treatment with the HA synthesis inhibitor 4-methylumbelliferone alleviated bile duct ligation-induced expression of these inflammatory markers. Collectively, our results suggest that HAS2 is negatively regulated by miR-200c and contributes to the development of acute liver injury and chronic liver inflammation via hyaluronan-mediated immune signaling.


Subject(s)
Hyaluronan Synthases , Liver Cirrhosis , MicroRNAs , Animals , Carbon Tetrachloride/adverse effects , Chemokine CCL3/metabolism , Chemokine CCL4/metabolism , Fibrosis , Hepatic Stellate Cells/metabolism , Hyaluronan Synthases/genetics , Hyaluronan Synthases/metabolism , Hyaluronic Acid/adverse effects , Hyaluronic Acid/metabolism , Inflammation/metabolism , Liver Cirrhosis/chemically induced , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism
4.
Gut Liver ; 16(4): 625-636, 2022 07 15.
Article in English | MEDLINE | ID: mdl-34916338

ABSTRACT

Background/Aims: Three-dimensional cultures of human pancreatic cancer tissue also known as "organoids" have largely been developed from surgical specimens. Given that most patients present with locally advanced and/or metastatic disease, such organoids are not representative of the majority of patients. Therefore, we used endoscopic ultrasound-guided fine-needle aspiration (EUS-FNA) to collect pancreatic cancer tissues from patients with advanced pancreatic cancer to create organoids, and evaluated their utility in pancreatic cancer research. Methods: Single-pass EUS-FNA samplings were employed to obtain the tissue for organoid generation. After establishment of the organoid, we compared the core biopsy tissues with organoids using hematoxylin and eosin staining, and performed whole exome sequencing (WES) to detect mutational variants. Furthermore, we compared patient outcome with the organoid drug response to determine the potential utility of the clinical application of such organoid-based assays. Results: Organoids were successfully generated in 14 of 20 tumors (70%) and were able to be passaged greater than 5 times in 12 of 20 tumors (60%). Among them, we selected eight pairs of organoid and core biopsy tissues for detailed analyses. They showed similar patterns in hematoxylin and eosin staining. WES revealed mutations in KRAS, TP53, CDKN2A, SMAD4, BRCA1, and BRCA2 which were 93% homologous, and the mean nonreference discordance rate was 5.47%. We observed moderate drug response correlations between the organoids and clinical outcomes in patients who underwent FOLFIRINOX chemotherapy. Conclusions: The established organoids from EUS-FNA core biopsies can be used for a suitable model system for pancreatic cancer research.


Subject(s)
Endoscopic Ultrasound-Guided Fine Needle Aspiration , Pancreatic Neoplasms , Antineoplastic Combined Chemotherapy Protocols , Endoscopic Ultrasound-Guided Fine Needle Aspiration/methods , Eosine Yellowish-(YS) , Hematoxylin , Humans , Organoids/pathology , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms
5.
Biochem Biophys Res Commun ; 430(3): 1109-13, 2013 Jan 18.
Article in English | MEDLINE | ID: mdl-23261432

ABSTRACT

The cytoprotective stress response factor HSF1 regulates the transcription of the chaperone HSP70, which exhibits anti-inflammatory effects and improves insulin sensitivity. We tested the therapeutic potential of this pathway in rodent models of diabetes using pharmacological tools. Activation of the HSF1 pathway was achieved using potent inhibitors of the upstream regulatory protein, HSP90. Treatment with AUY922, a selective HSP90 inhibitor led to robust inhibition of JNK1 phosphorylation, cytoprotection and improved insulin signaling in cells, consistent with effects observed with HSP70 treatment. Chronic dosing with HSP90 inhibitors reversed hyperglycemia in the diabetic db/db mouse model, and improved insulin sensitivity in the diet-induced obese mouse model of insulin resistance, further supporting the concept that the HSF1 pathway is a potentially viable anti-diabetes target.


Subject(s)
Blood Glucose/drug effects , DNA-Binding Proteins/agonists , Diabetes Mellitus, Type 2/drug therapy , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Hypoglycemic Agents/administration & dosage , Isoxazoles/administration & dosage , Resorcinols/administration & dosage , Transcription Factors/agonists , Animals , Benzoquinones/pharmacology , Blood Glucose/metabolism , Cells, Cultured , Cytoprotection , Diabetes Mellitus, Type 2/metabolism , Heat Shock Transcription Factors , Heat-Shock Response , Isoxazoles/chemistry , Lactams, Macrocyclic/pharmacology , Male , Mice , Mice, Inbred Strains , Myoblasts/drug effects , Myoblasts/metabolism , Resorcinols/chemistry
6.
J Neurosci Res ; 86(6): 1353-60, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18183617

ABSTRACT

Retinoic acid (RA) is a well-known antiinflammatory agent. In this study, we show that RA has a dual effect on cyclooxygenase-2 (COX-2) expression in inflammatory activated microglia, the resident brain macrophages. After treatment of microglia with LPS or thrombin, COX-2 expression was induced in two phases, specifically, an initial increase at about 12 hr after stimulation followed by a decrease, and another increase at about 48-72 hr. However, PGE(2) and 15d-PGJ(2) were detected at about 12 hr, and the levels continuously increased thereafter. Interestingly, all-trans retinoic acid (ATRA) suppressed the expression of early-phase COX-2 but augmented late-phase COX-2 and inhibited iNOS in the whole time sequence. ATRA enhanced PGE(2) production but had little effect on 15d-PGJ(2). Moreover, ATRA selectively up-regulated the expression of a PGE(2) synthase, mPGES-1, but had little effect on the PGD(2) synthase, H-PGDS. The results collectively suggest that ATRA modulates microglial responses to inflammatory stimulators, particularly at the late phase, via enhancement of COX-2 expression and PGE(2) production.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cyclooxygenase 2/biosynthesis , Dinoprostone/biosynthesis , Intramolecular Oxidoreductases/biosynthesis , Microglia/drug effects , Tretinoin/pharmacology , Animals , Blotting, Western , Brain/drug effects , Brain/metabolism , Cells, Cultured , Cyclooxygenase 2/drug effects , Enzyme-Linked Immunosorbent Assay , Inflammation/chemically induced , Lipopolysaccharides/toxicity , Microglia/metabolism , Microsomes/metabolism , Prostaglandin-E Synthases , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Thrombin/toxicity
7.
BMC Mol Biol ; 8: 46, 2007 Jun 05.
Article in English | MEDLINE | ID: mdl-17550601

ABSTRACT

BACKGROUND: Using a gene clustering strategy we determined intracellular pathway relationships within skeletal myotubes in response to an acute heat stress stimuli. Following heat shock, the transcriptome was analyzed by microarray in a temporal fashion to characterize the dynamic relationship of signaling pathways. RESULTS: Bioinformatics analyses exposed coordination of functionally-related gene sets, depicting mechanism-based responses to heat shock. Protein turnover-related pathways were significantly affected including protein folding, pre-mRNA processing, mRNA splicing, proteolysis and proteasome-related pathways. Many responses were transient, tending to normalize within 24 hours. CONCLUSION: In summary, we show that the transcriptional response to acute cell stress is largely transient and proteosome-centric.


Subject(s)
Gene Expression Regulation , Heat Stress Disorders , Multigene Family , Animals , Cell Line , Gene Expression Profiling , Mice , Muscle Fibers, Skeletal/physiology , Oligonucleotide Array Sequence Analysis , Proteome/analysis , Signal Transduction/physiology , Transcription, Genetic
8.
Mol Cancer ; 6: 7, 2007 Jan 18.
Article in English | MEDLINE | ID: mdl-17233903

ABSTRACT

BACKGROUND: Human mammary epithelial cells (HMEC) overcome two well-characterized genetic and epigenetic barriers as they progress from primary cells to fully immortalized cell lines in vitro. Finite lifespan HMEC overcome an Rb-mediated stress-associated senescence barrier (stasis), and a stringent, telomere-length dependent, barrier (agonescence or crisis, depending on p53 status). HMEC that have overcome the second senescence barrier are immortalized. METHODS: We have characterized pre-stasis, post-selection (post-stasis, with p16 silenced), and fully immortalized HMEC by transcription profiling and RT-PCR. Four pre-stasis and seven post-selection HMEC samples, along with 10 representatives of fully immortalized breast epithelial cell lines, were profiled using Affymetrix U133A/B chips and compared using both supervised and unsupervised clustering. Datasets were validated by RT-PCR for a select set of genes. Quantitative immunofluorescence was used to assess changes in transcriptional regulators associated with the gene expression changes. RESULTS: The most dramatic and uniform changes we observed were in a set of about 30 genes that are characterized as a "cancer proliferation cluster," which includes genes expressed during mitosis (CDC2, CDC25, MCM2, PLK1) and following DNA damage. The increased expression of these genes was particularly concordant in the fully immortalized lines. Additional changes were observed in IFN-regulated genes in some post-selection and fully immortalized cultures. Nuclear localization was observed for several transcriptional regulators associated with expression of these genes in post-selection and immortalized HMEC, including Rb, Myc, BRCA1, HDAC3 and SP1. CONCLUSION: Gene expression profiles and cytological changes in related transcriptional regulators indicate that immortalized HMEC resemble non-invasive breast cancers, such as ductal and lobular carcinomas in situ, and are strikingly distinct from finite-lifespan HMEC, particularly with regard to genes involved in proliferation, cell cycle regulation, chromosome structure and the DNA damage response. The comparison of HMEC profiles with lines harboring oncogenic changes (e.g. overexpression of Her-2neu, loss of p53 expression) identifies genes involved in tissue remodeling as well as proinflamatory cytokines and S100 proteins. Studies on carcinogenesis using immortalized cell lines as starting points or "normal" controls need to account for the significant pre-existing genetic and epigenetic changes inherent in such lines before results can be broadly interpreted.


Subject(s)
Breast Neoplasms/genetics , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Mammary Glands, Human/metabolism , Transcription, Genetic , Cell Nucleus/metabolism , Cluster Analysis , Epithelial Cells/physiology , Gene Expression Profiling , Humans , Mammary Glands, Human/cytology , Polymerase Chain Reaction/methods , Promoter Regions, Genetic , Receptor, ErbB-2/metabolism , Regulatory Elements, Transcriptional , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
9.
J Biotechnol ; 127(3): 355-60, 2007 Jan 10.
Article in English | MEDLINE | ID: mdl-16949174

ABSTRACT

Bio-cell chip is a chip that has hundreds of types of cells arrayed and immobilized on a small slide. To elucidate the role of deletion of the p16 gene in hematologic malignancies, the bio-cell chip technique was applied to fluorescent in situ hybridization (FISH) study. We made a bio-cell chip with bone marrow specimen from 109 patients with acute lymphoblastic leukemia (ALL), 102 patients with acute myelogenous leukemia (AML), 47 patients with chronic myelogenous leukemia (CML), and 25 patients with multiple myeloma (MM). A glass slide with 96 separated areas was fabricated, onto which was added methanol/acetic acid fixed cell suspensions for high-throughput FISH for p16. With the successful application of bio-cell chip technique, we found that the deletion of p16 contributed to the oncogenesis in acute leukemia, but not in chronic leukemia. In conclusion, the bio-cell chip, a cell version of ultrahigh-throughput technology, was successfully applied to the FISH study, which can be utilized efficiently in the molecular cytogenetic investigation of hematologic malignancies.


Subject(s)
Gene Deletion , Genes, p16 , In Situ Hybridization, Fluorescence , Leukemia/genetics , Microarray Analysis , Multiple Myeloma/genetics , Acute Disease , Chronic Disease , Cytogenetic Analysis/methods , Female , Humans , Leukemia/diagnosis , Male , Multiple Myeloma/diagnosis , Predictive Value of Tests
10.
Biochem Biophys Res Commun ; 341(3): 882-8, 2006 Mar 17.
Article in English | MEDLINE | ID: mdl-16442498

ABSTRACT

Here, we enriched a human cell population from adipose tissue that exhibited both mesenchymal plasticity, self-renewal capacity, and a cell-surface marker profile indistinguishable from that of bone marrow-derived mesenchymal stem cells. In addition to adipogenic and osteogenic differentiation, these adipose-derived stem cells displayed skeletal myogenic potential when co-cultured with mouse skeletal myocytes in reduced serum conditions. Physical incorporation of stem cells into multinucleated skeletal myotubes was determined by genetic lineage tracing, whereas human-specific antibody staining was employed to demonstrate functional contribution of the stem cells to a myogenic lineage. To investigate the effects of hypoxia, cells were maintained and differentiated at 2% O(2). In contrast with reports on bone marrow-derived stem cells, both osteogenic and adipogenic differentiation were significantly attenuated. In summary, the relative accessibility of adipose-derived mesenchymal stem cells from human donors provides opportunity for molecular investigation of mechanistic dysfunction in disease settings and may introduce new prospects for cell-based therapy.


Subject(s)
Adipocytes/cytology , Cell Differentiation , Cell Hypoxia/physiology , Stem Cells/cytology , Stem Cells/physiology , Animals , Antigens/metabolism , Cell Line , Cell Lineage , Coculture Techniques , Humans , Mice , Muscle Development , Osteogenesis
11.
Genomics ; 87(1): 129-38, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16300922

ABSTRACT

Stem cell differentiation is governed by extracellular signals that activate intracellular networks (or pathways) to drive phenotypic specification. Using a novel gene clustering strategy we determined pathway relationships from a genome-wide transcriptional dataset of skeletal myoblast differentiation. Established myogenic pathways, including cell contractility and cell-cycle arrest, were predicted with extreme statistical significance (p approximately 0). In addition, gene sets associated with angiogenesis, neuronal activity, and mRNA splicing were regulated, exposing developmental and therapeutic implications. Acquisition of transcriptional data spanning the entire differentiation time course provided context for a dynamic landscape of functional pathway regulation. This novel perspective on myogenic cell differentiation revealed previously unrecognized patterns of regulation. We predict that similar analyses will facilitate ongoing efforts to define molecular mechanisms in other stem cell and developmental paradigms. Finally, by combining an iterative process of analysis with supplementation of novel pathways, this application may evolve into a powerful discovery tool.


