Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
2.
Neurotox Res ; 40(5): 1526-1536, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35922689

ABSTRACT

Ferroptosis is a necrotic cell death caused by lipid oxidation that may be responsible for neural degeneration in Parkinson's disease. We assessed whether three neuronal cell lines are sensitive to killing by ferroptosis. Ferroptosis inducer erastin killed LUHMES neurons at sub-micromolar concentrations, whereas neuronal cells derived from SH-SY5Y cells or neural stem cells were at least 50-fold less sensitive. LUHMES differentiated neurons were likewise sensitive to killing by RSL3 or ML210, inhibitors of the glutathione peroxidase 4 enzyme (GPX4) that consumes GSH to detoxify lipid peroxides. Additional assays showed that erastin, RSL3, and ML210 increased lipid peroxide levels, and that LUHMES neurons were protected from both peroxide accumulation and cell death by ferrostatin-1. A possible role of iron was assessed by evaluating the effects of five metal chelators on cytotoxicity of erastin and RSL3. LUHMES neurons were protected from RSL3 by three of the chelators, 2,3-dimercapto-1-propanesulfonic acid (DMPS), deferoxiprone (DFX), and deferiprone (DFP). Collectively, these results demonstrate the vulnerability of LUHMES neurons to ferroptosis by chemical treatments that disrupt glutathione synthesis, lipid peroxide detoxification, or iron metabolism. The same vulnerabilities may occur in CNS neurons, which reportedly generate low levels of GSH and metallothioneins, limiting their ability to neutralize oxidative stresses and toxic metals. These results suggest a rationale and methods to search for environmental toxicants that may exploit these vulnerabilities and promote neurodegenerative diseases.


Subject(s)
Ferroptosis , Neuroblastoma , Humans , Carbolines/toxicity , Chelating Agents , Deferiprone , Dopaminergic Neurons/metabolism , Glutathione/metabolism , Iron/metabolism , Iron/toxicity , Lipid Peroxides , Phospholipid Hydroperoxide Glutathione Peroxidase , Unithiol
3.
Oncotarget ; 12(21): 2114-2130, 2021 Oct 12.
Article in English | MEDLINE | ID: mdl-34676045

ABSTRACT

The therapeutic efficacy of temozolomide (TMZ) is hindered by inherent and acquired resistance. Biomarkers such as MGMT expression and MMR proficiency are used as predictors of response. However, not all MGMTlow/-ve/MMRproficient patients benefit from TMZ treatment, indicating a need for additional patient selection criteria. We explored the role of ATR in mediating TMZ resistance and whether ATR inhibitors (ATRi) could reverse this resistance in multiple cancer lines. We observed that only 31% of MGMTlow/-ve/MMRproficient patient-derived and established cancer lines are sensitive to TMZ at clinically relevant concentrations. TMZ treatment resulted in DNA damage signaling in both sensitive and resistant lines, but prolonged G2/M arrest and cell death were exclusive to sensitive models. Inhibition of ATR but not ATM, sensitized the majority of resistant models to TMZ and resulted in measurable DNA damage and persistent growth inhibition. Also, compromised homologous recombination (HR) via RAD51 or BRCA1 loss only conferred sensitivity to TMZ when combined with an ATRi. Furthermore, low REV3L mRNA expression correlated with sensitivity to the TMZ and ATRi combination in vitro and in vivo. This suggests that HR defects and low REV3L levels could be useful selection criteria for enhanced clinical efficacy of an ATRi plus TMZ combination.

4.
Clin Epigenetics ; 8: 38, 2016.
Article in English | MEDLINE | ID: mdl-27081402

ABSTRACT

BACKGROUND: Oncogene overexpression in primary cells often triggers the induction of a cellular safeguard response promoting senescence or apoptosis. Secondary cooperating genetic events are generally required for oncogene-induced tumorigenesis to overcome these biologic obstacles. We employed comparative genomic hybridization for eight genetically engineered mouse models of mammary cancer to identify loci that might harbor genes that enhance oncogene-induced tumorigenesis. RESULTS: Unlike many other mammary tumor models, the MMTV-Myc tumors displayed few copy number variants except for amplification of distal mouse chromosome 11 in 80 % of the tumors (syntenic to human 17q23-qter often amplified in human breast cancer). Analyses of candidate genes located in this region identified JMJD6 as an epigenetic regulatory gene that cooperates with Myc to enhance tumorigenesis. It suppresses Myc-induced apoptosis under varying stress conditions through inhibition of p19ARF messenger RNA (mRNA) and protein, leading to reduced levels of p53. JMJD6 binds to the p19ARF promoter and exerts its inhibitory function through demethylation of H4R3me2a. JMJD6 overexpression in MMTV-Myc cell lines increases tumor burden, induces EMT, and greatly enhances tumor metastasis. Importantly, we demonstrate that co-expression of high levels of JMJD6 and Myc is associated with poor prognosis for human ER+ breast cancer patients. CONCLUSIONS: A novel epigenetic mechanism has been identified for how JMJD6 cooperates with Myc during oncogenic transformation. Combined high expression of Myc and JMJD6 confers a more aggressive phenotype in mouse and human tumors. Given the pleiotropic pro-tumorigenic activities of JMJD6, it may be useful as a prognostic factor and a therapeutic target for Myc-driven mammary tumorigenesis.


Subject(s)
Breast Neoplasms/pathology , Cell Transformation, Neoplastic/genetics , Epigenesis, Genetic , Jumonji Domain-Containing Histone Demethylases/genetics , Mammary Neoplasms, Experimental/pathology , Proto-Oncogene Proteins c-myc/genetics , Animals , Apoptosis , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Comparative Genomic Hybridization , Female , Gene Expression Regulation, Neoplastic , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mice , Neoplasm Metastasis , Prognosis , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction
5.
J Clin Invest ; 124(1): 156-68, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24316974

ABSTRACT

Breast cancer (BC) can recur as metastatic disease many years after primary tumor removal, suggesting that disseminated tumor cells survive for extended periods in a dormant state that is refractory to conventional therapies. We have previously shown that altering the tumor microenvironment through fibrosis with collagen and fibronectin deposition can trigger tumor cells to switch from a dormant to a proliferative state. Here, we used an in vivo preclinical model and a 3D in vitro model of dormancy to evaluate the role of Src family kinase (SFK) in regulating this dormant-to-proliferative switch. We found that pharmacological inhibition of SFK signaling or Src knockdown results in the nuclear localization of cyclin-dependent kinase inhibitor p27 and prevents the proliferative outbreak of dormant BC cells and metastatic lesion formation; however, SFK inhibition did not kill dormant cells. Dormant cell proliferation also required ERK1/2 activation. Combination treatment of cells undergoing the dormant-to-proliferative switch with the Src inhibitor (AZD0530) and MEK1/2 inhibitor (AZD6244) induced apoptosis in a large fraction of the dormant cells and delayed metastatic outgrowth, neither of which was observed with either inhibitor alone. Thus, targeting Src prevents the proliferative response of dormant cells to external stimuli, but requires MEK1/2 inhibition to suppress their survival. These data indicate that treatments targeting Src in combination with MEK1/2 may prevent BC recurrence.


Subject(s)
Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Benzodioxoles/pharmacology , Breast Neoplasms/drug therapy , Lung Neoplasms/drug therapy , MAP Kinase Kinase Kinases/antagonists & inhibitors , Quinazolines/pharmacology , src-Family Kinases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Cell Survival/drug effects , Female , Gene Knockdown Techniques , Humans , Lung Neoplasms/secondary , Mice , Mice, Nude , Neoplasm Recurrence, Local/prevention & control , RNA, Small Interfering/genetics , Xenograft Model Antitumor Assays , src-Family Kinases/genetics , src-Family Kinases/metabolism
6.
PLoS One ; 8(4): e61125, 2013.
Article in English | MEDLINE | ID: mdl-23577196

ABSTRACT

Transforming growth factor (ß1TGFß1) can promote proliferation in late stage cancers but acts as a tumor suppressor in normal epithelial cells and in early stage cancers. Although, the TGFß pathway has been shown to play a key role in tumorigenesis and metastasis, only a limited number of models have been developed to understand this process. Here, we present a novel model system to discern this paradoxical role of TGFß1 using the MDA-MB-231 (MB-231) cell line. The MB-231 triple-negative breast cancer cell line has been extensively characterized and has been shown to continue to proliferate and undergo epithelial-to-mesenchymal transition (EMT) upon TGFß1 stimulation. We have previously shown by microarray analysis that expression of GATA3 in MB-231 cells results in reprogramming of these cells from a basal to a luminal subtype associated with a reduction of metastasis and tumorigenesis when implanted as xenografts. We now demonstrate that GATA3 overexpression in these cells results in a reduction of TGFß1 response, reversal of EMT, and most importantly, restoration of sensitivity to the inhibitory effects on proliferation of TGFß1. Microarray analysis revealed that TGFß1 treatment resulted in reduction of several cell cycle effectors in 231-GATA3 cells but not in control cells. Furthermore, our microarray analysis revealed a significant increase of BMP5 in 231-GATA3 cells. We demonstrate that combined treatment of MB-231 control cells with TGFß1 and BMP5 results in a significant reduction of cellular proliferation. Thus, this model offers a means to further investigate potentially novel mechanisms involved in the switch in response to TGFß1 from tumor promoter to tumor suppressor through the reprogramming of a triple-negative breast cancer cell line by the GATA3 transcription factor.


Subject(s)
Breast Neoplasms/pathology , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , Transforming Growth Factor beta/pharmacology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Bone Morphogenetic Protein 5/metabolism , Breast Neoplasms/genetics , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Receptor, ErbB-2/genetics , Receptors, Estrogen/genetics , Receptors, Progesterone/genetics , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Transforming Growth Factor beta/metabolism
7.
Rejuvenation Res ; 13(5): 527-37, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20818983

ABSTRACT

Following spinal cord injury (SCI) or peripheral neuropathy, increased levels of the p75(NTR) death receptor initiate the signal transduction cascade leading to cell death. Investigations of compounds that may ameliorate neuronal cell death have largely used rodent models, which are time consuming, expensive, and cumbersome to perform. Previous studies had demonstrated that steroids, particularly dexamethasone and its analog methylprednisolone sodium succinate, exhibit limited neuroprotective effects against neuronal injury. Significantly, many naturally occurring nonsteroidal plant compounds exhibit structural overlap with steroids. In this report, we present an in vitro cellular screen model to practically examine the efficacy of various phytoestrogens in modulating the ibuprofen-induced expression of p75(NTR) and reduced cell survival of CCFSTTG1 and U87MG cells in a rescue (postinjury) or prevention (preinjury) regimen. We show that the phytoestrogen, biochanin A, and, to a lesser extent, genistein are more effective than dexamethasone at reducing p75(NTR) expression and improving the viability of U87MG and CCFSTTG1 before and after p75(NTR) induction. Furthermore, these studies implicate biochanin A's inactivation of p38-MAPK as a possible contributor to reducing p75(NTR) with associated increased cell survival. This new in vitro assay facilitates a more time-efficient screening of compounds to suppress p75(NTR) expression and increase neuronal cell viability prior to their evaluation in animal models of neurological diseases.


Subject(s)
Biological Assay/methods , Drug Evaluation, Preclinical/methods , Genistein/pharmacology , Ibuprofen/pharmacology , Receptors, Nerve Growth Factor/antagonists & inhibitors , Receptors, Nerve Growth Factor/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Dexamethasone/pharmacology , Enzyme Activation/drug effects , Humans , RNA, Small Interfering/metabolism , Time Factors , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Cancer Res ; 70(14): 5706-16, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20570886

ABSTRACT

Breast cancer that recurs as metastatic disease many years after primary tumor resection and adjuvant therapy seems to arise from tumor cells that disseminated early in the course of disease but did not develop into clinically apparent lesions. These long-term surviving, disseminated tumor cells maintain a state of dormancy, but may be triggered to proliferate through largely unknown factors. We now show that the induction of fibrosis, associated with deposition of type I collagen (Col-I) in the in vivo metastatic microenvironment, induces dormant D2.0R cells to form proliferative metastatic lesions through beta1-integrin signaling. In vitro studies using a three-dimensional culture system modeling dormancy showed that Col-I induces quiescent D2.0R cells to proliferate through beta1-integrin activation of SRC and focal adhesion kinase, leading to extracellular signal-regulated kinase (ERK)-dependent myosin light chain phosphorylation by myosin light chain kinase and actin stress fiber formation. Blocking beta1-integrin, Src, ERK, or myosin light chain kinase by short hairpin RNA or pharmacologic approaches inhibited Col-I-induced activation of this signaling cascade, cytoskeletal reorganization, and proliferation. These findings show that fibrosis with Col-I enrichment at the metastatic site may be a critical determinant of cytoskeletal reorganization in dormant tumor cells, leading to their transition from dormancy to metastatic growth. Thus, inhibiting Col-I production, its interaction with beta1-integrin, and downstream signaling of beta1-integrin may be important strategies for preventing or treating recurrent metastatic disease.


Subject(s)
Collagen Type I/metabolism , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Pulmonary Fibrosis/pathology , Actins/metabolism , Animals , Cell Growth Processes/physiology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Focal Adhesion Kinase 1/metabolism , Integrin beta1/metabolism , Lung Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Myosin-Light-Chain Kinase/metabolism , Phosphorylation , Pulmonary Fibrosis/metabolism , Rats , Signal Transduction , Transforming Growth Factor beta/metabolism , src-Family Kinases/metabolism
9.
Cancer Res ; 69(8): 3695-703, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19351854

ABSTRACT

Considered a chemopreventive agent, the ability of genistein to modulate the progression of existing prostate cancer (CaP) is not clear. We show here that the consumption of genistein (250 mg/kg diet) by 12-week-old transgenic adenocarcinoma mouse prostate (TRAMP-FVB) mice harboring prostatic intraepithelial neoplasia lesions until 20 weeks of age induces an aggressive progression of CaP, as evidenced by a 16% increase in the number of well-differentiated and poorly differentiated prostates, coinciding with a 70% incidence of pelvic lymph node (LN) metastases as opposed to 0% and 10% in 0 and 1,000 mg/kg groups, concomitant with elevated osteopontin (OPN) expression in prostates and LNs. Equivalent nanomolar (500 nmol/L) concentrations of genistein recapitulated these effects in human PC3 CaP cells as evidenced by increased proliferation, invasion, and matrix metalloproteinase-9 (MMP-9) activity (approximately 2-fold), accompanied by an up-regulation of OPN expression and secretion, compared with vehicle-treated cells. A pharmacologic dose (50 micromol/L) decreased proliferation, invasion, and MMP-9 activity (>2.0-fold) concomitant with OPN reduction. Upon OPN knockdown by short hairpin RNA, genistein was no longer effective in up-regulating PC3 cell proliferation, invasion, and MMP-9 activation, which were significantly reduced in the absence of OPN, highlighting the requirement for OPN in mediating the effects of genistein. Proliferation, invasion, and OPN levels were also nonsignificantly induced by genistein in the presence of ICI 182,780 or wortmannin, indicating a dependence on phosphatidylinositol 3-kinase and estrogen signaling. Our results suggest the presence of a biphasic regulation of CaP growth and metastasis by genistein, warranting careful examination of the effects of genistein on hormone-dependent cancers in a chemotherapeutic setting.


Subject(s)
Anticarcinogenic Agents/pharmacology , Genistein/pharmacology , Prostatic Neoplasms/pathology , Animals , Cell Growth Processes/drug effects , Cell Line, Tumor , Female , Humans , Lymphatic Metastasis , Male , Mice , Mice, Transgenic , Neoplasm Metastasis , Osteopontin/biosynthesis , Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms/metabolism , Up-Regulation/drug effects
10.
Biochem Biophys Res Commun ; 361(1): 169-75, 2007 Sep 14.
Article in English | MEDLINE | ID: mdl-17658479

ABSTRACT

Previous studies demonstrated a direct correlation with loss of kangai-1 (KAI1), a metastasis suppressor, and poor prognosis in human prostate and other cancers. In this study, we have characterized the age-dependent downregulation of KAI1 in the TRAMP model which was reversed when mice were fed a genistein-enriched diet. We demonstrated here that doses of genistein (5 and 10 microM)--achievable by supplement intake--significantly induced the expression of KAI1, both at the mRNA and protein levels (up to 2.5-fold), and decreased the invasiveness of TRAMP-C2 cells >2.0-fold. We have pinpointed KAI1 as the invasion suppressor, since its knockdown by siRNA restored the invasive potential of genistein-treated TRAMP-C2 cells to control levels. This work provides the first evidence that genistein treatment may counteract KAI1 downregulation, which is observed in many cancer types and therefore, could be used in anti-metastatic therapies.


Subject(s)
Anticarcinogenic Agents/pharmacology , Genistein/pharmacology , Kangai-1 Protein/biosynthesis , Prostatic Neoplasms/metabolism , Age Factors , Animals , Cell Line, Tumor , Disease Progression , Kangai-1 Protein/antagonists & inhibitors , Kangai-1 Protein/genetics , Male , Mice , Neoplasm Invasiveness , Prostatic Neoplasms/pathology , RNA Interference
11.
Carcinogenesis ; 28(11): 2282-90, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17615260

ABSTRACT

Telomerase contributes to the infinite replicative potential of cancer cells by conferring proliferation and survival through the regulation of growth factors and apoptotic proteins. Although it is generally known that the phytoestrogen, genistein, has telomerase-repressing and anti-proliferative effects on various cancer cells at pharmacological concentrations, we report here that physiologically achievable concentrations of genistein enhance telomerase activity, the proliferation of human prostate cancer cells and tumor growth in the transgenic adenocarcinoma mouse prostate model. In determining the mechanism for enhanced telomerase activity, we observed that physiological concentrations of genistein activated signal transducers and activators of transcription 3 (STAT3) both in vitro and in vivo and increased STAT3 binding to the telomerase reverse transcriptase promoter in human prostate cancer cells. These results demonstrate for the first time that physiologically achievable concentrations of genistein enhance telomerase reverse transcriptase transcriptional activity in prostate cancer cells via the activation of STAT3. Consequently, these concentrations of genistein will augment the growth of prostate cancer cells that could be detrimental to individuals with prostate cancer and therefore, caution should be exercised when genistein is considered for chemotherapeutic purposes.


Subject(s)
Genistein/pharmacology , Prostatic Neoplasms/enzymology , STAT3 Transcription Factor/metabolism , Telomerase/metabolism , Animals , Base Sequence , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Primers , Humans , Male , Mice , Mice, Transgenic , Phosphorylation , Promoter Regions, Genetic , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction , Telomerase/genetics
12.
Carcinogenesis ; 28(8): 1710-7, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17468512

ABSTRACT

Anti-proliferative properties of genistein in prostate and other cancers have been studied extensively. However, the identification of genistein targets that may mediate its chemopreventive effects in vivo requires further elucidation. In this study, we have demonstrated that the incorporation of genistein in the diet of transgenic adenocarcinoma mouse prostate model (TRAMP/FVB) mice resulted in a reduction in prostate size and the incidence of poorly differentiated (PD) cancer ensuing in an accumulation of prostates at the prostatic intra-epithelial neoplasia (PIN) stage. TRAMP/FVB prostate cancer progression and the onset of PD cancer were characterized by the activation of acutely transforming retrovirus AKT8 in rodent T cell lymphoma (Akt), phosphorylation of glycogen synthase kinase 3-beta (GSK-3beta), post-transcriptional up-regulation of cyclin D1 and repression of cadherin-1 via snail-1 up-regulation. Incorporation of genistein in the diet significantly inhibited the activation of Akt, restored the activation of GSK-3beta, reduced cyclin D1 levels post-transcriptionally and maintained the expression of the cadherin-1 complex via down-regulation of snail-1. By identifying the Akt-GSK-3 pathway and subsequently its downstream effectors, as targets for genistein chemopreventive action, we have elucidated one possible mechanism by which genistein decreases the proliferative potential, retards cancer progression and maintains the integrity of the prostatic epithelial cells in vivo.


Subject(s)
Adenocarcinoma/drug therapy , Cell Differentiation/genetics , Dietary Supplements , Genistein/administration & dosage , Glycogen Synthase Kinase 3/physiology , Prostatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/drug effects , Adenocarcinoma/enzymology , Animals , Antineoplastic Agents/administration & dosage , Cell Differentiation/drug effects , Disease Models, Animal , Disease Progression , Dose-Response Relationship, Drug , Drug Delivery Systems , Female , Male , Mice , Mice, Transgenic , Phenotype , Prostatic Neoplasms/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...