Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
PLoS One ; 18(4): e0271354, 2023.
Article in English | MEDLINE | ID: mdl-37022995

ABSTRACT

Tumors rich in stroma are associated with advanced stage and poor prognosis in colorectal adenocarcinoma (CRC). Abundance of stromal cells also has implications for genomic analysis of patient tumors as it may prevent detection of somatic mutations. As part of our efforts to interrogate stroma-cancer cell interactions and to identify actionable therapeutic targets in metastatic CRC, we aimed to determine the proportion of stroma embedded in hepatic CRC metastases by performing computational tumor purity analysis based on whole exome sequencing data (WES). Unlike previous studies focusing on histopathologically prescreened samples, we used an unbiased in-house collection of tumor specimens. WES from CRC liver metastasis samples were utilized to evaluate stromal content and to assess the performance of three in silico tumor purity tools, ABSOLUTE, Sequenza and PureCN. Matching tumor derived organoids were analyzed as a high purity control as they are enriched in cancer cells. Computational purity estimates were compared to those from a histopathological assessment conducted by a board-certified pathologist. According to all computational methods, metastatic specimens had a median tumor purity of 30% whereas the organoids were enriched for cancer cells with a median purity estimate of 94%. In line with this, variant allele frequencies (VAFs) of oncogenes and tumor suppressor genes were undetectable or low in most patient tumors, but higher in matching organoid cultures. Positive correlation was observed between VAFs and in silico tumor purity estimates. Sequenza and PureCN produced concordant results whereas ABSOLUTE yielded lower purity estimates for all samples. Our data shows that unbiased sample selection combined with molecular, computational, and histopathological tumor purity assessment is critical to determine the level of stroma embedded in metastatic colorectal adenocarcinoma.


Subject(s)
Adenocarcinoma , Colorectal Neoplasms , Liver Neoplasms , Humans , Exome Sequencing , Mutation , Exome/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Adenocarcinoma/genetics , Liver Neoplasms/genetics
2.
J Cell Mol Med ; 26(16): 4591-4601, 2022 08.
Article in English | MEDLINE | ID: mdl-35841281

ABSTRACT

Tumour Necrosis Factor (TNF) potently induces a transient inflammatory response that must be downregulated once any invasive stimulus has resolved. Yet, how TNF-induced inflammation is shut down in normal cells is incompletely understood. The present study shows that STAT3 was activated in mouse embryo fibroblasts (MEFs) by treatment with TNF or an agonist antibody to TNFR1. STAT3 activation was inhibited by pharmacological inhibition of the Jak2 tyrosine kinase that associates with TNFR1. To identify STAT3 target genes, global transcriptome analysis by RNA sequencing was performed in wild-type MEFs and MEFs from STAT3 knockout (STAT3KO ) mice that were stimulated with TNF, and the results were validated at the protein level by using multiplex cytokine assays and immunoblotting. After TNF stimulation, STAT3KO MEFs showed greater gene and protein induction of the inflammatory chemokines Ccl2, Cxcl1 and Cxcl10 than WT MEFs. These observations show that, by activating STAT3, TNF selectively modulates expression of a cohort of chemokines that promote inflammation. The greater induction by TNF of chemokines in STAT3KO than WT MEFs suggested that TNF induced an inhibitory protein in WT MEFs. Consistent with this possibility, STAT3 activation by TNFR1 increased the expression of Tnfaip3/A20, a ubiquitin modifying enzyme that inhibits inflammation, in WT MEFs but not in STAT3KO MEFs. Moreover, enforced expression of Tnfaip3/A20 in STAT3KO MEFs suppressed proinflammatory chemokine expression induced by TNF. Our observations identify Tnfaip3/A20 as a new downstream target for STAT3 which limits the induction of Ccl2, Cxcl1 and Cxcl10 and inflammation induced by TNF.


Subject(s)
Receptors, Tumor Necrosis Factor, Type I , Tumor Necrosis Factor-alpha , Animals , Gene Expression , Inflammation , Janus Kinase 2/metabolism , Mice , Mice, Knockout , NF-kappa B/metabolism , Receptors, Tumor Necrosis Factor, Type I/genetics , STAT3 Transcription Factor/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Tumor Necrosis Factor-alpha/metabolism
3.
PLoS One ; 14(7): e0219469, 2019.
Article in English | MEDLINE | ID: mdl-31291332

ABSTRACT

INTRODUCTION: Prognosis after resection of hepatocellular carcinoma (HCC) is highly variable. Compared to clinicopathologic factors, the use of molecular markers to predict outcome has not been well studied. We investigated the prognostic importance of thymidylate synthase (TS) gene expression and polymorphisms in patients after resection of HCC. METHODS: Patients who underwent complete resection of HCC for whom tissue was available were identified. TS gene expression level and polymorphisms were determined in HCC specimens. Prognostic factors were evaluated using Kaplan-Meier curves and Cox proportional hazard models. RESULTS: The study included 67 patients. In univariate analysis, variables that negatively influenced survival included TNM stage, microvascular invasion, and high TS expression. For the high TS expression group, median survival was 54 months and 5-year actuarial survival was 47%. For the low TS expression group, median survival was not reached and the 5-year actuarial survival was 91%. In multivariate analysis, only high TS expression remained an independent predictor of poor survival (HR = 10.77, 95% CI 1.36-84.91; P = 0.02). TS gene polymorphisms were not associated with TS expression or overall survival. CONCLUSIONS: High TS expression predicts poor outcome after resection of HCC. Molecular markers might be robust predictors of patient outcome after resection of HCC.


