Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters











Publication year range
1.
Oncogene ; 42(37): 2725-2736, 2023 09.
Article in English | MEDLINE | ID: mdl-37550562

ABSTRACT

PG545 (Pixatimod) is a highly sulfated small molecule known for its ability to inhibit heparanase and disrupt signaling mediated by heparan-binding-growth factors (HB-GF). Previous studies indicated that PG545 inhibits growth factor-mediated signaling in ovarian cancer (OC) to enhance response to chemotherapy. Here we investigated the previously unidentified mechanisms by which PG545 induces DNA damage in OC cells and found that PG545 induces DNA single- and double-strand breaks, reduces RAD51 expression in an autophagy-dependent manner and inhibits homologous recombination repair (HRR). These changes accompanied the ability of PG545 to inhibit endocytosis of the heparan-sulfate proteoglycan interacting DNA repair protein, DEK, leading to DEK sequestration in the tumor microenvironment (TME) and loss of nuclear DEK needed for HRR. As a result, PG545 synergized with poly (ADP-ribose) polymerase inhibitors (PARPis) in OC cell lines in vitro and in 55% of primary cultures of patient-derived ascites samples ex vivo. Moreover, PG545/PARPi synergy was observed in OC cells exhibiting either de novo or acquired resistance to PARPi monotherapy. PG545 in combination with rucaparib also generated increased DNA damage, increased antitumor effects and increased survival of mice bearing HRR proficient OVCAR5 xenografts compared to monotherapy treatment in vivo. Synergistic antitumor activity of the PG545/rucaparib combination was likewise observed in an immunocompetent syngeneic ID8F3 OC model. Collectively, these results suggest that targeting DEK-HSPG interactions in the TME through the use of PG545 may be a novel method of inhibiting DNA repair and sensitizing cells to PARPis.


Subject(s)
Ovarian Neoplasms , Poly(ADP-ribose) Polymerase Inhibitors , Saponins , Animals , Female , Humans , Mice , Angiogenesis Inhibitors/pharmacology , Cell Line, Tumor , DNA Repair , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Tumor Microenvironment , Saponins/pharmacology , Saponins/therapeutic use
2.
Gynecol Oncol ; 174: 11-20, 2023 07.
Article in English | MEDLINE | ID: mdl-37141817

ABSTRACT

OBJECTIVE: Alterations in DNA methylation are early events in endometrial cancer (EC) development and may have utility in EC detection via tampon-collected vaginal fluid. METHODS: For discovery, DNA from frozen EC, benign endometrium (BE), and benign cervicovaginal (BCV) tissues underwent reduced representation bisulfite sequencing (RRBS) to identify differentially methylated regions (DMRs). Candidate DMRs were selected based on receiver operating characteristic (ROC) discrimination, methylation level fold-change between cancers and controls, and absence of background CpG methylation. Methylated DNA marker (MDM) validation was performed using qMSP on DNA from independent EC and BE FFPE tissue sets. Women ≥45 years of age with abnormal uterine bleeding (AUB) or postmenopausal bleeding (PMB) or any age with biopsy-proven EC self-collected vaginal fluid using a tampon prior to clinically indicated endometrial sampling or hysterectomy. Vaginal fluid DNA was assayed by qMSP for EC-associated MDMs. Random forest modeling analysis was performed to generate predictive probability of underlying disease; results were 500-fold in-silico cross-validated. RESULTS: Thirty-three candidate MDMs met performance criteria in tissue. For the tampon pilot, 100 EC cases were frequency matched by menopausal status and tampon collection date to 92 BE controls. A 28-MDM panel highly discriminated between EC and BE (96% (95%CI 89-99%) specificity; 76% (66-84%) sensitivity (AUC 0.88). In PBS/EDTA tampon buffer, the panel yielded 96% (95% CI 87-99%) specificity and 82% (70-91%) sensitivity (AUC 0.91). CONCLUSION: Next generation methylome sequencing, stringent filtering criteria, and independent validation yielded excellent candidate MDMs for EC. EC-associated MDMs performed with promisingly high sensitivity and specificity in tampon-collected vaginal fluid; PBS-based tampon buffer with added EDTA improved sensitivity. Larger tampon-based EC MDM testing studies are warranted.


Subject(s)
Endometrial Neoplasms , Humans , Female , Genetic Markers , Edetic Acid/metabolism , Endometrial Neoplasms/diagnosis , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Endometrium/metabolism , DNA , DNA Methylation
4.
Br J Cancer ; 127(7): 1352-1364, 2022 10.
Article in English | MEDLINE | ID: mdl-35794237

ABSTRACT

BACKGROUND: Malignant pleural mesothelioma (MPM) is an aggressive neoplasm and often acquires chemoresistance by increasing stemness in tumour tissue, thereby generating cancer stem cells (CSCs). CSCs escape treatment by deploying metabolic pathways to trigger dormancy or proliferation, also gaining the ability to exit and re-enter the cell cycle to hide their cellular identity. METHODS: We employed various cellular and biochemical assays to identify the role of the glycolytic enzyme PFKFB3, by knocking it down and pharmacologically inhibiting it with PFK158, to determine its anticancer effects in vitro and in vivo by targeting the CSC population in MPM. RESULTS: Here, we have identified PFKFB3 as a strategic player to target the CSC population in MPM and demonstrated that both pharmacologic (PFK158) and genetic inhibition of PFKFB3 destroy the FAK-Stat3-SOX2 nexus resulting in a decline in conspicuous stem cell markers viz. ALDH, CD133, CD44, SOX2. Inhibition of PFKFB3 accumulates p21 and p27 in the nucleus by decreasing SKP2. Lastly, PFK158 diminishes tumour-initiating cells (TICs) mediated MPM xenograft in vivo. CONCLUSIONS: This study confers a comprehensive and mechanistic function of PFKFB3 in CSC maintenance that may foster exceptional opportunities for targeted small molecule blockade of the TICs in MPM.


Subject(s)
Mesothelioma, Malignant , Quinolines , Cell Line, Tumor , Cell Proliferation , Humans , Neoplastic Stem Cells/pathology , Phosphofructokinase-2/genetics , Phosphofructokinase-2/metabolism , Pyridines/pharmacology , Quinolines/pharmacology , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , SOXB1 Transcription Factors/pharmacology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
5.
Oncogene ; 41(33): 4003-4017, 2022 08.
Article in English | MEDLINE | ID: mdl-35804016

ABSTRACT

PFKFB3 (6-phosphofructo-2-kinase) is the rate-limiting enzyme of glycolysis and is overexpressed in several human cancers that are associated with poor prognosis. High PFKFB3 expression in cancer stem cells promotes glycolysis and survival in the tumor microenvironment. Inhibition of PFKFB3 by the glycolytic inhibitor PFK158 and by shRNA stable knockdown in small cell lung carcinoma (SCLC) cell lines inhibited glycolysis, proliferation, spheroid formation, and the expression of cancer stem cell markers CD133, Aldh1, CD44, Sox2, and ABCG2. These factors are also associated with chemotherapy resistance. We found that PFK158 treatment and PFKFB3 knockdown enhanced the ABCG2-interacting drugs doxorubicin, etoposide, and 5-fluorouracil in reducing cell viability under conditions of enriched cancer stem cells (CSC). Additionally, PFKFB3 inhibition attenuated the invasion/migration of SCLC cells by downregulating YAP/TAZ signaling while increasing pLATS1 via activation of pMST1 and NF2 and by reducing the mesenchymal protein expression. PFKFB3 knockdown and PFK158 treatment in a H1048 SCLC cancer stem cell-enriched mouse xenograft model showed significant reduction in tumor growth and weight with reduced expression of cancer stem cell markers, ABCG2, and YAP/TAZ. Our findings identify that PFKFB3 is a novel target to regulate cancer stem cells and its associated therapeutic resistance markers YAP/TAZ and ABCG2 in SCLC models.


Subject(s)
Lung Neoplasms , Small Cell Lung Carcinoma , Animals , Cell Line, Tumor , Cell Proliferation , Glycolysis , Hippo Signaling Pathway , Humans , Lung Neoplasms/pathology , Mice , Phosphofructokinase-2/metabolism , Pyridines , Quinolines , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/genetics , Tumor Microenvironment
6.
Gynecol Oncol ; 165(3): 568-576, 2022 06.
Article in English | MEDLINE | ID: mdl-35370009

ABSTRACT

OBJECTIVE: Aberrant DNA methylation is an early event in carcinogenesis which could be leveraged to detect ovarian cancer (OC) in plasma. METHODS: DNA from frozen OC tissues, benign fallopian tube epithelium (FTE), and buffy coats from cancer-free women underwent reduced representation bisulfite sequencing (RRBS) to identify OC MDMs. Candidate MDM selection was based on receiver operating characteristic (ROC) discrimination, methylation fold change, and low background methylation among controls. Blinded biological validation was performed using methylated specific PCR on DNA extracted from independent OC and FTE FFPE tissues. MDMs were tested using Target Enrichment Long-probe Quantitative Amplified Signal (TELQAS) assays in pre-treatment plasma from women newly diagnosed with OC and population-sampled healthy women. A random forest modeling analysis was performed to generate predictive probability of disease; results were 500-fold in silico cross-validated. RESULTS: Thirty-three MDMs showed marked methylation fold changes (10 to >1000) across all OC subtypes vs FTE. Eleven MDMs (GPRIN1, CDO1, SRC, SIM2, AGRN, FAIM2, CELF2, RIPPLY3, GYPC, CAPN2, BCAT1) were tested on plasma from 91 women with OC (73 (80%) high-grade serous (HGS)) and 91 without OC; the cross-validated 11-MDM panel highly discriminated OC from controls (96% (95% CI, 89-99%) specificity; 79% (69-87%) sensitivity, and AUC 0.91 (0.86-0.96)). Among the 5 stage I/II HGS OCs included, all were correctly identified. CONCLUSIONS: Whole methylome sequencing, stringent filtering criteria, and biological validation yielded candidate MDMs for OC that performed with high sensitivity and specificity in plasma. Larger plasma-based OC MDM studies, including testing of pre-diagnostic specimens, are warranted.


Subject(s)
DNA Methylation , Ovarian Neoplasms , Biomarkers, Tumor/genetics , CELF Proteins/genetics , Carcinoma, Ovarian Epithelial/diagnosis , Carcinoma, Ovarian Epithelial/genetics , Feasibility Studies , Female , Genetic Markers , Humans , Nerve Tissue Proteins/genetics , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/genetics , Transaminases/genetics
7.
Cancer Res ; 82(6): 1038-1054, 2022 03 15.
Article in English | MEDLINE | ID: mdl-34654724

ABSTRACT

Dissemination of ovarian cancer cells can lead to inoperable metastatic lesions in the bowel and omentum that cause patient death. Here we show that LRRC15, a type-I 15-leucine-rich repeat-containing membrane protein, highly overexpressed in ovarian cancer bowel metastases compared with matched primary tumors and acts as a potent promoter of omental metastasis. Complementary models of ovarian cancer demonstrated that LRRC15 expression leads to inhibition of anoikis-induced cell death and promotes adhesion and invasion through matrices that mimic omentum. Mechanistically, LRRC15 interacted with ß1-integrin to stimulate activation of focal adhesion kinase (FAK) signaling. As a therapeutic proof of concept, targeting LRRC15 with the specific antibody-drug conjugate ABBV-085 in both early and late metastatic ovarian cancer cell line xenograft models prevented metastatic dissemination, and these results were corroborated in metastatic patient-derived ovarian cancer xenograft models. Furthermore, treatment of 3D-spheroid cultures of LRRC15-positive patient-derived ascites with ABBV-085 reduced cell viability. Overall, these data uncover a role for LRRC15 in promoting ovarian cancer metastasis and suggest a novel and promising therapy to target ovarian cancer metastases.Significance: This study identifies that LRRC15 activates ß1-integrin/FAK signaling to promote ovarian cancer metastasis and shows that the LRRC15-targeted antibody-drug conjugate ABBV-085 suppresses ovarian cancer metastasis in preclinical models.


Subject(s)
Immunoconjugates , Ovarian Neoplasms , Carcinoma, Ovarian Epithelial , Cell Adhesion , Cell Line, Tumor , Female , Humans , Immunoconjugates/pharmacology , Integrins , Membrane Proteins/genetics , Membrane Proteins/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology
8.
J Exp Clin Cancer Res ; 40(1): 182, 2021 Jun 03.
Article in English | MEDLINE | ID: mdl-34082797

ABSTRACT

BACKGROUND: Aberrant lipogenicity and deregulated autophagy are common in most advanced human cancer and therapeutic strategies to exploit these pathways are currently under consideration. Group III Phospholipase A2 (sPLA2-III/PLA2G3), an atypical secretory PLA2, is recognized as a regulator of lipid metabolism associated with oncogenesis. Though recent studies reveal that high PLA2G3 expression significantly correlates with poor prognosis in several cancers, however, role of PLA2G3 in ovarian cancer (OC) pathogenesis is still undetermined. METHODS: CRISPR-Cas9 and shRNA mediated knockout and knockdown of PLA2G3 in OC cells were used to evaluate lipid droplet (LD) biogenesis by confocal and Transmission electron microscopy analysis, and the cell viability and sensitization of the cells to platinum-mediated cytotoxicity by MTT assay. Regulation of primary ciliation by PLA2G3 downregulation both genetically and by metabolic inhibitor PFK-158 induced autophagy was assessed by immunofluorescence-based confocal analysis and immunoblot. Transient transfection with GFP-RFP-LC3B and confocal analysis was used to assess the autophagic flux in OC cells. PLA2G3 knockout OVCAR5 xenograft in combination with carboplatin on tumor growth and metastasis was assessed in vivo. Efficacy of PFK158 alone and with platinum drugs was determined in patient-derived primary ascites cultures expressing PLA2G3 by MTT assay and immunoblot analysis. RESULTS: Downregulation of PLA2G3 in OVCAR8 and 5 cells inhibited LD biogenesis, decreased growth and sensitized cells to platinum drug mediated cytotoxicity in vitro and in in vivo OVCAR5 xenograft. PLA2G3 knockdown in HeyA8MDR-resistant cells showed sensitivity to carboplatin treatment. We found that both PFK158 inhibitor-mediated and genetic downregulation of PLA2G3 resulted in increased number of percent ciliated cells and inhibited cancer progression. Mechanistically, we found that PFK158-induced autophagy targeted PLA2G3 to restore primary cilia in OC cells. Of clinical relevance, PFK158 also induces percent ciliated cells in human-derived primary ascites cells and reduces cell viability with sensitization to chemotherapy. CONCLUSIONS: Taken together, our study for the first time emphasizes the role of PLA2G3 in regulating the OC metastasis. This study further suggests the therapeutic potential of targeting phospholipases and/or restoration of PC for future OC treatment and the critical role of PLA2G3 in regulating ciliary function by coordinating interface between lipogenesis and metastasis.


Subject(s)
Cell Proliferation/drug effects , Group III Phospholipases A2/genetics , Lipogenesis/drug effects , Ovarian Neoplasms/drug therapy , Animals , Autophagy/drug effects , CRISPR-Cas Systems/genetics , Cell Survival/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Heterografts , Humans , Lipid Droplets/drug effects , Mice , Microscopy, Electron, Transmission , Neoplasm Metastasis , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Platinum/pharmacology , Pyridines/pharmacology , Quinolines/pharmacology
9.
Cancers (Basel) ; 13(9)2021 Apr 21.
Article in English | MEDLINE | ID: mdl-33919392

ABSTRACT

We previously reported that the antimalarial compound quinacrine (QC) induces autophagy in ovarian cancer cells. In the current study, we uncovered that QC significantly upregulates cathepsin L (CTSL) but not cathepsin B and D levels, implicating the specific role of CTSL in promoting QC-induced autophagic flux and apoptotic cell death in OC cells. Using a Magic Red® cathepsin L activity assay and LysoTracker red, we discerned that QC-induced CTSL activation promotes lysosomal membrane permeability (LMP) resulting in the release of active CTSL into the cytosol to promote apoptotic cell death. We found that QC-induced LMP and CTSL activation promotes Bid cleavage, mitochondrial outer membrane permeabilization (MOMP), and mitochondrial cytochrome-c release. Genetic (shRNA) and pharmacological (Z-FY(tBU)-DMK) inhibition of CTSL markedly reduces QC-induced autophagy, LMP, MOMP, apoptosis, and cell death; whereas induced overexpression of CTSL in ovarian cancer cell lines has an opposite effect. Using recombinant CTSL, we identified p62/SQSTM1 as a novel substrate of CTSL, suggesting that CTSL promotes QC-induced autophagic flux. CTSL activation is specific to QC-induced autophagy since no CTSL activation is seen in ATG5 knockout cells or with the anti-malarial autophagy-inhibiting drug chloroquine. Importantly, we showed that upregulation of CTSL in QC-treated HeyA8MDR xenografts corresponds with attenuation of p62, upregulation of LC3BII, cytochrome-c, tBid, cleaved PARP, and caspase3. Taken together, the data suggest that QC-induced autophagy and CTSL upregulation promote a positive feedback loop leading to excessive autophagic flux, LMP, and MOMP to promote QC-induced cell death in ovarian cancer cells.

10.
Oncogene ; 40(8): 1409-1424, 2021 02.
Article in English | MEDLINE | ID: mdl-33420377

ABSTRACT

The advanced or recurrent endometrial cancer (EC) has a poor prognosis because of chemoresistance. 6-Phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3), a glycolytic enzyme, is overexpressed in a variety of human cancers and plays important roles in promoting tumor cell growth. Here, we showed that high expression of PFKFB3 in EC cell lines is associated with chemoresistance. Pharmacological inhibition of PFKFB3 with PFK158 and or genetic downregulation of PFKFB3 dramatically suppressed cell proliferation and enhanced the sensitivity of EC cells to carboplatin (CBPt) and cisplatin (Cis). Moreover, PFKFB3 inhibition resulted in reduced glucose uptake, ATP production, and lactate release. Notably, we found that PFK158 with CBPt or Cis exerted strong synergistic antitumor activity in chemoresistant EC cell lines, HEC-1B and ARK-2 cells. We also found that the combination of PFK158 and CBPt/Cis induced apoptosis- and autophagy-mediated cell death through inhibition of the Akt/mTOR signaling pathway. Mechanistically, we found that PFK158 downregulated the CBPt/Cis-induced upregulation of RAD51 expression and enhanced CBPt/Cis-induced DNA damage as demonstrated by an increase in γ-H2AX levels in HEC-1B and ARK-2 cells, potentially revealing a means to enhance PFK158-induced chemosensitivity. More importantly, PFK158 treatment, either as monotherapy or in combination with CBPt, led to a marked reduction in tumor growth in two chemoresistant EC mouse xenograft models. These data suggest that PFKFB3 inhibition alone or in combination with standard chemotherapy may be used as a novel therapeutic strategy for improved therapeutic efficacy and outcomes of advanced and recurrent EC patients.


Subject(s)
Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Endometrial Neoplasms/drug therapy , Phosphofructokinase-2/genetics , Apoptosis/drug effects , Carboplatin/pharmacology , Cell Line, Tumor , Cisplatin/pharmacology , Endometrial Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Phosphofructokinase-2/antagonists & inhibitors , Pyridines/pharmacology , Quinolines/pharmacology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/genetics , Xenograft Model Antitumor Assays
11.
Gynecol Oncol ; 156(2): 387-392, 2020 02.
Article in English | MEDLINE | ID: mdl-31787246

ABSTRACT

OBJECTIVE: We aimed to assess whether endometrial cancer (EC) can be detected in shed DNA collected with vaginal tampon by analyzing copy number, methylation markers, and mutations. METHODS: Tampons were collected prior to hysterectomy from 38 EC patients and 28 women with benign indications. Extracted tampon DNA underwent the following: 1) low-coverage whole genome sequencing (LC-WGS) to assess copy number, 2) pyrosequencing to measure percent promotor methylation of HOXA9, RASSF1, and CDH13 and 3) next generation sequencing (NGS) to identify mutations in 19 genes associated with EC identified through The Cancer Genome Atlas. Sensitivity and specificity for each test and test combinations were calculated. RESULTS: Methylation analysis yielded the highest specificities but lowest sensitivities (37-40% sensitivity; 100% specificity for HOXA9, RASSF1 and HTR1B) while mutation analysis had improved sensitivity (50% sensitivity; 83% specificity). Only one "false positive" result for copy number variants was identified among women with benign surgical indications, which was based on detection of copy number changes, and associated with a leiomyosarcoma that was only recognized at hysterectomy. Considering any of the 3 biomarker classes as a positive, resulted in a sensitivity of 92% and specificity of 86%. Mutation analysis did not add sensitivity to the combination of analysis of copy number and methylation. CONCLUSIONS: This study demonstrates a proof-of-principle for non-invasive yet precise detection of endometrial cancer. We propose that with improved biomarker testing, it may be possible to develop a clinically useful test for detecting EC.