Subject(s)
Cell Differentiation/physiology , Gene Expression Regulation/physiology , Muscle Development/genetics , Myoblasts/physiology , Signal Transduction/physiology , Stem Cells/physiology , Animals , Cell Cycle/genetics , Gene Expression Profiling/methods , Mice , Muscle Contraction/physiology , Myoblasts/cytology , Neovascularization, Physiologic/genetics , Neurons/cytology , Neurons/physiology , Oligonucleotide Array Sequence Analysis/methods , Stem Cells/cytology
12.
Cancer Genet Cytogenet ; 162(2): 176-8, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16213368

ABSTRACT

TEL/AML1 (also known as ETV6/RUNX1) rearrangement is the most frequent genetic change in childhood B-acute lymphoblastic leukemia (ALL) and is associated with a favorable prognosis. Its presence in adult ALL, however, has been questionable, and any association between TEL/AML1 rearrangement and clinical prognosis is unknown. To reveal the presence and incidence of the TEL/AML1 rearrangement in adult ALL, we applied fluorescence in situ hybridization (FISH). We conducted extra-signal, dual-color fluorescence in situ hybridization (ES-FISH) for TEL/AML1 rearrangement on bone marrow cells from 74 adult ALL patients and analyzed the survival time. We demonstrated the TEL/AML1 rearrangement in 2 patients (2.7%) among 74 patients with ALL. Of 74 adult ALL patients, 3 (4.0%) showed deletion of the TEL gene without TEL/AML1 rearrangement. The mean survival time of patients with TEL/AML1+ALL and patients with cryptic TEL deletion was 33 and 5 months, respectively, whereas the mean survival time of 53 TEL/AML1-ALL patients was 16.7 months. TEL/AML1 rearrangement is not unique in childhood ALL, and cryptic TEL deletion without TEL/AML1 rearrangement was more frequent than the TEL/AML1 rearrangement in adult ALL. We recommend TEL/AML1 FISH in adult ALL patients because conventional cytogenetic studies so far have yielded uninformative results.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Gene Deletion , Gene Rearrangement , Oncogene Proteins, Fusion/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins c-ets/genetics , Repressor Proteins/genetics , Adult , Aged , Female , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , ETS Translocation Variant 6 Protein
13.
Exp Cell Res ; 307(1): 174-82, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15922737

ABSTRACT

Much attention is focused on characterizing the contribution of bone marrow (BM)-derived cells to regenerating skeletal muscle, fuelled by hopes for stem cell-mediated therapy of muscle degenerative diseases. Though physical integration of BM stem cells has been well documented, little evidence of functional commitment to myotube phenotype has been reported. This is due to the innate difficulty in distinguishing gene products derived from donor versus host nuclei. Here, we demonstrate that BM-derived stem cells contribute via gene expression following incorporation to skeletal myotubes. By co-culturing human BM-derived mesenchymal stem cells (MSC) with mouse skeletal myoblasts, physical incorporation was observed by genetic lineage tracing and species-specific immunofluorescence. We used a human-specific antibody against the intermediate filament protein nestin, a marker of regenerating skeletal muscle, to identify functional contribution of MSC to myotube formation. Although nestin expression was never detected in MSC, human-specific expression was detected in myotubes that also contained MSC-derived nuclei. This induction of gene expression following myotube integration suggests that bone marrow-derived stem cells can reprogram and functionally contribute to the muscle cell phenotype. We propose that this model of myogenic commitment may provide the means to further characterize functional reprogramming of MSC to skeletal muscle.


Subject(s)
Bone Marrow Cells/cytology , Gene Expression Regulation , Mesenchymal Stem Cells/cytology , Muscle Development , Muscle Fibers, Skeletal/cytology , Adipocytes/cytology , Animals , Biomarkers , Cell Culture Techniques , Cell Differentiation , Cell Fusion , Cell Line , Cell Lineage , Cell Nucleus/metabolism , Cell Separation/methods , Coculture Techniques , Endothelium, Vascular/cytology , Fluorescent Antibody Technique , Green Fluorescent Proteins/metabolism , Humans , Intermediate Filament Proteins/metabolism , Mice , Microscopy, Fluorescence , Models, Biological , Myoblasts/cytology , Neovascularization, Physiologic , Nerve Tissue Proteins/metabolism , Nestin , Osteocytes/cytology , Species Specificity , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...