Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
2.
Mol Cancer ; 14: 200, 2015 Nov 23.
Article in English | MEDLINE | ID: mdl-26597727

ABSTRACT

BACKGROUND: Pancreatic Ductal Adenocarcinoma (PDAC) is a devastating disease hallmarked by limited patient survival. Resistance to chemotherapy, a major cause of treatment failure in PDAC patients, is often attributed to Cancer Stem Cells (CSCs). Pancreatic CSCs are a small subset of quiescent cells within a tumor represented by surface markers like CD133. These cells are responsible not only for tumor recurrence, but also poor prognosis based on their "stem-like" characteristics. At present, conventional therapy is directed towards rapidly dividing PDAC cells and thus fails to target the CSC population. METHODS: MIA PaCa-2, S2-013 and AsPC-1 were treated with 12.5 nM triptolide (12 T cells) for 7 days. The surviving cells were recovered briefly in drug-free growth media and then transferred to Cancer Stem cell Media (CSM). As a control, untreated cells were also transferred to CSM media (CSM). The 12 T and CSM cells were tested for stemness properties using RNA and protein markers. Low numbers of CSM and 12 T cells were implanted subcutaneously in athymic nude mice to study their tumorigenic potential. 12 T and CSM cells were sorted for CD133 expression and assayed for their colony forming ability and sphere forming ability. Invasiveness of 12 T cells, CSM and MIA PaCa-2 were compared using Boyden chamber assays. RESULTS: Treated 12 T cells displayed increased expression of the surface marker CD133 and the drug transporter ABCG2 compared to untreated cells (CSM cells). Both 12 T and CSM cells formed subcutaneous tumors in mice confirming their tumor-initiating properties. When tested for invasion, 12 T cells had increased invasiveness compared to CSM cells. CD133(+) cells in both CSM and 12 T showed greater colony and sphere forming ability compared to CD133(-) cells from each group. Consistent with these data, when injected subcutaneously in mice, CD133(-) cells from CSM or 12 T did not form any tumors whereas CD133(+) cells from both groups showed tumor formation at a very low cell number. Despite pre-exposure to triptolide in 12 T CD133(+) cells, treatment of tumors formed by these cells with Minnelide, a triptolide pro-drug, showed significant tumor regression. CONCLUSION: Our results indicated that triptolide enhanced and enriched the "stemness" in the PDAC cell lines at a low dose of 12.5 nM, but also resulted in the regression of tumors derived from these cells.


Subject(s)
Antigens, CD/metabolism , Antineoplastic Agents/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Glycoproteins/metabolism , Organophosphates/pharmacology , Pancreatic Neoplasms/drug therapy , Peptides/metabolism , Phenanthrenes/pharmacology , AC133 Antigen , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Survival/drug effects , Diterpenes/pharmacology , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Epoxy Compounds/pharmacology , Humans , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/physiology , Pancreatic Neoplasms/pathology , Side-Population Cells/drug effects
3.
J Biol Chem ; 288(47): 33927-33938, 2013 Nov 22.
Article in English | MEDLINE | ID: mdl-24129563

ABSTRACT

Pancreatic cancer, the fourth most prevalent cancer-related cause of death in the United States, is a disease with a dismal survival rate of 5% 5 years after diagnosis. One of the survival proteins responsible for its extraordinary ability to evade cell death is HSP70. A naturally derived compound, triptolide, and its water-soluble prodrug, Minnelide, down-regulate the expression of this protein in pancreatic cancer cells, thereby causing cell death. However, the mechanism of action of triptolide has not been elucidated. Our study shows that triptolide-induced down-regulation of HSP70 expression is associated with a decrease in glycosylation of the transcription factor Sp1. We further show that triptolide inhibits glycosylation of Sp1, inhibiting the hexosamine biosynthesis pathway, particularly the enzyme O-GlcNAc transferase. Inhibition of O-GlcNAc transferase prevents nuclear localization of Sp1 and affects its DNA binding activity. This in turn down-regulates prosurvival pathways like NF-κB, leading to inhibition of HSF1 and HSP70 and eventually to cell death. In this study, we evaluated the mechanism by which triptolide affects glycosylation of Sp1, which in turn affects downstream pathways controlling survival of pancreatic cancer cells.


Subject(s)
Acetylglucosamine/metabolism , Antineoplastic Agents, Alkylating/pharmacology , Cell Nucleus/metabolism , Diterpenes/pharmacology , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/drug therapy , Phenanthrenes/pharmacology , Sp1 Transcription Factor/metabolism , Acetylglucosamine/genetics , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/genetics , Cell Death/drug effects , Cell Death/genetics , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/pathology , Cell Survival/drug effects , Cell Survival/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Down-Regulation/drug effects , Down-Regulation/genetics , Epoxy Compounds/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Glycosylation/drug effects , HSP70 Heat-Shock Proteins/biosynthesis , HSP70 Heat-Shock Proteins/genetics , Heat Shock Transcription Factors , Humans , N-Acetylglucosaminyltransferases/antagonists & inhibitors , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasm Proteins/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Signal Transduction/drug effects , Signal Transduction/genetics , Sp1 Transcription Factor/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Mol Cancer Res ; 20(9): 1443-1455, 2022 09 02.
Article in English | MEDLINE | ID: mdl-35639459

ABSTRACT

Basal-like breast cancers (BLBC) are the most common triple-negative subtype (hormone receptor and HER2 negative) with poor short-term disease outcome and are commonly identified by expression of basal cytokeratins (CK) 5 and 17. The goal of this study was to investigate whether CK5 and CK17 play a role in adverse behavior of BLBC cells. BLBC cell lines contain heterogeneous populations of cells expressing CK5, CK17, and the mesenchymal filament protein vimentin. Stable shRNA knockdown of either CK5 or CK17 compared with non-targeting control in BLBC cells was sufficient to promote an epithelial-mesenchymal transition (EMT) gene signature with loss of E-cadherin and an increase in vimentin expression. Relative to control cells, CK5 and CK17 knockdown cells acquired a more spindle-like morphology with increased cell scattering and were more invasive in vitro. However, CK5 or CK17 knockdown compared with control cells generated decreased lymph node and lung metastases in vivo. Loss of CK5 or CK17 moderately reduced the IC50 dose of doxorubicin in vitro and led to increased doxorubicin efficacy in vivo. Single-cell RNA-sequencing of BLBC patient-derived xenografts identified heterogeneous populations of CK5/CK17, vimentin, and dual basal CK/vimentin-positive cells that fell on an EMT spectrum of epithelial, mesenchymal, and intermediate, respectively, whereas knockdown of CK5 transitioned cells toward a more mesenchymal score. IMPLICATIONS: This study supports that basal CKs 5 and 17 contribute to the adverse behavior of BLBC cells and could be an untapped source of therapeutic vulnerability for this aggressive disease.


Subject(s)
Breast Neoplasms , Keratin-17/metabolism , Keratin-5/metabolism , Biomarkers, Tumor/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Doxorubicin , Female , Humans , Vimentin/genetics , Vimentin/metabolism
5.
Nat Commun ; 13(1): 2011, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35440136

ABSTRACT

Estrogen receptor alpha (ER/ESR1) is frequently mutated in endocrine resistant ER-positive (ER+) breast cancer and linked to ligand-independent growth and metastasis. Despite the distinct clinical features of ESR1 mutations, their role in intrinsic subtype switching remains largely unknown. Here we find that ESR1 mutant cells and clinical samples show a significant enrichment of basal subtype markers, and six basal cytokeratins (BCKs) are the most enriched genes. Induction of BCKs is independent of ER binding and instead associated with chromatin reprogramming centered around a progesterone receptor-orchestrated insulated neighborhood. BCK-high ER+ primary breast tumors exhibit a number of enriched immune pathways, shared with ESR1 mutant tumors. S100A8 and S100A9 are among the most induced immune mediators and involve in tumor-stroma paracrine crosstalk inferred by single-cell RNA-seq from metastatic tumors. Collectively, these observations demonstrate that ESR1 mutant tumors gain basal features associated with increased immune activation, encouraging additional studies of immune therapeutic vulnerabilities.


Subject(s)
Breast Neoplasms , Estrogen Receptor alpha/genetics , Breast Neoplasms/pathology , Estrogen Receptor alpha/metabolism , Female , Humans , Mutation
6.
Oncogene ; 39(12): 2478-2492, 2020 03.
Article in English | MEDLINE | ID: mdl-31988452

ABSTRACT

Estrogen receptor (ER) positive breast cancers often contain subpopulations of cells that express the intermediate filament protein cytokeratin 5 (CK5). CK5+ cells are enriched in cancer stem cell (CSC) properties, can be induced by progestins, and predict poor prognosis in ER+ breast cancer. We established through CK5 knockout and overexpression in ER+ breast cancer cell lines that CK5 is important for tumorsphere formation, prompting us to speculate that CK5 has regulatory activity in CSCs. To interrogate CK5 interacting proteins that may be functionally cooperative, we performed immunoprecipitation-mass spectrometry for CK5 in ER+ breast cancer cells. Focusing on proteins with signaling activity, we identified ß-catenin, a key transcription factor of the Wnt signaling pathway and cell adhesion molecule, as a CK5 interactor, which we confirmed by co-immunoprecipitation in several breast cancer models. We interrogated the dual functions of ß-catenin in relation to CK5. Knockout or knockdown of CK5 ablated ß-catenin transcriptional activity in response to progestins and Wnt stimuli. Conversely, CK5 induced by progestins or overexpression was sufficient to promote the loss of ß-catenin at the cell membrane and total E-cadherin loss. A breast cancer patient-derived xenograft showed similar loss of membrane ß-catenin and E-cadherin in CK5+ but not intratumoral CK5- cells and single-cell RNA sequencing found the top enriched pathways in the CK5+ cell cluster were cell junction remodeling and signaling. This report highlights that CK5 actively remodels cell morphology and that blockade of CK5-ß-catenin interaction may reverse the detrimental properties of CK5+ breast cancer cells.


Subject(s)
Breast Neoplasms/metabolism , Keratin-5/metabolism , Neoplastic Stem Cells/metabolism , beta Catenin/metabolism , Adherens Junctions/metabolism , Animals , Antigens, CD/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cadherins/metabolism , Carcinogenesis/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Female , Gene Knockout Techniques , Humans , Immunoprecipitation , Keratin-5/genetics , Mass Spectrometry , Mice , Progestins/pharmacology , Protein Interaction Mapping , Receptors, Estrogen/metabolism , Transcription, Genetic , Wnt Signaling Pathway
7.
Sci Rep ; 7(1): 7872, 2017 08 11.
Article in English | MEDLINE | ID: mdl-28801636

ABSTRACT

Pancreatic tumors are renowned for their extremely hypoxic centers, resulting in upregulation of a number of hypoxia mediated signaling pathways including cell proliferation, metabolism and cell survival. Previous studies from our laboratory have shown that Minnelide, a water-soluble pro-drug of triptolide (anti-cancer compound), decreases viability of cancer cells in vitro as well as in vivo. However, its mechanism of action remain elusive. In the current study we evaluated the effect of Minnelide, on hypoxia mediated oncogenic signaling as well as stemness in pancreatic cancer. Minnelide has just completed Phase 1 trial against GI cancers and is currently awaiting Phase 2 trials. Our results showed that upon treatment with triptolide, HIF-1α protein accumulated in pancreatic cancer cells even though hypoxic response was decreased in them. Our studies showed even though HIF-1α is accumulated in the treated cells, there was no decrease in HIF-1 binding to hypoxia response elements. However, the HIF-1 transcriptional activity was significantly reduced owing to depletion of co-activator p300 upon treatment with triptolide. Further, treatment with triptolide resulted in a decreased activity of Sp1 and NF-kB the two major oncogenic signaling pathway in pancreatic cancer along with a decreased tumor initiating cell (TIC) population in pancreatic tumor.


Subject(s)
Carcinogenesis/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplastic Stem Cells/drug effects , Organophosphates/pharmacology , Pancreatic Neoplasms/drug therapy , Phenanthrenes/pharmacology , Xenograft Model Antitumor Assays , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Diterpenes , Epoxy Compounds , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice, SCID , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
8.
Sci Rep ; 7(1): 1564, 2017 05 08.
Article in English | MEDLINE | ID: mdl-28484232

ABSTRACT

Endoplasmic reticulum (ER) stress initiates an important mechanism for cell adaptation and survival, named the unfolded protein response (UPR). Severe or chronic/prolonged UPR can breach the threshold for survival and lead to cell death. There is a fundamental gap in knowledge on the molecular mechanism of how chronic ER stress is stimulated and leads to cell death in pancreatic ductal adenocarcinoma (PDAC). Our study shows that downregulating specificity protein 1 (Sp1), a transcription factor that is overexpressed in pancreatic cancer, activates UPR and results in chronic ER stress. In addition, downregulation of Sp1 results in its decreased binding to the ER stress response element present in the promoter region of Grp78, the master regulator of ER stress, thereby preventing homeostasis. We further show that inhibition of Sp1, as well as induction of ER stress, leads to lysosomal membrane permeabilization (LMP), a sustained accumulation of cytosolic calcium, and eventually cell death in pancreatic cancer.


Subject(s)
Endoplasmic Reticulum/metabolism , Homeostasis , Intracellular Membranes/metabolism , Lysosomes/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Sp1 Transcription Factor/metabolism , Calcium/metabolism , Cell Death , Cell Line, Tumor , Cytosol/metabolism , Down-Regulation , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Humans , Models, Biological , Permeability
9.
Clin Cancer Res ; 22(2): 415-25, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26405195

ABSTRACT

PURPOSE: Pancreatic cancer stromal microenvironment is considered to be the major reason for failure of conventional and targeted therapy for this disease. The desmoplastic stroma, comprising mainly collagen and glycosaminoglycans like hyaluronan (HA), is responsible for compression of vasculature in the tumor resulting in impaired drug delivery and poor prognosis. Minnelide, a water-soluble prodrug of triptolide currently in phase I clinical trial, has been very effective in multiple animal models of pancreatic cancer. However, whether Minnelide will have efficacious delivery into the tumor despite the desmoplastic stroma has not been evaluated before. EXPERIMENT DESIGN: Patient tumor-derived xenografts (PDX) and spontaneous pancreatic cancer mice were treated with 0.42 and 0.21 mg/kg body weight for 30 days. Stromal components were determined by IHC and ELISA-based assays. Vascular functionality and drug delivery to the tumor were assessed following treatment with Minnelide. RESULT: Our current study shows that treatment with Minnelide resulted in reduction of ECM components like HA and collagen in the pancreatic cancer stroma of both the spontaneous KPC mice as well as in patient tumor xenografts. Furthermore, treatment with Minnelide improved functional vasculature in the tumors resulting in four times more functional vessels in the treated animals compared with untreated animals. Consistent with this observation, Minnelide also resulted in increased drug delivery into the tumor compared with untreated animals. Along with this, Minnelide also decreased viability of the stromal cells along with the tumor cells in pancreatic adenocarcinoma. CONCLUSIONS: In conclusion, these results are extremely promising as they indicate that Minnelide, along with having anticancer effects is also able to deplete stroma in pancreatic tumors, which makes it an effective therapy for pancreatic cancer.


Subject(s)
Organophosphates/pharmacology , Pancreatic Neoplasms/drug therapy , Phenanthrenes/pharmacology , Stromal Cells/drug effects , Adenocarcinoma/drug therapy , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Diterpenes/pharmacology , Drug Delivery Systems/methods , Epoxy Compounds/pharmacology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Tumor Microenvironment/drug effects , Pancreatic Neoplasms
10.
Oncotarget ; 7(35): 56324-56337, 2016 Aug 30.
Article in English | MEDLINE | ID: mdl-27472388

ABSTRACT

Chemoresistance in pancreatic cancer has been attributed to tumor-initiating cells (TICs), a minor sub-population of tumor cells. However, the mechanism of chemo-resistance in these cells is still unclear.In the current study, immunohistochemical analysis of LSL-KrasG12D; LSL-Trp53R172H;PdxCre (KPC) murine tumors indicated that hypoxic regions developed through tumor progression. This hypoxic "niche" correlated with increased CD133+ population that had an increased HIF1A activity. Consistent with this observation, CD133+ cells had increased glucose uptake and activity of glycolytic pathway enzymes compared to CD133- cells. Mass spectrometric analysis (UPLC-TQD) following metabolic labeling of CD133+ cells with [13C]-U6 glucose confirmed this observation. Furthermore, although both populations had functionally active mitochondria, CD133+ cells had low mitochondrial complex I and complex IV activity and lesser accumulation of ROS in response to standard chemotherapeutic compounds like paclitaxel, 5FU and gemcitabine. CD133+ cells also showed increased resistance to all three chemotherapeutic compounds and treatment with Glut1 inhibitor (STF31) reversed this resistance, promoting apoptotic death in these cells similar to CD133- cells.Our study indicates that the altered metabolic profile of CD133+ pancreatic TIC protects them against apoptosis, by reducing accumulation of ROS induced by standard chemotherapeutic agents, thereby confering chemoresistance. Since resistance to existing chemotherapy contributes to the poor prognosis in pancreatic cancer, our study paves the way for identifying novel therapeutic targets for managing chemoresistance and tumor recurrence in pancreatic cancer.


Subject(s)
Metabolic Networks and Pathways , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/pathology , Tumor Microenvironment , AC133 Antigen/genetics , AC133 Antigen/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Cell Hypoxia , Cell Line, Tumor , Cell Separation/methods , Chromatography, High Pressure Liquid , Drug Resistance, Neoplasm , Electron Transport Complex I/metabolism , Electron Transport Complex IV/metabolism , Glucose/metabolism , Glucose Transporter Type 1/antagonists & inhibitors , Glucose Transporter Type 1/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Metabolome , Mice , Mice, Transgenic , Mitochondria/drug effects , Mitochondria/metabolism , Neoplasms, Experimental/pathology , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL