Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Hum Pathol ; 88: 48-59, 2019 06.
Article in English | MEDLINE | ID: mdl-30946934

ABSTRACT

Some rhabdomyosarcomas and sarcomatoid carcinomas with heterologous rhabdomyosarcomatous elements resemble high-grade neuroendocrine carcinoma, creating a diagnostic difficulty. The purpose of this study was to characterize the overlap of adult genitourinary rhabdomyosarcomas, excluding those occurring at paratesticular sites, with high-grade neuroendocrine carcinoma and identify features helpful in their separation. Seventeen cases of rhabdomyosarcoma (11 from the urinary bladder and 3 each from kidney and prostate) were compared to 10 cases of high-grade neuroendocrine carcinoma from the urinary bladder. These tumors were analyzed by immunohistochemistry for desmin, MyoD1, myogenin, chromogranin, synaptophysin, CD56, TTF1, and ASCL1, and RNA sequencing was performed on 4 cases of bladder rhabdomyosarcoma (2 rhabdomyosarcomas and 2 sarcomatoid-rhabdomyosarcoma) and 10 cases of bladder high-grade neuroendocrine carcinoma. This was compared to public data from 414 typical urothelial carcinomas from The Cancer Genome Atlas dataset. Morphologic and immunophenotypic overlap with high-grade neuroendocrine carcinoma was seen in half of the bladder tumors, which included 4 rhabdomyosarcomas and 2 sarcomatoid rhabdomyosarcomas. RNA sequencing confirmed expression of neuroendocrine markers in these cases (2 rhabdomyosarcomas and 2 sarcomatoid rhabdomyosarcomas). Differential neuroendocrine differentiation was highlighted by ASCL1 protein expression only in high-grade neuroendocrine carcinoma. Moreover, both a pure alveolar rhabdomyosarcoma and sarcomatoid rhabdomyosarcoma of the urinary bladder demonstrated a fusion involving PPP1R12A. In summary, adult rhabdomyosarcomas of the urinary bladder are molecularly distinct from high-grade neuroendocrine carcinomas based on specific patterns of expression of myogenic and epithelial to mesenchymal transition-related transcription factors as well as the presence of a novel PPP1R12A fusion which is seen in a subset of cases.


Subject(s)
Carcinoma, Neuroendocrine/genetics , Gene Fusion/genetics , Myosin-Light-Chain Phosphatase/genetics , Rhabdomyosarcoma/genetics , Urinary Bladder Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Basic Helix-Loop-Helix Transcription Factors/genetics , Biomarkers, Tumor/analysis , Diagnosis, Differential , Female , Gene Expression , Humans , Kidney Neoplasms/genetics , Male , Middle Aged , Muscle Development/genetics , Prostatic Neoplasms/genetics
2.
Integr Biol (Camb) ; 8(12): 1221-1231, 2016 12 05.
Article in English | MEDLINE | ID: mdl-27796394

ABSTRACT

Carcinoma progression is influenced by interactions between epithelial tumor cells and components of their microenvironment. In particular, cell-extracellular matrix (ECM) interactions are known to drive tumor growth, metastatic potential, and sensitivity or resistance to therapy. Yet the intrinsic complexity of ECM composition within the tumor microenvironment remains a barrier to comprehensive investigation of these interactions. We present here a high-throughput cell microarray-based approach to study the impact of defined combinations of ECM proteins on tumor cell drug responses. Using this approach, we quantitatively evaluated the effects of 55 different ECM environments representing all single and two-factor combinations of 10 ECM proteins on the responses of lung adenocarcinoma cells to a selection of cancer-relevant small molecule drugs. This drug panel consisted of an alkylating agent and five receptor tyrosine kinase inhibitors. We further determined that expression of the neuroendocrine transcription factor ASCL1, which has been previously associated with poor patient outcome when co-expressed with the RET oncogene, altered cell responses to drugs and modulated cleavage of the pro-apoptotic protein caspase-3 depending on ECM context. Our results suggest that co-expression of specific ECM proteins with known genetic drivers in lung adenocarcinoma may impact therapeutic efficacy. Furthermore, this approach could be utilized to define the molecular mechanisms by which cell-matrix interactions drive drug resistance through integration with clinical cell samples and genomics data.


Subject(s)
Biomarkers, Tumor/metabolism , Drug Screening Assays, Antitumor/instrumentation , Extracellular Matrix Proteins/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , A549 Cells , Antineoplastic Agents/therapeutic use , Drug Screening Assays, Antitumor/methods , Equipment Design , Equipment Failure Analysis , Genome , High-Throughput Screening Assays/instrumentation , High-Throughput Screening Assays/methods , Humans , Protein Array Analysis/instrumentation , Protein Array Analysis/methods , Tissue Array Analysis/instrumentation , Tissue Array Analysis/methods
3.
BMC Res Notes ; 7: 601, 2014 Sep 03.
Article in English | MEDLINE | ID: mdl-25187308

ABSTRACT

BACKGROUND: Fusarochromanone (FC101) is a small molecule fungal metabolite with a host of interesting biological functions, including very potent anti-angiogenic and direct anti-cancer activity. RESULTS: Herein, we report that FC101 exhibits very potent in-vitro growth inhibitory effects (IC50 ranging from 10nM-2.5 µM) against HaCat (pre-malignant skin), P9-WT (malignant skin), MCF-7 (low malignant breast), MDA-231 (malignant breast), SV-HUC (premalignant bladder), UM-UC14 (malignant bladder), and PC3 (malignant prostate) in a time-course and dose-dependent manner, with the UM-UC14 cells being the most sensitive. FC101 induces apoptosis and an increase in proportion of cells in the sub-G1 phase in both HaCat and P9-WT cell lines as evidenced by cell cycle profile analysis. In a mouse xenograft SCC tumor model, FC101 was well tolerated, non-toxic, and achieved a 30% reduction in tumor size at a dose of 8 mg/kg/day. FC101 is also a potent anti-angiogenenic agent. At nanomolar doses, FC101 inhibits the vascular endothelial growth factor-A (VEGF-A)-mediated proliferation of endothelial cells. CONCLUSIONS: Our data presented here indicates that FC101 is an excellent lead candidate for a small molecule anti-cancer agent that simultaneously affects angiogenesis signaling, cancer signal transduction, and apoptosis. Further understanding of the underlying FC101's molecular mechanism may lead to the design of novel targeted and selective therapeutics, both of which are pursued targets in cancer drug discovery.


Subject(s)
Antineoplastic Agents/pharmacology , Chromones/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation , Drug Screening Assays, Antitumor , Humans , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/physiology
4.
Bioorg Med Chem ; 22(4): 1412-20, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24457089

ABSTRACT

Multidrug-resistance is a major cause of cancer chemotherapy failure in clinical treatment. Evidence shows that multidrug-resistant cancer cells are as sensitive as corresponding regular cancer cells under the exposure to anticancer ceramide analogs. In this work we designed five new ceramide analogs with different backbones, in order to test the hypothesis that extending the conjugated system in ceramide analogs would lead to an increase of their anticancer activity and selectivity towards resistant cancer cells. The analogs with the 3-ketone-4,6-diene backbone show the highest apoptosis-inducing efficacy. The most potent compound, analog 406, possesses higher pro-apoptotic activity in chemo-resistant cell lines MCF-7TN-R and NCI/ADR-RES than the corresponding chemo-sensitive cell lines MCF-7 and OVCAR-8, respectively. However, this compound shows the same potency in inhibiting the growth of another pair of chemo-sensitive and chemo-resistant cancer cells, MCF-7 and MCF-7/Dox. Mechanism investigations indicate that analog 406 can induce apoptosis in chemo-resistant cancer cells through the mitochondrial pathway. Cellular glucosylceramide synthase assay shows that analog 406 does not interrupt glucosylceramide synthase in chemo-resistant cancer cell NCI/ADR-RES. These findings suggest that due to certain intrinsic properties, ceramide analogs' pro-apoptotic activity is not disrupted by the normal drug-resistance mechanisms, leading to their potential use for overcoming cancer multidrug-resistance.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzeneacetamides/chemistry , Ceramides/chemistry , Ceramides/pharmacology , Ketones/chemistry , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Benzeneacetamides/chemical synthesis , Benzeneacetamides/pharmacology , Cell Line, Tumor , Ceramides/chemical synthesis , Drug Resistance, Neoplasm/drug effects , Glucosyltransferases/antagonists & inhibitors , Glucosyltransferases/metabolism , Humans , Isomerism , MCF-7 Cells , Molecular Conformation
5.
J Biol Chem ; 287(44): 37195-205, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-22936806

ABSTRACT

Cancer stem cells are distinguished from normal adult stem cells by their stemness without tissue homeostasis control. Glycosphingolipids (GSLs), particularly globo-series GSLs, are important markers of undifferentiated embryonic stem cells, but little is known about whether or not ceramide glycosylation, which controls glycosphingolipid synthesis, plays a role in modulating stem cells. Here, we report that ceramide glycosylation catalyzed by glucosylceramide synthase, which is enhanced in breast cancer stem cells (BCSCs) but not in normal mammary epithelial stem cells, maintains tumorous pluripotency of BCSCs. Enhanced ceramide glycosylation and globotriosylceramide (Gb3) correlate well with the numbers of BCSCs in breast cancer cell lines. In BCSCs sorted with CD44(+)/ESA(+)/CD24(-) markers, Gb3 activates c-Src/ß-catenin signaling and up-regulates the expression of FGF-2, CD44, and Oct-4 enriching tumorigenesis. Conversely, silencing glucosylceramide synthase expression disrupts Gb3 synthesis and selectively kills BCSCs through deactivation of c-Src/ß-catenin signaling. These findings highlight the unexploited role of ceramide glycosylation in selectively maintaining the tumorous pluripotency of cancer stem cells. It speculates that disruption of ceramide glycosylation or globo-series GSL is a useful approach to specifically target BCSCs specifically.


Subject(s)
Breast Neoplasms/enzymology , Ceramides/metabolism , Glucosyltransferases/metabolism , Neoplastic Stem Cells/enzymology , Animals , Antibiotics, Antineoplastic/pharmacology , Breast Neoplasms/pathology , CD24 Antigen/metabolism , Cell Separation , Cell Survival/drug effects , Cell Transformation, Neoplastic , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Female , Glycosylation , Humans , Hyaluronan Receptors/metabolism , Immunomagnetic Separation , MCF-7 Cells , Mice , Mice, Nude , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Protein Processing, Post-Translational , Signal Transduction , Spheroids, Cellular/drug effects , beta Catenin/metabolism
6.
Int J Biochem Cell Biol ; 44(11): 1770-8, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22728310

ABSTRACT

Myelosuppression and drug resistance are common adverse effects in cancer patients with chemotherapy, and those severely limit the therapeutic efficacy and lead treatment failure. It is unclear by which cellular mechanism anticancer drugs suppress bone marrow, while drug-resistant tumors survive. We report that due to the difference of glucosylceramide synthase (GCS), catalyzing ceramide glycosylation, doxorubicin (Dox) eliminates bone marrow stem cells (BMSCs) and expands breast cancer stem cells (BCSCs). It was found that Dox decreased the numbers of BMSCs (ABCG2(+)) and the sphere formation in a dose-dependent fashion in isolated bone marrow cells. In tumor-bearing mice, Dox treatments (5mg/kg, 6 days) decreased the numbers of BMSCs and white blood cells; conversely, those treatments increased the numbers of BCSCs (CD24(-)/CD44(+)/ESA(+)) more than threefold in the same mice. Furthermore, therapeutic-dose of Dox (1mg/kg/week, 42 days) decreased the numbers of BMSCs while it increased BCSCs in vivo. Breast cancer cells, rather than bone marrow cells, highly expressed GCS, which was induced by Dox and correlated with BCSC pluripotency. These results indicate that Dox may have opposite effects, suppressing BMSCs versus expanding BCSCs, and GCS is one determinant of the differentiated responsiveness of bone marrow and cancer cells.


Subject(s)
Bone Marrow Cells/enzymology , Bone Marrow Cells/pathology , Breast Neoplasms/pathology , Doxorubicin/toxicity , Glucosyltransferases/metabolism , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Animals , Blood Cell Count , Bone Marrow Cells/drug effects , Breast Neoplasms/blood , Breast Neoplasms/enzymology , Dose-Response Relationship, Drug , Female , Humans , Mice , Neoplastic Stem Cells/drug effects , Paclitaxel/toxicity , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...