Subject(s)
Carcinoma, Hepatocellular/surgery , Liver Neoplasms/surgery , Thymidylate Synthase/genetics , Aged , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Kaplan-Meier Estimate , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , Prognosis , Proportional Hazards Models , Treatment Outcome
4.
Transl Oncol ; 12(3): 550-560, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30639964

ABSTRACT

PURPOSE: Prognostic schemes that rely on clinical variables to predict outcome after resection of colorectal metastases remain imperfect. We hypothesized that molecular markers can improve the accuracy of prognostic schemes. METHODS: We screened the transcriptome of matched colorectal liver metastases (CRCLM) and primary tumors from 42 patients with unresected CRCLM to identify differentially expressed genes. Among the differentially expressed genes identified, we looked for associations between expression and time to disease progression or overall survival. To validate such associations, mRNA levels of the candidate genes were assayed by qRT-PCR from CRCLM in 56 additional patients who underwent hepatectomy. RESULTS: Seven candidate genes were selected for validation based on their differential expression between metastases and primary tumors and a correlation between expression and surgical outcome: lumican; tissue inhibitor metalloproteinase 1; basic helix-loop-helix domain containing class B2; fibronectin; transmembrane 4 superfamily member 1; mitogen inducible gene 6 (MIG-6); and serpine 2. In the hepatectomy group, only MIG-6 expression was predictive of poor survival after hepatectomy. Quantitative PCR of MIG-6 mRNA was performed on 25 additional hepatectomy patients to determine if MIG-6 expression could substratify patients beyond the clinical risk score. Patients within defined clinical risk score categories were effectively substratified into distinct groups by relative MIG-6 expression. CONCLUSIONS: MIG-6 expression is inversely associated with survival after hepatectomy and may be used to improve traditional prognostic schemes that rely on clinicopathologic data such as the Clinical Risk Score.

5.
Mol Cancer Ther ; 17(12): 2702-2709, 2018 12.
Article in English | MEDLINE | ID: mdl-30254185

ABSTRACT

Patients with pancreatic neuroendocrine tumors (PNET) commonly develop advanced disease and require systemic therapy. However, treatment options remain limited, in part, because experimental models that reliably emulate PNET disease are lacking. We therefore developed a patient-derived xenograft model of PNET (PDX-PNET), which we then used to evaluate two mTOR inhibitor drugs: FDA-approved everolimus and the investigational new drug sapanisertib. PDX-PNETs maintained a PNET morphology and PNET-specific gene expression signature with serial passage. PDX-PNETs also harbored mutations in genes previously associated with PNETs (such as MEN1 and PTEN), displayed activation of the mTOR pathway, and could be detected by Gallium-68 DOTATATE PET-CT. Treatment of PDX-PNETs with either everolimus or sapanisertib strongly inhibited growth. As seen in patients, some PDX-PNETs developed resistance to everolimus. However, sapanisertib, a more potent inhibitor of the mTOR pathway, caused tumor shrinkage in most everolimus-resistant tumors. Our PDX-PNET model is the first available, validated PDX model for PNET, and preclinical data from the use of this model suggest that sapanisertib may be an effective new treatment option for patients with PNET or everolimus-resistant PNET.


Subject(s)
Benzoxazoles/therapeutic use , Drug Resistance, Neoplasm , Everolimus/therapeutic use , Neuroendocrine Tumors/drug therapy , Pancreatic Neoplasms/drug therapy , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Xenograft Model Antitumor Assays , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Humans , Mice, Nude , Neuroendocrine Tumors/diagnostic imaging , Neuroendocrine Tumors/pathology , Organometallic Compounds/chemistry , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/pathology , Positron Emission Tomography Computed Tomography , Protein Kinase Inhibitors/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism
6.
PLoS One ; 8(9): e73817, 2013.
Article in English | MEDLINE | ID: mdl-24040083

ABSTRACT

SALL2- a member of the Spalt gene family- is a poorly characterized transcription factor found deregulated in various cancers, which suggests it plays a role in the disease. We previously identified SALL2 as a novel interacting protein of neurotrophin receptors and showed that it plays a role in neuronal function, which does not necessarily explain why or how SALL2 is deregulated in cancer. Previous evidences indicate that SALL2 gene is regulated by the WT1 and AP4 transcription factors. Here, we identified SALL2 as a novel downstream target of the p53 tumor suppressor protein. Bioinformatic analysis of the SALL2 gene revealed several putative p53 half sites along the promoter region. Either overexpression of wild-type p53 or induction of the endogenous p53 by the genotoxic agent doxorubicin repressed SALL2 promoter activity in various cell lines. However R175H, R249S, and R248W p53 mutants, frequently found in the tumors of cancer patients, were unable to repress SALL2 promoter activity, suggesting that p53 specific binding to DNA is important for the regulation of SALL2. Electrophoretic mobility shift assay demonstrated binding of p53 to one of the identified p53 half sites in the Sall2 promoter, and chromatin immunoprecipitation analysis confirmed in vivo interaction of p53 with the promoter region of Sall2 containing this half site. Importantly, by using a p53ER (TAM) knockin model expressing a variant of p53 that is completely dependent on 4-hydroxy-tamoxifen for its activity, we show that p53 activation diminished SALL2 RNA and protein levels during genotoxic cellular stress in primary mouse embryo fibroblasts (MEFs) and radiosensitive tissues in vivo. Thus, our finding indicates that p53 represses SALL2 expression in a context-specific manner, adding knowledge to the understanding of SALL2 gene regulation, and to a potential mechanism for its deregulation in cancer.


Subject(s)
DNA Damage , Gene Expression Regulation/genetics , Intracellular Signaling Peptides and Proteins/genetics , Tumor Suppressor Protein p53/genetics , Animals , Antibiotics, Antineoplastic/pharmacology , Base Sequence , Binding Sites/genetics , Blotting, Western , Cell Line, Tumor , Cells, Cultured , DNA-Binding Proteins , Doxorubicin/pharmacology , Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , HCT116 Cells , HEK293 Cells , Humans , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation , Promoter Regions, Genetic/genetics , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , Transcription Factors , Tumor Suppressor Protein p53/metabolism
7.
Clin Cancer Res ; 19(20): 5777-87, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23983256

ABSTRACT

PURPOSE: The TP53 tumor suppressor is frequently mutated in colon cancer, but the influence of such mutations on survival remains controversial. We investigated whether mutations in the DNA-binding domain of TP53 are associated with survival in stage III colon cancer. EXPERIMENTAL DESIGN: The impact of TP53 genotype was prospectively evaluated in Cancer and Leukemia Group B 89803, a trial that randomized stage III colon cancer patients to receive adjuvant 5-fluorouracil/leucovorin (5FU/LV) or 5FU/LV with irinotecan (IFL). RESULTS: TP53 mutations were identified in 274 of 607 cases. The presence of any TP53 mutation did not predict disease-free survival (DFS) or overall survival with either adjuvant regimen when men and women were considered together or as separate groups. However, outcome differences among women became apparent when tumor TP53 genotype was stratified as wild-type versus zinc- or non-zinc-binding mutations in the TP53 DNA-binding domain. DFS at 5 years was 0.59, 0.52, and 0.78 for women with TP53 wild-type tumors, and tumors with zinc- or non-zinc-binding mutations, respectively. Survival at 5 years for these same women was 0.72, 0.59, and 0.90, respectively. No differences in survival by TP53 genotype were observed in men. CONCLUSIONS: The presence of any TP53 mutation within the DNA-binding domain did not predict survival in stage III colon cancer. However, TP53 genotype was predictive of survival in women following adjuvant therapy. Future colon cancer therapeutic trials, with inclusion of correlative molecular markers, should be designed to permit evaluation of survival and/or response to treatment in women separately from men.


Subject(s)
Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Mutation , Tumor Suppressor Protein p53/genetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chemotherapy, Adjuvant , Colonic Neoplasms/mortality , Colonic Neoplasms/therapy , DNA/chemistry , DNA/metabolism , Female , Genotype , Humans , Male , Microsatellite Instability , Models, Molecular , Molecular Conformation , Neoplasm Staging , Protein Binding , Sex Factors , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism , Zinc/chemistry , Zinc/metabolism
8.
Surgery ; 150(6): 1295-302, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22136853

ABSTRACT

BACKGROUND: Mitogen-inducible gene 6 (Mig-6) is a putative tumor suppressor gene and prognostic biomarker in papillary thyroid cancer. We hypothesized that Mig-6 knockout would activate pro-oncogenic signaling in mouse thyrocytes. METHODS: We performed a thyroid-specific knockout using the Cre/loxP recombinase system. RESULTS: Four knockout and 4 control mouse thyroids were harvested at 2 months of age. Immunoblotting confirmed Mig-6 ablation in knockout mice thyrocytes. Epidermal growth factor receptor (EGFR) and extracellular signal-regulated kinase (ERK) phosphorylation levels were increased in Mig-6 knockout compared to wild-type mice. Total EGFR levels were similar in knockout and wild-type mice. However, EGFR was absent in the caveolae-containing membrane fraction of knockout mice, indicating that Mig-6 depletion is associated with a change in the membrane distribution of EGFR. Although p65 localized to the nucleus in wild-type mice, it was distributed in both cytoplasm and nucleus in knockouts, suggesting that Mig-6 loss decreases p65 activity. CONCLUSION: Our results confirm the feasibility of targeted, thyroid-specific gene knockout as a strategy for studying the relevance of specific genes in thyroid oncogenesis. We suggest that the loss of Mig-6 alters the membrane distribution of EGFR, which may limit receptor degradation and activate this oncogenic signaling pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Epidermal Growth Factor/metabolism , Genes, Tumor Suppressor/physiology , NF-kappa B/metabolism , Signal Transduction/genetics , Thyroid Gland/metabolism , Thyroid Neoplasms/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Biomarkers/metabolism , Carcinoma , Carcinoma, Papillary , Gene Knockout Techniques , Intracellular Signaling Peptides and Proteins , Mice , Mice, Knockout , NF-kappa B/antagonists & inhibitors , Signal Transduction/physiology , Thyroid Cancer, Papillary , Thyroid Neoplasms/metabolism , Transcription Factor RelA/metabolism
9.
J Biol Chem ; 286(20): 17543-59, 2011 May 20.
Article in English | MEDLINE | ID: mdl-21402692

ABSTRACT

CYP3A4 expression in breast cancer correlates with decreased overall survival, but the mechanisms are unknown. Cytochrome P450 gene profiling by RNAi silencing demonstrates that CYP3A or 2C8 gene expression is specifically required for growth of the breast cancer lines MCF7, T47D, and MDA-MB-231. CYP3A4 silencing blocks the cell cycle at the G(2)/M checkpoint and induces apoptosis in the MCF7 line, thereby inhibiting anchorage-dependent growth and survival. CYP3A4 was profiled for NADPH-dependent arachidonic acid (AA) metabolism and synthesized AA epoxygenase products (±)-8,9-, (±)-11,12-, and (±)-14,15-epoxyeicosatrienoic acid (EET) (total turnover of ∼2 pmol/pmol CYP3A4/min) but not hydroxylase products (±)-15-, (±)-19-, or 20-hydroxyeicosatetraenoic acid. Furthermore, eicosanoid profiling revealed that MCF7 cells synthesize EETs in a CYP3A4-dependent manner. The (±)-14,15-EET regioisomer selectively rescues breast cancer cells from CYP3A4 silencing in a concentration-dependent fashion and promotes mitogenesis and anchorage-dependent cloning. Stat3 (Tyr-705) phosphorylation was inhibited by CYP3A4 silencing, providing a potential mechanism for CYP3A4 involvement in breast cancer cell growth. Silencing Stat3 blocks breast cancer cell growth and abrogates (±)-14,15-EET-induced proliferation, indicating a Stat3 requirement for (±)-14,15-EET-mediated cell growth. Although silencing of CYP3A4 reduces nuclear Tyr(P)-705-Stat3, (±)-14,15-EET restores this signaling process and promotes Tyr(P)-705-Stat3 translocation to the nucleus, suggesting that (±)-14,15-EET may be involved in an autocrine/paracrine pathway driving cell growth. These studies indicate that CYP3A4 is a highly active AA epoxygenase that promotes Stat3-mediated breast cancer cell growth in part through (±)-14,15-EET biosynthesis. Furthermore, these studies indicate an essential role for Stat3 as a mediator of epoxygenase activity in breast cancer.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Breast Neoplasms/metabolism , Cell Division , Cytochrome P-450 CYP3A/metabolism , G2 Phase , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , STAT3 Transcription Factor/metabolism , 8,11,14-Eicosatrienoic Acid/genetics , 8,11,14-Eicosatrienoic Acid/metabolism , Active Transport, Cell Nucleus/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cytochrome P-450 CYP3A/genetics , Female , Gene Silencing , Humans , Phosphorylation/genetics , STAT3 Transcription Factor/genetics , Signal Transduction/genetics
10.
J Clin Endocrinol Metab ; 96(3): E554-65, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21190978

ABSTRACT

CONTEXT: Low tumoral expression of mitogen-inducible gene-6 (Mig-6) is associated with papillary thyroid cancer (PTC) recurrence after thyroidectomy. OBJECTIVE: We hypothesize that Mig-6 behaves as a tumor suppressor in PTC. DESIGN: Mig-6 expression and promoter methylation status were compared in 31 PTC specimens with matched normal thyroid tissue from the same patient. The impact of Mig-6 loss and gain of function on nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) activation, global tyrosine kinase phosphorylation, and cellular invasion was determined in vitro. RESULTS: Mig-6 protein was abundant in all normal thyroid specimens, whereas 77% of PTC had low Mig-6 expression. Mig-6 promoter methylation was found in 79% of PTC with low Mig-6 expression. Low Mig-6 expression in PTC specimens was associated with low NF-κB activity but high levels of epidermal growth factor receptor (EGFR) and ERK phosphorylation. Mig-6 expression inversely correlated with PTC size but had no association with other clinicopathological variables including age, extrathyroidal extension, lymphovascular invasion, or histological subtype. Mig-6 knockdown in thyroid cancer cell lines resulted in EGFR phosphorylation and diminished NF-κB activity, whereas Mig-6 overexpression had the opposite effects. Mig-6 knockdown activated ErbB2, Met, and Src phosphorylation. Furthermore, Mig-6 regulated ERK phosphorylation independent from its effects on EGFR. Mig-6 knockdown promoted cellular proliferation, as determined by clonogenic survival. Lastly, Mig-6 knockdown increased matrix metalloproteinase-2 and -9 activities and increased cellular invasion. CONCLUSIONS: Mig-6 has tumor suppressor-like activity in PTC. In vivo studies are required to confirm that Mig-6 is a putative tumor suppressor in PTC, and future studies should investigate the utility of Mig-6 as a diagnostic marker.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Carcinoma, Papillary/genetics , Genes, Tumor Suppressor/physiology , Tumor Suppressor Proteins/genetics , Blotting, Western , Carcinoma , Carcinoma, Papillary/pathology , Cell Line, Tumor , Cell Nucleus/chemistry , Cells, Cultured , Cytosol/chemistry , DNA, Neoplasm/genetics , DNA, Neoplasm/isolation & purification , Diffusion Chambers, Culture , Down-Regulation , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , NF-kappa B/genetics , Neoplasm Invasiveness/genetics , Phosphorylation , Reverse Transcriptase Polymerase Chain Reaction , Thyroid Cancer, Papillary , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Tumor Stem Cell Assay
11.
Mol Cancer Res ; 8(9): 1217-26, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20736296

ABSTRACT

Although autophagy is generally considered a prosurvival mechanism that preserves viability, there is evidence that it could drive an alternative programmed cell death pathway in cells with defects in apoptosis. Because the inhibition of autophagic activity promotes resistance to both chemotherapy and external beam radiation in papillary thyroid cancer (PTC), we determined if RAD001, a potent activator of autophagy, improves the efficacy of either therapy. We found that RAD001 increased the expression level of light chain 3-II, a marker for autophagy, as well as autophagosome formation in cell lines and in human PTC ex vivo. RAD001 sensitized PTC to doxorubicin and external beam radiation in a synergistic fashion, suggesting that combination therapy could improve therapeutic response at less toxic concentrations. The effects of RAD001 were abrogated by RNAi knockdown of the autophagy-related gene 5, suggesting that RAD001 acts, in part, by enhancing autophagy. Because the synergistic activity of RAD001 with doxorubicin and external radiation suggests distinct and complementary mechanisms of action, we characterized how autophagy modulates signaling pathways in PTC. To do so, we performed kinome profiling and discovered that autophagic activation resulted in Src phosphorylation and Met dephosphorylation. Src inhibition did not reverse the effects of RAD001, whereas Met inhibition reversed the effects of autophagy blockade on chemosensitivity. These results suggest that the anticancer effects of autophagic activation are mediated largely through Met. We conclude that RAD001 induces autophagy, which enhances the therapeutic response to cytotoxic chemotherapy and external beam radiation in PTC.


Subject(s)
Autophagy/drug effects , Carcinoma, Papillary/pathology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Radiation Tolerance/drug effects , Sirolimus/analogs & derivatives , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Autophagy-Related Protein 5 , Carcinoma, Papillary/enzymology , Carcinoma, Papillary/metabolism , Cell Count , Cell Line, Tumor , Cell Survival/drug effects , Doxorubicin/pharmacology , Drug Screening Assays, Antitumor , Drug Synergism , Everolimus , Humans , Microtubule-Associated Proteins/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-met/metabolism , RNA Interference/drug effects , Radiation-Sensitizing Agents/pharmacology , Sirolimus/pharmacology , Thyroid Neoplasms/enzymology , Vacuoles/drug effects , Vacuoles/ultrastructure , src-Family Kinases/metabolism
12.
J Surg Res ; 163(1): 47-51, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20599218

ABSTRACT

BACKGROUND: Patients with neuroendocrine tumors (NETs) may have metastatic disease and unknown primary site. NETs commonly arise from the bronchopulmonary (BP) and gastrointestinal (GI) tract. The largest subgroups of well-differentiated BP-NETs are typical carcinoids (TCs). The homeodomain transcription factor NKX2.2 regulates development of gut serotonin cells and is a marker of GI-NETs. Previous work on a limited number of samples suggested that BP-TCs do not express NKX2.2. We hypothesized that lack of NKX2.2 expression in BP-TCs might be useful to distinguish BP- from GI-NETs, and evaluated NKX2.2 expression in a larger number of BP-TCs. METHODS: Archived formalin-fixed, paraffin-embedded tissues were obtained from 13 previously undescribed patients with BP-TCs. Expression of NKX2.2, serotonin, and the NE marker chromogranin A (CgA) were assessed by immunohistochemistry. RESULTS: CgA expression was robust in all 13 BP-TCs, confirming the NE phenotype. Serotonin expression was less frequent (9/13; 69%). Two patients with BP-TCs in which serotonin expression was absent exhibited Cushing's syndrome due to ectopic ACTH production. NKX2.2 expression was not observed in any of the 13 tumors. CONCLUSIONS: Bronchopulmonary TCs uniformly express CgA but not NKX2.2. Because most of these tumors express serotonin, our findings suggest that NKX2.2 may not be required for serotonin production by BP-TCs. We conclude that the presence or absence of NKX2.2 expression may assist in the determination of the primary tumor site in patients with NET metastases of unknown origin. NET metastases that are CgA-positive/NKX2.2-negative would suggest a BP primary, whereas those that are CgA-positive/NKX2.2-positive would suggest a GI primary.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoid Tumor/metabolism , Gastrointestinal Neoplasms/metabolism , Homeodomain Proteins/metabolism , Lung Neoplasms/metabolism , Transcription Factors/metabolism , Adolescent , Adult , Aged , Carcinoid Tumor/diagnosis , Female , Gastrointestinal Neoplasms/diagnosis , Homeobox Protein Nkx-2.2 , Humans , Immunohistochemistry , Lung Neoplasms/diagnosis , Male , Middle Aged , Nuclear Proteins , Young Adult , Zebrafish Proteins
13.
Endocr Relat Cancer ; 17(1): 283-91, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20048018

ABSTRACT

Neuroendocrine (NE) or carcinoid tumors of the small intestine (SI) frequently metastasize and produce the hormone serotonin, causing significant morbidity and mortality. A member of the ETS oncogene family of transcription factors, Fev, acts with the homeodomain transcription factor Nkx2.2 in the development of serotonin neurons in mice. In this study, we investigated the role of Fev in normal and neoplastic SI. In NE tumors (NETs) of the SI, serotonin stimulates tumor growth and causes debilitating symptoms, such as diarrhea, flushing, wheezing, and right-sided valvular heart disease (i.e. carcinoid syndrome). Compared with those in the matched normal human SI, FEV expression levels were significantly elevated in primary NETs (20-fold, P<0.0001), lymph node metastases (35-fold, P=0.004), and NET liver metastases (22-fold, P<0.0001) resected from patients with serotonin excess. Fev is expressed in the wild type but not in Nkx2.2 (-/-) mouse SI, in which cells producing serotonin are absent. Using recombination-based cell lineage tracing, we found that FEV-positive cells give rise to serotonin-producing cells in the SI. In Fev (-/-) mouse SI, we observed no difference in the number of cells producing serotonin or other hormones. We conclude that FEV expression identifies serotonin-producing cells in normal and neoplastic SI and is a novel target for diagnosis of patients with NETs of the SI.


Subject(s)
DNA-Binding Proteins/physiology , Enteroendocrine Cells/metabolism , Intestinal Neoplasms/pathology , Intestine, Small/metabolism , Neuroendocrine Tumors/pathology , Nuclear Proteins/physiology , Serotonin/metabolism , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoid Tumor/genetics , Carcinoid Tumor/metabolism , Carcinoid Tumor/pathology , Case-Control Studies , Cell Separation/methods , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enteroendocrine Cells/cytology , Enteroendocrine Cells/pathology , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Intestinal Neoplasms/genetics , Intestinal Neoplasms/metabolism , Intestine, Small/cytology , Intestine, Small/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Mice , Mice, Transgenic , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish Proteins
14.
Surgery ; 146(6): 1208-14, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19958950

ABSTRACT

BACKGROUND: Autophagy is a conserved response to stress that facilitates cell survival in some contexts and promotes cell death in others. We sought to characterize autophagy in papillary thyroid cancer (PTC), and to determine the effects of autophagy inhibition on chemosensitivity and radiosensitivity. METHODS: The human thyroid papillary carcinoma cell lines TPC-1 and 8505-C were treated with doxorubicin or radiation in the presence or absence of the autophagy-specific inhibitor 3-methyladenine (3-MA). RESULTS: Although light chain 3 (LC3)-II protein levels were undetectable in normal thyroid and PTC specimens at baseline, doxorubicin exposure induced LC3-II expression and the formation of autophagosomes. Both PTC cell lines expressed low levels of LC3-II under standard conditions. Treatment of these cells with doxorubicin strongly induced LC3-II expression and the formation of autophagosomes; however, doxorubicin-mediated induction of LC3-II was abrogated by 3-MA. Moreover, 3-MA significantly increased the doxorubicin IC(50) in both PTC cell lines. Radiation exposure also induced LC3-II expression. Treatment with 3-MA abrogated the radiation-induced increase in LC3-II in both cell lines and reduced radiosensitivity by 49% and 31% in 8505-C and TPC-1 cells, respectively. CONCLUSION: Autophagy inhibition promotes PTC resistance to doxorubicin and radiation. Therefore, autophagy activation may be a useful adjunct treatment for patients with PTC that is refractory to conventional therapy.


Subject(s)
Autophagy , Carcinoma, Papillary/drug therapy , Carcinoma, Papillary/radiotherapy , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/radiotherapy , Adenine/analogs & derivatives , Adenine/pharmacology , Antibiotics, Antineoplastic/pharmacology , Autophagy/drug effects , Carcinoma, Papillary/metabolism , Carcinoma, Papillary/pathology , Cell Line, Tumor , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Humans , In Vitro Techniques , Microtubule-Associated Proteins/metabolism , Radiation Tolerance/drug effects , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology
15.
Mol Cancer Res ; 7(10): 1655-62, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19825987

ABSTRACT

Although most patients with papillary thyroid cancer (PTC) have favorable outcomes, some have advanced PTC that is refractory to external beam radiation and systemic chemotherapy. Galectin-3 (Gal-3) is a beta-galactoside-binding protein with antiapoptotic activity that is consistently overexpressed in PTC. The purpose of this study is to determine if Gal-3 inhibition promotes apoptosis, chemosensitivity, and radiosensitivity in PTC. PTC cell lines (8505-C and TPC-1) and human ex vivo PTC were treated with a highly specific small molecule inhibitor of Gal-3 (Td131_1). Apoptotic activity was determined by flow cytometric analysis as well as caspase-3 and PARP cleavage. The minimum inhibitory concentrations of Td131_1 and doxorubicin were determined, and their combined effects were measured to test for synergistic activity. The effects of Td131_1 on radiosensitivity were determined by a clonogenic assay. Td131_1 promoted apoptosis, improved radiosensitivity, and synergistically enhanced chemosensitivity to doxorubicin in PTC cell lines. In PTC ex vivo, Td131_1 treatment alone induced the cleavage of caspase-3 and PARP. Td131_1 and doxorubicin together activated apoptosis in PTC ex vivo to a greater degree than their combined individual effects. Td131_1 activated apoptosis and had synergistic activity with doxorubicin in PTC. We conclude that Gal-3 targeted therapy is a promising therapeutic strategy for advanced PTC that is refractory to surgery and radioactive iodine therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Papillary/drug therapy , Galectin 3/antagonists & inhibitors , Radiation-Sensitizing Agents/therapeutic use , Thioglycosides/therapeutic use , Thyroid Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Apoptosis/physiology , Carcinoma, Papillary/metabolism , Carcinoma, Papillary/radiotherapy , Caspase 3/drug effects , Caspase 3/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/physiology , Drug Synergism , Flow Cytometry , Galectin 3/metabolism , Humans , Molecular Structure , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Radiation Tolerance/drug effects , Radiation Tolerance/physiology , Radiation-Sensitizing Agents/chemical synthesis , Rats , Thiogalactosides/chemical synthesis , Thiogalactosides/therapeutic use , Thioglycosides/chemical synthesis , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/radiotherapy
16.
J Biol Chem ; 284(23): 15390-9, 2009 Jun 05.
Article in English | MEDLINE | ID: mdl-19324884

ABSTRACT

Tissue transglutaminase 2 (TG2) is overexpressed in epithelial ovarian cancer (EOC) and promotes intraperitoneal metastasis. How TG2 facilitates the spread of EOC is unknown. Here, we show that TG2 regulates the expression and function of matrix metalloproteinase-2 (MMP-2), a critical mediator of tissue invasiveness. TG2 knockdown down-regulates MMP-2 protein and mRNA expression in SKOV3, IGROV-1, MDA-MB-436, and PC-3 cancer cells. TG2 knockdown or inhibition of TG2 activity using KCC009 decreases MMP-2 gelatinase activity in cancer cells. MMP-2 expression and function are regulated by TG2 at transcriptional level, as demonstrated by quantitative PCR and reporter assays. We used bioinformatics and chromatin immunoprecipitation to identify a CREB binding site in the MMP-2 promoter. Binding of CREB to the MMP-2 promoter was diminished in cells that expressed decreased TG2 levels. TG2 knockdown decreased CREB phosphorylation, and CREB knockdown decreased MMP-2 expression. The effect of TG2 on CREB activity and MMP-2 transcription is mediated by TG2-dependent degradation of protein phosphatase 2 (PP2A-alpha). We show that PP2A-alpha complexes with and is targeted for degradation by TG2. In addition to their related in vitro expression levels, TG2 and MMP-2 expression were significantly correlated in vivo, as shown by concordant immunostaining in peritoneal xenografts and in human ovarian tumors. The capacity of TG2 to regulate MMP-2 expression in vitro and in vivo identifies a mechanism that may facilitate tissue invasion and the spread of EOC. The demonstration that TG2 induced degradation of PP2A-alpha activates CREB, and thereby increases MMP-2 transcription, provides novel mechanistic insight into the pro- metastatic function of TG2.


Subject(s)
Cyclic AMP Response Element Modulator/metabolism , Matrix Metalloproteinase 2/genetics , Ovarian Neoplasms/metabolism , Transglutaminases/genetics , Cell Line, Tumor , Cloning, Molecular , Cyclic AMP Response Element-Binding Protein/metabolism , DNA Primers , Female , GTP-Binding Proteins , Humans , Matrix Metalloproteinase 2/metabolism , Neoplasm Metastasis , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Protein Glutamine gamma Glutamyltransferase 2 , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Transglutaminases/deficiency
17.
Biochem Biophys Res Commun ; 379(2): 626-31, 2009 Feb 06.
Article in English | MEDLINE | ID: mdl-19124005

ABSTRACT

A subset of patients with papillary thyroid cancer (PTC) present with aggressive disease that is refractory to conventional treatment. Novel therapies are needed to treat this group of patients. Galectin-3 (Gal-3) is a beta-galactoside-binding protein with anti-apoptotic activity. Over 30 studies in the last 3 years have reported that Gal-3 is highly expressed in PTC relative to normal thyrocytes. In this study, we show that Gal-3 silencing with RNA interference stimulates apoptosis, while Gal-3 overexpression protects against both TRAIL- and doxorubicin-induced apoptosis in PTC cells. The anti-apoptotic activity and chemoresistance related to Gal-3 function can be partially reversed through the inhibition of the PI3K-Akt pathway, suggesting that Gal-3 acts, at least in part, on the PI3K-Akt axis. These observations support further evaluation of Gal-3 as a potential therapeutic target in patients with aggressive PTC.


Subject(s)
Adenocarcinoma, Papillary/metabolism , Antibiotics, Antineoplastic/pharmacology , Apoptosis/genetics , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/genetics , Galectin 3/physiology , Thyroid Neoplasms/metabolism , Adenocarcinoma, Papillary/pathology , Cell Line, Tumor , Galectin 3/genetics , Humans , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Thyroid Neoplasms/pathology
18.
EMBO J ; 28(3): 261-73, 2009 Feb 04.
Article in English | MEDLINE | ID: mdl-19131967

ABSTRACT

By screening a fetal brain two-hybrid library with the death domain of the p75 neurotrophin receptor (NTR), we identified the Sall2 transcription factor as a novel interacting protein. Sall2 is a unique member of the Sall gene family, which is believed to be a tumour suppressor. Here, we show that Sall2 contains a p75NTR interaction domain not found in other Sall proteins and that p75NTR/Sall2 complexes co-immunoprecipitate from brain lysates. NGF dissociates p75NTR/Sall2 complexes and activates TrkA, which has an obligate function in the nuclear translocation of Sall2. NGF also increases Sall2 expression and this is mediated by p75NTR, but may not require TrkA. Depletion of Sall2 from cells decreases the expression and activity of p21(WAF1/CIP1), as well as the ability of NGF to induce growth arrest and the development of neurites. Overexpression of Sall2 activates p21(WAF1/CIP1), induces growth arrest, and promotes neurite outgrowth independently of NGF. These data establish Sall2 as a link between NTRs and transcriptional events that regulate the growth and development of neuronal cells.


Subject(s)
Cell Cycle/drug effects , Nerve Growth Factor/pharmacology , Neurites/drug effects , Neurites/metabolism , Receptor, Nerve Growth Factor/metabolism , Signal Transduction/drug effects , Transcription Factors/metabolism , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA-Binding Proteins , Gene Silencing/drug effects , Hippocampus/cytology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Neurons/cytology , Neurons/drug effects , PC12 Cells , Protein Binding/drug effects , Protein Structure, Tertiary , Protein Transport/drug effects , Rats , Receptor, Nerve Growth Factor/chemistry , Receptor, trkA/metabolism
19.
Endocr Relat Cancer ; 16(1): 267-79, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18987169

ABSTRACT

The homeodomain transcription factor NKX2.2 is necessary for neuroendocrine (NE) differentiation in the central nervous system and pancreas. NE tumors derived from the gut are defined by their NE phenotype, which is used for diagnosis and contributes to tumorigenicity. We hypothesized that NKX2.2 is important for NE differentiation in normal and neoplastic gut. NKX2.2 and NE marker expression was investigated in the small intestine of embryonic and adult mice using immunofluorescence (IF). To determine the role of NKX2.2 in NE differentiation of the intestine, the phenotype of Nkx2.2 (-/-) mice was examined by IF and real-time (RT)-PCR. NKX2.2 and NE marker expression in human NE tumors of the gut and normal tissues were evaluated by immunohistochemistry and qRT-PCR. NKX2.2 expression was detected in the intervillus/crypt regions of embryonic and adult mouse intestine. Co-expression of Nkx2.2 with neurogenin3 (NEUROG3) and hormones was observed in the adult intestinal crypt compartment, suggesting NKX2.2 functions in NEUROG3-positive endocrine progenitors and newly differentiated endocrine cells. In the intestine of Nkx2.2 (-/-) mice, we found a dramatic reduction in the number of cells producing numerous hormones, such as serotonin, gastrin, cholecystokinin, somatostatin, glucagon-like peptide 1 (GLP-1), and secretin, but an increase in cells producing ghrelin. NKX2.2 was expressed in most (24 of 29) human NE tumors derived from diverse primary sites. We conclude NKX2.2 functions in immature endocrine cells to control NE differentiation in normal intestine and is expressed in most NE tumors of the gut, and is therefore a novel target of diagnosis for patients with gastrointestinal NE tumors.


Subject(s)
Gastrointestinal Neoplasms/genetics , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Neuroendocrine Tumors/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Apoptosis/physiology , Cell Differentiation/physiology , Cell Division/physiology , Endocrine Cells/cytology , Endocrine Cells/physiology , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Neoplasms/pathology , Ghrelin/metabolism , Homeobox Protein Nkx-2.2 , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/embryology , Intestine, Small/cytology , Intestine, Small/embryology , Mice , Mice, Mutant Strains , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Nuclear Proteins , Zebrafish Proteins
20.
Surgery ; 144(6): 908-13; discussion 913-4, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19040996

ABSTRACT

BACKGROUND: Mitogen-inducible gene-6 (Mig-6) is an immediate early response gene that negatively regulates signaling. EGFR overexpression and activating mutations in MAPK signaling effectors are common events in papillary thyroid cancer (PTC). The purpose of this study was to determine if Mig-6 expression is associated with EGFR expression or surgical outcomes in PTC. METHODS: We determined Mig-6 transcript levels from a microarray in 19 patients with PTC who underwent thyroidectomy. We established a maximally selected cutoff to discriminate Kaplan-Meier survival estimates. For cross-validation, we performed quantitative RT-PCR on resected well-differentiated PTC from an additional 106 patients. RESULTS: Mig-6 and EGFR mRNA levels correlated directly (P < .0001). Mig-6 expression above the cutoff of 1.10 (2;-dCt[Mig6-GUS]) was associated with greater survival (P = .008). When this cutoff was applied in the cross-validation, high Mig-6 expression was associated with longer survival (P = .03) and disease-free survival (P = .07). Furthermore, high Mig-6 expression was independently predictive of greater disease-free survival in BRAF(V600E)-positive PTC. CONCLUSION: High Mig-6 expression in PTC is associated with favorable outcomes. Mig-6 is a novel tumor suppressor that may be a candidate for targeted cancer therapeutics in patients with PTC refractory to conventional therapy.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adenocarcinoma, Papillary/genetics , Neoplasm Recurrence, Local/genetics , Proto-Oncogene Proteins B-raf/metabolism , Thyroid Neoplasms/genetics , Adaptor Proteins, Signal Transducing/biosynthesis , Adenocarcinoma, Papillary/metabolism , Adenocarcinoma, Papillary/surgery , Adult , Biomarkers, Tumor/analysis , ErbB Receptors/biosynthesis , Female , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Predictive Value of Tests , Survival Analysis , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/surgery , Thyroidectomy , Treatment Outcome , Tumor Suppressor Proteins , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...