Subject(s)
DNA Methylation , Endometrial Neoplasms/genetics , Gene Dosage , Menstrual Hygiene Products , Biomarkers, Tumor/genetics , Diagnosis, Differential , Endometrial Neoplasms/diagnosis , Endometrial Neoplasms/pathology , Female , Humans , Middle Aged , Mutation , Uterine Diseases/diagnosis , Uterine Diseases/genetics , Uterine Diseases/pathology , Vaginal Smears/methods
12.
Cell Death Dis ; 10(10): 725, 2019 09 27.
Article in English | MEDLINE | ID: mdl-31562297

ABSTRACT

The metabolic signatures of cancer cells are often associated with elevated glycolysis. Pharmacological (PFK158 treatment) and genetic inhibition of 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3), a critical control point in the glycolytic pathway, decreases glucose uptake, ATP production, and lactate dehydrogenase activity and arrests malignant pleural mesothelioma (MPM) cells in the G0/G1 phase to induce cell death. To overcome this nutrient stress, inhibition of PFKFB3 activity led to an escalation in endoplasmic reticulum (ER) activity and aggravated ER stress mostly by upregulating BiP and GADD153 expression and activation of the endocytic Rac1-Rab5-Rab7 pathway resulting in a unique form of cell death called "methuosis" in both the sarcomatoid (H28) and epithelioid (EMMeso) cells. Transmission electron microscopy (TEM) analysis showed the formation of nascent macropinocytotic vesicles, which rapidly coalesced to form large vacuoles with compromised lysosomal function. Both immunofluorescence microscopy and co-immunoprecipitation analyses revealed that upon PFKFB3 inhibition, two crucial biomolecules of each pathway, Rac1 and Calnexin interact with each other. Finally, PFK158 alone and in combination with carboplatin-inhibited tumorigenesis of EMMeso xenografts in vivo. Since most cancer cells exhibit an increased glycolytic rate, these results provide evidence for PFK158, in combination with standard chemotherapy, may have a potential in the treatment of MPM.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Lung Neoplasms/genetics , Mesothelioma/genetics , Phosphofructokinase-2/antagonists & inhibitors , Pyridines/pharmacology , Animals , Apoptosis/drug effects , Calcium/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cellular Reprogramming/drug effects , Endoplasmic Reticulum Stress/drug effects , Female , Glycolysis , Heterografts , Humans , Lung Neoplasms/metabolism , Mesothelioma/metabolism , Mesothelioma, Malignant , Mice , Mice, Nude , Phosphofructokinase-2/genetics , Phosphofructokinase-2/metabolism , Pinocytosis , Pleural Neoplasms/genetics , Pleural Neoplasms/metabolism
13.
Gynecol Oncol ; 154(3): 495-504, 2019 09.
Article in English | MEDLINE | ID: mdl-31204077

ABSTRACT

OBJECTIVE: This study is designed to identify genes and pathways that could promote metastasis to the bowel in high-grade serous ovarian cancer (OC) and evaluate their associations with clinical outcomes. METHODS: We performed RNA sequencing of OC primary tumors (PTs) and their corresponding bowel metastases (n = 21 discovery set; n = 18 replication set). Differentially expressed genes (DEGs) were those expressed at least 2-fold higher in bowel metastases (BMets) than PTs in at least 30% of patients (P < .05) with no increased expression in paired benign bowel tissue and were validated with quantitative reverse transcription PCR. Using an independent OC cohort (n = 333), associations between DEGs in PTs and surgical and clinical outcomes were performed. Immunohistochemistry and mouse xenograft studies were performed to confirm the role of LRRC15 in promoting metastasis. RESULTS: Among 27 DEGs in the discovery set, 21 were confirmed in the replication set: SFRP2, Col11A1, LRRC15, ADAM12, ADAMTS12, MFAP5, LUM, PLPP4, FAP, POSTN, GRP, MMP11, MMP13, C1QTNF3, EPYC, DIO2, KCNA1, NETO1, NTM, MYH13, and PVALB. Higher expression of more than half of the genes in the PT was associated with an increased requirement for bowel resection at primary surgery and an inability to achieve complete cytoreduction. Increased expression of LRRC15 in BMets was confirmed by immunohistochemistry and knockdown of LRRC15 significantly inhibited tumor progression in mice. CONCLUSIONS: We identified 21 genes that are overexpressed in bowel metastases among patients with OC. Our findings will help select potential molecular targets for the prevention and treatment of malignant bowel obstruction in OC.


Subject(s)
Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/pathology , Intestinal Neoplasms/genetics , Intestinal Neoplasms/secondary , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Animals , Cell Line, Tumor , Cohort Studies , Female , Gene Knockdown Techniques , Heterografts , High-Throughput Nucleotide Sequencing , Humans , Membrane Proteins/genetics , Mice , Mice, Nude , RNA, Neoplasm/genetics , Transcriptome , Up-Regulation
14.
Int J Cancer ; 144(1): 178-189, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30226266

ABSTRACT

Metabolic alterations are increasingly recognized as important novel anti-cancer targets. Among several regulators of metabolic alterations, fructose 2,6 bisphosphate (F2,6BP) is a critical glycolytic regulator. Inhibition of the active form of PFKFB3ser461 using a novel inhibitor, PFK158 resulted in reduced glucose uptake, ATP production, lactate release as well as induction of apoptosis in gynecologic cancer cells. Moreover, we found that PFK158 synergizes with carboplatin (CBPt) and paclitaxel (PTX) in the chemoresistant cell lines, C13 and HeyA8MDR but not in their chemosensitive counterparts, OV2008 and HeyA8, respectively. We determined that PFK158-induced autophagic flux leads to lipophagy resulting in the downregulation of cPLA2, a lipid droplet (LD) associated protein. Immunofluorescence and co-immunoprecipitation revealed colocalization of p62/SQSTM1 with cPLA2 in HeyA8MDR cells uncovering a novel pathway for the breakdown of LDs promoted by PFK158. Interestingly, treating the cells with the autophagic inhibitor bafilomycin A reversed the PFK158-mediated synergy and lipophagy in chemoresistant cells. Finally, in a highly metastatic PTX-resistant in vivo ovarian mouse model, a combination of PFK158 with CBPt significantly reduced tumor weight and ascites and reduced LDs in tumor tissue as seen by immunofluorescence and transmission electron microscopy compared to untreated mice. Since the majority of cancer patients will eventually recur and develop chemoresistance, our results suggest that PFK158 in combination with standard chemotherapy may have a direct clinical role in the treatment of recurrent cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Autophagy/drug effects , Enzyme Inhibitors/pharmacology , Ovarian Neoplasms/drug therapy , Phosphofructokinase-2/antagonists & inhibitors , Pyridines/pharmacology , Quinolines/pharmacology , Xenograft Model Antitumor Assays , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Carboplatin/administration & dosage , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Enzyme Inhibitors/therapeutic use , Female , Glycolysis/drug effects , Humans , Lipid Droplets/drug effects , Lipid Droplets/metabolism , Mice, Nude , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Paclitaxel/administration & dosage , Phosphofructokinase-2/metabolism , Pyridines/therapeutic use , Quinolines/therapeutic use
15.
Clin Cancer Res ; 16(2): 398-409, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20068077

ABSTRACT

PURPOSE: Some 85% of lung cancers are smoking related. Here, we investigate the role of serine protease HtrA3 in smoking-related lung cancer. EXPERIMENTAL DESIGN: We assess HtrA3 methylation and its corresponding expression in the human bronchial cell line BEAS-2B following cigarette smoke carcinogen treatment, in lung cancer cell lines and in primary lung tumors from light, moderate, and heavy smokers. We also show the effects of HtrA3 downregulation on MTT reduction and clonogenic survival with etoposide and cisplatin treatment and the corresponding effects of HtrA3 re-expression during treatment. RESULTS: We show for the first time that HtrA3 expression is reduced or completely lost in over 50% of lung cancer cell lines and primary lung tumors from heavy smokers. Treatment of HtrA3-deficient cell lines with 5-aza-2'-deoxycytidine resulted in a dose-dependent increase in HtrA3 transcription. Further, sequence analysis of bisulfite-modified DNA from lung cancer cell lines and from primary lung tumors showed an increased frequency of methylation within the first exon of HtrA3 with a corresponding loss of HtrA3 expression, particularly in tumors from smokers. In BEAS-2B, treatment with the cigarette smoke carcinogen 4-(methylnitrosamino)-I-(3-pyridyl)-1-butanone resulted in HtrA3 downregulation with a corresponding increase in methylation. Additional studies indicate resistance to etoposide and cisplatin cytotoxicity as a functional consequence of HtrA3 loss. Finally, immunohistochemical analysis of primary lung tumors revealed a strong correlation between low HtrA3 expression and heavy smoking history. CONCLUSIONS: Collectively, these results suggest that cigarette smoke-induced methylation of HtrA3 could contribute to the etiology of chemoresistant disease in smoking-related lung cancer.


Subject(s)
Carcinoma, Bronchogenic/etiology , DNA Methylation/physiology , Gene Silencing , Lung Neoplasms/etiology , Serine Endopeptidases/genetics , Smoking/adverse effects , Adenocarcinoma/etiology , Adenocarcinoma/genetics , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Bronchogenic/drug therapy , Carcinoma, Bronchogenic/genetics , Carcinoma, Large Cell/etiology , Carcinoma, Large Cell/genetics , Carcinoma, Squamous Cell/etiology , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Cells, Cultured , Cisplatin/administration & dosage , Drug Resistance, Neoplasm/genetics , Etoposide/administration & dosage , Gene Silencing/physiology , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mice , Mice, Inbred BALB C , Nitrosamines/adverse effects , Serine Endopeptidases/metabolism
16.
J Biol Chem ; 282(19): 14413-20, 2007 May 11.
Article in English | MEDLINE | ID: mdl-17363371

ABSTRACT

Heparan sulfate (HS) glycosaminoglycans are the oligosaccharide chains of heparan sulfate proteoglycans. The sulfation of HS glycosaminoglycan residues is required for its interaction with various heparin-binding growth factors to promote their biological activities to activate their high affinity receptor tyrosine kinases. We have identified HS glycosaminoglycan-6-O-endosulfatase HSulf-1 as a down-regulated gene in ovarian, breast, and several other cancer cell lines. Here we have shown that HSulf-1 inhibits autocrine activation of the EGFR-ERK (epidermal growth factor receptor-extracellular signal-regulated kinase) pathway induced by serum withdrawal in MDA-MB-468 breast cancer cells. Short hairpin RNA-mediated down-regulation of HSulf-1 in HSulf-1 clonal lines of MDA-MB-468 led to a significant increase in autocrine activation of ERK compared with vector only control. The autocrine signaling was also inhibited with neutralization antibodies against amphiregulin and HB-EGF, the heparin-binding growth factor family of the EGF superfamily. Furthermore, HSulf-1-mediated inhibition of autocrine signaling was associated with reduced cyclin D1 levels, leading to decreased S phase fraction and increased G(2)-M fraction, as well as increased cell death. Finally, evaluation of HSulf-1 expression levels in primary invasive breast tumors by RNA in situ hybridization indicated that HSulf-1 is down-regulated in the majority (60%) of tumors, with a predominant association with lobular histology. These data suggest a potential role of HSulf-1 down-regulation in mammary carcinogenesis.


Subject(s)
Autocrine Communication , Breast Neoplasms/metabolism , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Sulfotransferases/metabolism , Amphiregulin , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Carcinoma, Lobular/metabolism , Carcinoma, Lobular/pathology , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cyclin D , Cyclins/metabolism , Down-Regulation , EGF Family of Proteins , Enzyme-Linked Immunosorbent Assay , Epidermal Growth Factor/antagonists & inhibitors , Epidermal Growth Factor/immunology , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , Female , Gene Expression Regulation, Neoplastic , Glycoproteins/antagonists & inhibitors , Glycoproteins/immunology , Heparin-binding EGF-like Growth Factor , Humans , In Situ Hybridization , Intercellular Signaling Peptides and Proteins/immunology , Middle Aged , Neoplasm Invasiveness/pathology , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , RNA, Small Interfering/pharmacology , Retroviridae , S Phase , Signal Transduction , Sulfotransferases/antagonists & inhibitors , Sulfotransferases/genetics , Tissue Array Analysis
17.
J Clin Invest ; 116(7): 1994-2004, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16767218

ABSTRACT

Resistance to chemotherapy presents a serious challenge in the successful treatment of various cancers and is mainly responsible for mortality associated with disseminated cancers. Here we show that expression of HtrA1, which is frequently downregulated in ovarian cancer, influences tumor response to chemotherapy by modulating chemotherapy-induced cytotoxicity. Downregulation of HtrA1 attenuated cisplatin- and paclitaxel-induced cytotoxicity, while forced expression of HtrA1 enhanced cisplatin- and paclitaxel-induced cytotoxicity. HtrA1 expression was upregulated by both cisplatin and paclitaxel treatment. This upregulation resulted in limited autoproteolysis and activation of HtrA1. Active HtrA1 induces cell death in a serine protease-dependent manner. The potential role of HtrA1 as a predictive factor of clinical response to chemotherapy was assessed in both ovarian and gastric cancer patients receiving cisplatin-based regimens. Patients with ovarian or gastric tumors expressing higher levels of HtrA1 showed a higher response rate compared with those with lower levels of HtrA1 expression. These findings uncover what we believe to be a novel pathway by which serine protease HtrA1 mediates paclitaxel- and cisplatin-induced cytotoxicity and suggest that loss of HtrA1 in ovarian and gastric cancers may contribute to in vivo chemoresistance.


Subject(s)
Drug Therapy , Neoplasms , Serine Endopeptidases/metabolism , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , Antineoplastic Agents, Phytogenic/therapeutic use , Caspases/metabolism , Cell Death , Cell Line, Tumor , Cisplatin/therapeutic use , Enzyme Activation , Female , High-Temperature Requirement A Serine Peptidase 1 , Humans , Male , Middle Aged , Molecular Sequence Data , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Paclitaxel/therapeutic use , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Serine Endopeptidases/genetics , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
18.
Cancer Res ; 66(12): 6025-32, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16778174

ABSTRACT

We previously identified HSulf-1 as a down-regulated gene in several tumor types including ovarian, breast, and hepatocellular carcinomas. Loss of HSulf-1, which selectively removes 6-O-sulfate from heparan sulfate, up-regulates heparin-binding growth factor signaling and confers resistance to chemotherapy-induced apoptosis. Here we report that HSulf-1 expression in MDA-MB-468 breast carcinoma clonal lines leads to reduced proliferation in vitro and reduced tumor burden in athymic nude mice in vivo. Additionally, xenografts derived from HSulf-1-expressing stable clones of carcinoma cells showed reduced vessel density, marked necrosis, and apoptosis, indicative of inhibition of angiogenesis. Consistent with this observation, HSulf-1-expressing clonal lines showed reduced staining with the endothelial marker CD31 in Matrigel plug assay, indicating that HSulf-1 expression inhibits angiogenesis. More importantly, HSulf-1 expression in the xenografts was associated with a reduced ability of vascular endothelial cell heparan sulfate to participate in a complex with fibroblast growth factor 2 (FGF-2) and its receptor tyrosine kinase FGF receptor 1c. In vitro, short hairpin RNA-mediated down-regulation of HSulf-1 in human umbilical vein endothelial cells (HUVEC) resulted in an increased proliferation mediated by heparan sulfate-dependent FGF-2, hepatocyte growth factor, and vascular endothelial growth factor 165 (VEGF165) but not by heparan sulfate-independent VEGF121. HSulf-1 down-regulation also enhanced downstream signaling through the extracellular signal-regulated kinase pathway compared with untreated cells. Consistent with the role of heparan sulfate glycosaminoglycan sulfation in VEGF-mediated signaling, treatment of HUVEC cells with chlorate, which inhibits heparan sulfate glycosaminoglycan sulfation and therefore mimics HSulf-1 overexpression, led to an attenuated VEGF-mediated signaling. Collectively, these observations provide the first evidence of a novel mechanism by which HSulf-1 modulates the function of heparan sulfate binding VEGF165 in proliferation and angiogenesis.


Subject(s)
Breast Neoplasms/blood supply , Breast Neoplasms/therapy , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/therapy , Sulfotransferases/physiology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Growth Processes/physiology , Cell Line, Tumor , Down-Regulation , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelial Cells/physiology , Female , Fibroblast Growth Factor 2/metabolism , Heparitin Sulfate/metabolism , Humans , Mice , Mice, Nude , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , RNA, Small Interfering/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Sulfotransferases/biosynthesis , Sulfotransferases/genetics , Transfection , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
19.
Mol Cell Neurosci ; 32(1-2): 82-90, 2006.
Article in English | MEDLINE | ID: mdl-16624571

ABSTRACT

We traced the cause of a slow-channel syndrome (SCS) in a patient with progressive muscle weakness, repetitive compound muscle action potential and prolonged low amplitude synaptic currents to a V --> F substitution in the M1 domain of the beta subunit (betaV229F) of the muscle acetylcholine receptor (AChR). In vitro expression studies in Xenopus oocytes indicated that the novel mutation betaV229F expressed normal amounts of AChRs and decreased the ACh EC50 by 10-fold compared to wild type. Kinetic analysis indicated that the mutation displayed prolonged mean open duration and repeated openings during activation. Prolonged openings caused by the betaV229F mutation were due to a reduction in the channel closing rate and an increase in the effective channel opening rate. Repeated openings of the channel during activation were caused by a significant reduction in the agonist dissociation constant. In addition, the betaV229F mutation produced an increase in calcium permeability. The kinetic and permeation studies presented in this work are sufficient to explain the consequences of the betaV229F mutation on the miniature endplate currents and thus are direct evidence that the betaV229F mutation is responsible for compromising the safety margin of neuromuscular transmission in the patient.


Subject(s)
Acetylcholine/metabolism , Mutation/genetics , Myasthenic Syndromes, Congenital/genetics , Neuromuscular Junction/genetics , Receptors, Nicotinic/genetics , Synaptic Transmission/genetics , Aged , Amino Acid Substitution , Animals , Calcium Signaling/genetics , Cell Membrane Permeability/genetics , Excitatory Postsynaptic Potentials/genetics , Female , Humans , Ion Channels/genetics , Ion Channels/metabolism , Kinetics , Male , Muscle, Skeletal/innervation , Muscle, Skeletal/physiopathology , Myasthenic Syndromes, Congenital/metabolism , Myasthenic Syndromes, Congenital/physiopathology , Neuromuscular Junction/metabolism , Neuromuscular Junction/physiopathology , Oocytes , Pedigree , Xenopus laevis
20.
Oncogene ; 24(32): 5089-100, 2005 Jul 28.
Article in English | MEDLINE | ID: mdl-15870691

ABSTRACT

Epigenetic silencing by hypermethylation of CpGs represents a mechanism of inactivation of tumor suppressors. Here we report on the cloning of a novel candidate tumor suppressor gene TCEAL7 inactivated by methylation in ovarian cancer. TCEAL codes for a 1.35 kb transcript that was previously reported to be downregulated in ovarian cancer by cDNA microarray and suppression subtraction cDNA (SSH) analyses. This report focuses on the elucidation of mechanisms associated with TCEAL7 downregulation. Expression of TCEAL7 is downregulated in a majority of ovarian tumors and cancer cell lines but induced by 5-aza-2'-deoxycytidine treatment in a dose-dependant manner, implicating methylation as a mechanism of TCEAL7 inactivation. Sequence analyses of bisufite-modified genomic DNA from somatic cell hybrids with either the active or the inactive human X chromosome reveal that TCEAL7 is subjected to X chromosome inactivation. Loss of TCEAL7 expression in primary tumors and cell lines correlates with methylation of a CpG site within the promoter. In vitro methylation of the CpG site suppresses promoter activity whereas selective demethylation of the SmaI site attenuates the suppression. Finally, re-expression of TCEAL7 in cancer cell lines induces cell death and reduces colony formation efficiency. These data implicate TCEAL7 as a cell death regulatory protein that is frequently inactivated in ovarian cancers, and suggest that it may function as a tumor suppressor.


Subject(s)
Gene Expression Regulation, Neoplastic , Gene Silencing , Genes, Tumor Suppressor , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Ovarian Neoplasms/genetics , Amino Acid Sequence , Apoptosis Regulatory Proteins , Base Sequence , Cell Line, Tumor , Chromosomes, Human, X , Conserved Sequence , DNA Methylation , DNA, Complementary/genetics , Female , Humans , Molecular Sequence Data , Neoplasm Proteins/chemistry , Nuclear Proteins/chemistry , Proteins/chemistry , Proteins/genetics , Sequence Alignment , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL