Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
BMC Cancer ; 22(1): 976, 2022 Sep 12.
Article in English | MEDLINE | ID: mdl-36096767

ABSTRACT

BACKGROUND: Fatty acid synthase (FASN) expression is associated with a more aggressive breast cancer phenotype and is regulated downstream of receptor tyrosine kinase (RTK) signaling pathways. Recently, post transcriptional regulation of lipogenic transcripts have been demonstrated as being mediated downstream of serine-arginine rich protein kinase 2 (SRPK2), which acts to phosphorylate serine-arginine rich splicing factors (SRSFs), resulting in RNA binding and various RNA regulatory processes. Though post-transcriptional regulation of FASN has been studied previously, the upstream mediators of these pathways have not been elucidated. METHODS: Western blotting and RT-qPCR were utilized to demonstrate alterations in FASN and mRNA expression upon modulation of the IGF-1-mTORC1-SRPK2 pathway by small molecule inhibitors or RNAi mediated silencing. RNA stability was accessed by using the transcriptional inhibitor actinomycin-D followed by RT-qPCR. Further, we employed RNA-immunoprecipitation to demonstrate the direct binding of SRSF-1 to FASN transcripts. RESULTS: In the current study, we demonstrated an IGF-1 induced increase in FASN mRNA and protein expression that was attenuated by mTORC1 inhibition. This mTORC1 inhibition also resulted in decreases in total and nuclear p-SRPK2 in response to IGF-1 exposure. Upon SRPK2 knockdown and inhibition, we observed a decrease in FASN protein and mRNA stability, respectively, in response to IGF-1 exposure that was specific to triple negative and HER2+ breast cancer cell lines. As we explored further, IGF-1 exposure resulted in an altered localization of eGFP expressed SRSF-1, pEGFP-SRSF-1 that was rescued upon both SRPK2 knockdown and mTORC1 inhibition. Further, we observed an increase binding of SRSF-1 to FASN RNA upon IGF-1 exposure, which was abrogated by SRPK2 knockdown. CONCLUSION: These current findings establish a potential IGF-1-mTORC1-SRPK2-FASN axis in breast cancer, which could be a potential therapeutic target for cancers that overexpress FASN and components of the IGF-1R pathway.


Subject(s)
Insulin-Like Growth Factor I , Neoplasms , Arginine , Cell Line, Tumor , Fatty Acid Synthases/metabolism , Insulin-Like Growth Factor I/genetics , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Protein Kinases , Protein Serine-Threonine Kinases , RNA , RNA, Messenger , Serine
2.
Nutr Cancer ; 74(2): 650-659, 2022.
Article in English | MEDLINE | ID: mdl-33715540

ABSTRACT

Obesity is associated with low-grade chronic inflammation, and metabolic dysregulation. Evidence shows that chronic inflammation inhibits protective immunity mediated by CD4+ T cells. Additionally, obesity-induced inflammation affects prostate cancer progression. However, the effect of obesity on CD4+ T-cell- response to prostate cancer is not well understood. To investigate whether obesity induces changes in CD4+ T cell cytokine profile, cytokine expression was measured in splenic CD4+ T-cells from 10-week-old male C57Bl/6 mice exposed to conditioned media (CM) from macrophages grown in sera from obese subjects. Additionally, expression levels of key regulators of Epithelial-Mesenchymal Transition (EMT) were measure in prostate cancer epithelial cells exposed to conditioned media from obesity-modified T-cells. Cell migration and invasion was measured in prostate cancer epithelial cells exposed to CM from obesity-modified CD4+ T-cells. Obesity suppressed the expression of IFNγ and IL-2 in CD4+ T-cells but up-regulated the expression of IL-6. Prostate epithelial cancer cells exposed to conditioned media from obesity-modified T cell increased the expression of EMT markers and showed a higher invasive and migratory capacity.


Subject(s)
Prostate , Prostatic Neoplasms , Animals , CD4-Positive T-Lymphocytes , Cell Line, Tumor , Cell Movement , Epithelial-Mesenchymal Transition/genetics , Humans , Male , Mice , Obesity/complications , Phenotype , Prostate/metabolism , Prostatic Neoplasms/metabolism
3.
Breast Cancer Res Treat ; 187(2): 375-386, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33893909

ABSTRACT

PURPOSE: The majority of breast cancers are estrogen receptor (ERα) positive making endocrine therapy a mainstay for these patients. Unfortunately, resistance to endocrine therapy is a common occurrence. Fatty acid synthase (FASN) is a key enzyme in lipid biosynthesis and its expression is commensurate with tumor grade and resistance to numerous therapies. METHODS: The effect of the FASN inhibitor TVB-3166 on ERα expression and cell growth was characterized in tamoxifen-resistant cell lines, xenografts, and patient explants. Subcellular localization of ERα was assessed using subcellular fractionations. Palmitoylation and ubiquitination of ERα were assessed by immunoprecipitation. ERα and p-eIF2α protein levels were analyzed by Western blotting after treatment with TVB-3166 with or without the addition of palmitate or BAPTA. RESULTS: TVB-3166 treatment leads to a marked inhibition of proliferation in tamoxifen-resistant cells compared to the parental cells. Additionally, TVB-3166 significantly inhibited tamoxifen-resistant breast tumor growth in mice and decreased proliferation of primary tumor explants compared to untreated controls. FASN inhibition significantly reduced ERα levels most prominently in endocrine-resistant cells and altered its subcellular localization. Furthermore, we showed that the reduction of ERα expression upon TVB-3166 treatment is mediated through the induction of endoplasmic reticulum stress. CONCLUSION: Our preclinical data provide evidence that FASN inhibition by TVB-3166 presents a promising therapeutic strategy for the treatment of endocrine-resistant breast cancer. Further clinical development of FASN inhibitors for endocrine-resistant breast cancer should be considered.


Subject(s)
Breast Neoplasms , Enzyme Inhibitors/therapeutic use , Fatty Acid Synthase, Type I/antagonists & inhibitors , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm , Estrogen Receptor alpha/genetics , Fatty Acid Synthase, Type I/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Tamoxifen/pharmacology
4.
Nutr Cancer ; 68(6): 1021-33, 2016.
Article in English | MEDLINE | ID: mdl-27367296

ABSTRACT

Epidemiological studies have correlated frequent omega-3 (n-3) fatty acid consumption with a lower risk for breast cancer; however, recent prospective studies have been less conclusive. Efforts in the preventive setting have focused on the use of n-3 fatty acids, and the pharmaceutical ethyl esters (EE) of these natural compounds, for high-risk patient populations. Limited understanding of specific mechanisms by which these agents function has hampered identification of the cancer subtype(s) that would gain the greatest therapeutic benefit. In this study, we investigated the in vitro effects of n-3 EEs in four distinct breast cancer subtypes and explored how they affect not only breast cancer cell survival but also modulate the nuclear factor kappa-light-chain enhancer of activated B cells (NF-κB) and peroxisome proliferator-activated receptor gamma signaling pathways. Similar to the high variance in response observed in human studies, we found that the effectiveness of n-3 EEs depends on the molecular characteristics of the MCF-7, CAMA-1, MDA-MB-231, and SKBR3 breast cancer cell lines and is closely associated with the suppression of NF-κB. These data strongly suggest that the use of n-3 fatty acids and their pharmaceutical ether esters in the prevention and therapeutic setting should be guided by specific tumor characteristics.


Subject(s)
Anticarcinogenic Agents/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Docosahexaenoic Acids/metabolism , Eicosapentaenoic Acid/metabolism , Fatty Acids, Omega-3/metabolism , Neoplasm Proteins/genetics , Polymorphism, Single Nucleotide , Anticarcinogenic Agents/therapeutic use , Breast Neoplasms/pathology , Breast Neoplasms/prevention & control , Cell Line, Tumor , Cell Survival , Colony-Forming Units Assay , Dietary Supplements , Docosahexaenoic Acids/therapeutic use , Drug Combinations , Eicosapentaenoic Acid/therapeutic use , Esters/metabolism , Esters/therapeutic use , Fatty Acids, Omega-3/therapeutic use , Female , Gene Expression Regulation, Neoplastic , Genes, Reporter , Humans , Kinetics , Mutation , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , PPAR gamma/antagonists & inhibitors , PPAR gamma/genetics , PPAR gamma/metabolism , Signal Transduction
5.
Biochim Biophys Acta ; 1842(10): 1475-82, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25066474

ABSTRACT

Glycerol-3-phosphate acyltransferase-1 is the first rate limiting step in de novo glycerophospholipid synthesis. We have previously demonstrated that GPAT-1 deletion can significantly alter T cell function resulting in a T cell phenotype similar to that seen in aging. Recent studies have suggested that changes in the metabolic profile of T cells are responsible for defining specific effector functions and T cell subsets. Therefore, we determined whether T cell dysfunction in GPAT-1 (-/-) CD4(+) T cells could be explained by changes in cellular metabolism. We show here for the first time that GPAT-1 (-/-) CD4(+) T cells exhibit several key metabolic defects. Striking decreases in both the oxygen consumption rate (OCR) and the extracellular acidification rate (ECAR) were observed in GPAT-1 (-/-) CD4(+) T cells following CD3/CD28 stimulation indicating an inherent cellular defect in energy production. In addition, the spare respiratory capacity (SRC) of GPAT-1 (-/-) CD4+ T cells, a key indicator of their ability to cope with mitochondrial stress was significantly decreased. We also observed a significant reduction in mitochondrial membrane potential in GPAT-1 (-/-) CD4(+) T cells compared to their WT counterparts, indicating that GPAT-1 deficiency results in altered or dysfunctional mitochondria. These data demonstrate that deletion of GPAT-1 can dramatically alter total cellular metabolism under conditions of increased energy demand. Furthermore, altered metabolic response following stimulation may be the defining mechanism underlying T cell dysfunction in GPAT-1 (-/-) CD4(+) T cells. Taken together, these results indicate that GPAT-1 is essential for the response to the increased metabolic demands associated with T cell activation.

6.
Prostate ; 75(5): 449-62, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25560177

ABSTRACT

BACKGROUND: A close relationship between aging, inflammation, and prostate cancer is widely accepted. Aging is accompanied by a progressive increase in pro-inflammatory cytokines, including interleukin 17 (IL-17), a key pro-inflammatory cytokine that becomes dysregulated with age. However, the contribution of IL-17 to age-related prostate tumorigenesis remains unclear. The aim of this study was to investigate the role of age-related IL-17 dysregulation in prostate tumorigenesis. METHODS: Serum and splenic T-lymphocytes from young GPAT-1 knock-out aging-mimic T cell mice as well as young and aged wild-type mice were collected. shRNA was used to knock down the IL-17 receptor in LNCaP prostate cancer cells and RWPE-1 non-transformed prostate epithelial cells, which were then exposed to the mouse sera or conditioned media from stimulated T-lymphocytes. NF-κB activation, NF-κB target gene expression, and cell proliferation were all measured in these cells by luciferase assay, qPCR, Western blot analysis, and MTT assay, respectively. RESULTS: T-lymphocyte-secreted IL-17 from aging-mimic mice induced NF-κB activity and target gene expression in LNCaP and RWPE-1 cells. It also promoted proliferation of these cells. CONCLUSION: Aging-mimic T cell mice produce increased levels of IL-17, which stimulates the pro-inflammatory NF-κB pathway in prostate epithelial cells. NF-κB increases inflammation, carcinogenesis and metastatic potential in the prostate. These findings provide evidence that the dysregulation of cytokine production seen in aged T cells may directly contribute to the increased risk for prostate cancer in the elderly.


Subject(s)
Aging/physiology , Interleukin-17/metabolism , Prostatic Neoplasms/metabolism , Signal Transduction/physiology , Animals , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Epithelial Cells/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , Receptors, Interleukin-17/genetics , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology
7.
Breast Cancer Res Treat ; 149(1): 49-57, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25476497

ABSTRACT

Obesity is associated with a worse breast cancer prognosis, particularly in estrogen receptor alpha (ERα) positive, postmenopausal patients. We hypothesized that this is mediated in part by an elevation in breast cancer cell cyclooxygenase-2 (COX-2) expression and prostaglandin E2 (PGE2) production that results in greater local pre-adipocyte aromatase expression. We utilized an in vitro model of the obese patient's tumor microenvironment in which cultured MCF-7 breast cancer cells and pre-adipocytes were exposed to pooled serum from obese (OB; BMI ≥ 30.0 kg/m(2)) or normal weight (N; BMI 18.5-24.9 kg/m(2)) postmenopausal women. Exposure to OB versus N sera significantly increased MCF-7 cell COX-2 expression and PGE2 production. Pre-adipocyte aromatase expression was 89 % greater following culture in conditioned media (CM) from MCF-7 cells exposed to OB versus N sera (OB-CM and N-CM, respectively), a difference nullified by MCF-7 cell treatment with the COX-2 inhibitor celecoxib. Previous analysis of the sera revealed significantly higher interleukin-6 (IL-6) concentrations in the OB versus N samples. Depletion of IL-6 from the sera neutralized the difference in pre-adipocyte aromatase expression stimulated by OB-CM versus N-CM. Finally, CM from pre-adipocyte/MCF-7 cell co-cultures exposed to OB sera stimulated greater MCF-7 and T47D breast cancer cell ERα activity and proliferation in comparison to N sera. This study indicates that obesity-associated systemic IL-6 indirectly enhances pre-adipocyte aromatase expression via increased breast cancer cell PGE2 production. Investigation regarding the efficacy of a COX-2 inhibitor/aromatase inhibitor combination therapy in the obese postmenopausal patient population is warranted.


Subject(s)
Aromatase/biosynthesis , Breast Neoplasms/genetics , Dinoprostone/biosynthesis , Interleukin-6/genetics , Obesity/genetics , Adipocytes/enzymology , Aromatase Inhibitors/administration & dosage , Breast Neoplasms/complications , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2 Inhibitors/administration & dosage , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-6/antagonists & inhibitors , Interleukin-6/immunology , MCF-7 Cells , Obesity/complications , Obesity/pathology
8.
Nutr Cancer ; 66(7): 1179-86, 2014.
Article in English | MEDLINE | ID: mdl-25264717

ABSTRACT

Obesity is the leading preventable comorbidity associated with increased prostate cancer-related recurrence and mortality. Epidemiological and clinical studies indicate that a body mass index >30 is associated with increased oxidative DNA damage within the prostate gland and increased prostate cancer-related mortality. Here we provide evidence that obesity promotes worse clinical outcome through induction of metabolic abnormalities known to promote genotoxic stress. We have previously reported that blood serum derived from obese mice may enhance the proliferative and invasive potential of human prostate cancer cell lines ex vivo. Here we show that a 1-h exposure of LNCaP or PacMetUT1 prostate cancer cell lines and nonmalignant RWPE-1 prostate epithelial cells to 2% serum from obese mice induces markers of aerobic glycolysis relative to those exposed to serum from nonobese mice. This metabolic change was correlated with accumulation of reactive oxygen species (ROS) and increased frequency of DNA double-strand breaks. Interestingly, N-tert-Butylhydroxylamine, an antioxidant, significantly suppressed markers of aerobic glycolysis in the cells exposed to the blood serum of obese mice, suggesting that ROS contributes to a metabolic shift toward aerobic glycolysis. Here we describe obesity-induced changes in key metabolic markers that impact prostate cancer cell progression and explore the role of antioxidants in ameliorating these effects.


Subject(s)
Glycolysis , Obesity/physiopathology , Prostatic Neoplasms/physiopathology , Animals , Antioxidants/pharmacology , Cell Line, Tumor , DNA Damage/drug effects , Disease Progression , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Hydroxylamines/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Oxidative Stress/drug effects , Pyruvate Kinase/metabolism , Reactive Oxygen Species/metabolism , Reproducibility of Results
9.
Oncogene ; 43(14): 1063-1074, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38374406

ABSTRACT

Flotillin-1 contributes to invasion and metastasis in triple negative breast cancer (TNBC) and is modified post-translationally through palmitoylation. Palmitoylation, the process of conjugating palmitoyl-CoA to proteins, plays an essential role in protein stability and trafficking. Thus far, there has not been any investigation into the role of flotillin-1 palmitoylation in the context of metastasis in vivo. To address the role of flotillin-1 palmitoylation in metastasis, MDA-MB-231 cells expressing palmitoylation defective flotillin-1 constructs were used as models. Compared to flotillin-1 WT expressing tumors, flotillin-1 palmitoylation defective displayed abrogated tumor progression and lung metastasis in vivo in both spontaneous and experimental models. Further mechanistic investigation led to the identification of zDHHC5 as the main palmitoyl acyltransferase responsible for palmitoylating endogenous flotillin-1. Modulation of flotillin-1 palmitoylation status through mutagenesis, zDHHC5 silencing, and 2-bromopalmitate inhibition all resulted in the proteasomal degradation of flotillin-1 protein. To assess if flotillin-1 palmitoylation can be inhibited for potential clinical relevance, we designed a competitive peptide fused to a cell penetrating peptide sequence, which displayed efficacy in blocking flotillin-1 palmitoylation in vitro without altering palmitoylation of other zDHHC5 substrates, highlighting its specificity. Additionally, TNBC xenograft tumor models expressing a doxycycline inducible flotillin-1 palmitoylation inhibiting peptide displayed attenuated tumor growth and lung metastasis. Collectively, these results reveal a novel palmitoylation dependent mechanism which is essential for the stability of flotillin-1 protein. More specifically, disruption of flotillin-1 palmitoylation through mutagenesis or competitive peptide promoted flotillin-1 protein degradation, subsequently impeding its tumor promoting and metastasis-inducing effects in TNBC tumor models.


Subject(s)
Lung Neoplasms , Membrane Proteins , Triple Negative Breast Neoplasms , Humans , Cell Line, Tumor , Lipoylation , Lung Neoplasms/genetics , Membrane Proteins/metabolism , Peptides , Triple Negative Breast Neoplasms/genetics
10.
Breast Cancer Res ; 15(4): R59, 2013.
Article in English | MEDLINE | ID: mdl-23880059

ABSTRACT

INTRODUCTION: Epidemiological and clinical studies indicate that obesity is associated with a worse postmenopausal breast cancer prognosis and an increased risk of endocrine therapy resistance. However, the mechanisms mediating these effects remain poorly understood. Here we investigate the molecular pathways by which obesity-associated circulating factors in the blood enhance estrogen receptor alpha (ERα) positive breast cancer cell viability and growth. METHODS: Blood serum was collected from postmenopausal breast cancer patients and pooled by body mass index (BMI) category (Control: 18.5 to 24.9 kg/m²; Obese: ≥30.0 kg/m²). The effects of patient sera on MCF-7 and T47D breast cancer cell viability and growth were examined by MTT and colony formation assays, respectively. Insulin-like growth factor receptor 1(IGF-1R), Akt, and ERK1/2 activation and genomic ERα activity were assessed to determine their possible contribution to obese patient sera-induced cell viability and growth. To further define the relative contribution of these signaling pathways, cells grown in patient sera were treated with various combinations of ERα, PI3K/Akt and MAPK targeted therapies. Comparisons between cells exposed to different experimental conditions were made using one-way analysis of variance (ANOVA) and Student's t test. RESULTS: Cells grown in media supplemented with obese patient sera displayed greater cell viability and growth as well as IGF-1R, Akt and ERK1/2 activation relative to control sera. Despite the lack of a significant difference in genomic ERα activity following growth in obese versus control patient sera, we observed a dramatic reduction in cell viability and growth after concurrent inhibition of the ERα and PI3K/Akt signaling pathways. Further, we demonstrated that ERα inhibition was sufficient to attenuate obese serum-induced Akt and ERK1/2 activation. Together, these data suggest that obesity promotes greater ERα positive breast cancer cell viability and growth through enhanced crosstalk between nongenomic ERα signaling and the PI3K/Akt and MAPK pathways. CONCLUSIONS: Circulating factors in the serum of obese postmenopausal women stimulate ERα positive breast cancer cell viability and growth by facilitating non-genomic ERα crosstalk with the PI3K/Akt and MAPK signaling pathways. These findings provide valuable insight into one mechanism by which obesity may promote ERα positive postmenopausal breast cancer progression and endocrine therapy resistance.


Subject(s)
Breast Neoplasms/complications , Breast Neoplasms/metabolism , MAP Kinase Signaling System , Obesity/complications , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor Cross-Talk , Receptors, Estrogen/metabolism , Biomarkers/blood , Biomarkers/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Progression , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/metabolism , Female , Humans , Middle Aged , Obesity/blood , Obesity/metabolism , Phosphoinositide-3 Kinase Inhibitors , Receptor, IGF Type 1/metabolism , Receptors, Estrogen/antagonists & inhibitors , Risk Factors
11.
Prostate ; 73(8): 855-64, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23532664

ABSTRACT

BACKGROUND: The primary risk for prostate cancer is aging, often associated with inflammation. Evidence implicates progressive age-related immune dysfunction with increased prostate cancer incidence and mortality. The aged T-cell response is characterized by increased production of pro-inflammatory cytokines, which could significantly contribute to prostate tumorigenesis through induction of key inflammation-mediated pro-survival factors. METHODS: T-cell function of the young (<6 month-old) glycerol-3-phosphate acyltransferase-1 (GPAT-1) knock-out mouse mimics many of the hallmarks observed in an aged (>22-month-old) mouse. Serum and splenic T-lymphocytes from young GPAT-1(-/-) (6 months) and aged wild type (22 months) mice were collected for in vitro studies, including a cytokine immunoarray for serum cytokine levels, luciferase assays for NF-κB activation and Western blot analyses for protein expression. RESULTS: The T-cell cytokine profile of the GPAT-1(-/-) mice mirrored that observed in aged wild type mice, including higher expression levels of IL-17. Serum- and T-cell-derived factors induced NF-κB activity in normal, non-transformed and prostate cancer epithelial cells, correlating with re-localization of NF-κB and increased protein expression of downstream targets of NF-κB. CONCLUSION: The aging and aging-mimic mice produced circulating factors that induce pro-inflammatory pathways in prostate cells, most notably NF-κB. These findings provide evidence that an aged T-cell may directly contribute to the increased risk for prostate cancer in the elderly and establish that the GPAT-1(-/-) model, which mimics many of the characteristics of an aged immune system, is a viable tool for investigating this novel area of cancer risk.


Subject(s)
Aging/immunology , Cell Transformation, Neoplastic/immunology , Glycerol-3-Phosphate O-Acyltransferase/immunology , Inflammation/immunology , Prostatic Neoplasms/immunology , T-Lymphocytes/immunology , Animals , Blotting, Western , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Cytokines/blood , Cytokines/immunology , Disease Models, Animal , Female , Immunoassay , Inflammation/enzymology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/immunology , Prostatic Neoplasms/enzymology , T-Lymphocytes/enzymology
12.
Mol Carcinog ; 52(6): 446-58, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22290600

ABSTRACT

The prevalence of obesity, an established risk and progression factor for postmenopausal breast cancer, remains high in US women. Activation of Akt/mammalian target of rapamycin (mTOR) signaling plays a key role in the obesity-breast cancer link. However, the impact of weight normalization in obese postmenopausal women on breast tumorigenesis and/or Akt/mTOR activation is poorly characterized. To model this, ovariectomized female C57BL/6 mice were fed a control diet (n = 20), a calorie restriction (CR) regimen (n = 20), or a diet-induced obesity (DIO) diet (n = 30). At week 17, DIO mice were switched to control diet, resulting in formerly obese (FOb) mice with weights identical to the controls by week 20. MMTV-Wnt-1 mammary tumor cells were injected at 20 wk into each mouse. Two weeks post-injection, vehicle or the mTOR inhibitor RAD001 at 10 or 15 mg/kg body weight (n = 10/diet group) was administered by gavage twice/week until termination. Relative to controls, CR mice had decreased (and DIO mice had increased) serum insulin-like growth factor-1 (IGF-1) and phosphorylation of Akt/mTOR pathway components. RAD001 decreased tumor growth in the CR, control, and FOb mice. Wnt-1 tumor cells treated in vitro with serum from mice from each group established that diet-dependent circulating factors contribute to tumor growth and invasiveness. These findings suggest weight normalization in obese mice does not immediately reverse tumor progression or Akt/mTOR activation. Treatment with RAD001 blocked mammary tumor development and mTOR activation observed in the FOb mice, suggesting combination of lifestyle and pharmacologic strategies may be effective for breaking the obesity-breast cancer link.


Subject(s)
Antineoplastic Agents/therapeutic use , Mammary Neoplasms, Experimental/complications , Mammary Neoplasms, Experimental/drug therapy , Obesity/complications , Proto-Oncogene Proteins c-akt/metabolism , Sirolimus/analogs & derivatives , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Line, Tumor , Diet , Everolimus , Female , Hormones/blood , Insulin-Like Growth Factor I/metabolism , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Obese , Signal Transduction/drug effects , Sirolimus/therapeutic use , Weight Loss/drug effects , Wnt1 Protein/metabolism
13.
Nutr Cancer ; 65(4): 556-62, 2013.
Article in English | MEDLINE | ID: mdl-23659447

ABSTRACT

Prostate cancer (PCa) is the second leading cause of cancer-related deaths in men. Studies show that consumption of polyunsaturated fatty acids (PUFA) modulates the development and progression of prostate cancer. High amounts of omega-6 fatty acids have been linked with increased prostate cancer risk, whereas omega-3 fatty acids have been shown to inhibit PCa growth. However, because omega-3 and omega-6 are both essential fatty acids and part of a complete diet, it is more relevant to determine the ideal ratio of the two that would allow patients to benefit from the therapeutic properties of omega-3 fatty acids. LNCaP prostate cancer cells were treated with dietary-based ratios of omega-6 to omega-3 fatty acids under hormone-deprivation conditions, and effects on various cellular processes were determined. A low omega-6 to omega-3 PUFA ratio can delay the progression of cells toward castration-resistance by suppressing pathways involved in prostate cancer progression, such as the Akt/mTOR/NFκB axis. It also suppresses the expression of cyclin D1, and activation of caspase-3 and annexin V staining shows induction of proapoptotic events. Taken together, our data demonstrates that maintaining a low omega-6 to omega-3 fatty acids ratio can enhance efficacy of hormone ablation therapy.


Subject(s)
Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-6/pharmacology , Prostatic Neoplasms/diet therapy , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin D1/metabolism , Humans , Male , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/prevention & control , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
14.
Breast Cancer (Auckl) ; 16: 11782234221111374, 2022.
Article in English | MEDLINE | ID: mdl-36035625

ABSTRACT

Purpose: The objective of this study is to determine the impact of exposure to obesity-related systemic factors on fatty acid synthase enzyme (FASN) expression in breast cancer cells. Methods: MCF-7 breast cancer cells were exposed to sera from patients having obesity or not having obesity and subjected to quantitative reverse transcription polymerase chain reaction (RT-qPCR). Subsequent MTT and colony-forming assays using both MCF-7 and T-47D cells exposed to sera and treated with or without FASN inhibitor, TVB-3166, were used. MCF-7 cells were then treated with insulin and the sterol regulatory element-binding protein (SREBP) processing inhibitor, betulin, prior to analysis of FASN expression by quantitative RT-qPCR and western blot. Insulin-induced SREBP-FASN promoter binding was analyzed by chromatin immunoprecipitation with an anti-SREBP antibody. Results: In response to sera exposure (body mass index [BMI] >30) there was an increase in FASN expression in breast cancer cells. Furthermore, treatment with the FASN inhibitor, TVB-3166, resulted in a decreased breast cancer cell survival and proliferation while increasing apoptosis upon sera exposure (BMI >30). Insulin-exposed MCF-7 cells exhibited an increased FASN messenger RNA and protein expression, which is abrogated upon SREBP inhibition. In addition, insulin exposure induced enhanced SREBP binding to the FASN promoter. Conclusions: Our results implicate FASN as a potential mediator of obesity-induced breast cancer aggression and a therapeutic target of patients with obesity-induced breast cancer.

15.
Breast Cancer Res ; 13(6): R120, 2011.
Article in English | MEDLINE | ID: mdl-22115051

ABSTRACT

INTRODUCTION: Adjuvant treatment with tamoxifen substantially improves survival of women with estrogen-receptor positive (ER+) tumors. Tamoxifen resistance (TAMR) limits clinical benefit. RRR-α-tocopherol ether-linked acetic acid analogue (α-TEA) is a small bioactive lipid with potent anticancer activity. We evaluated the ability of α-TEA in the presence of tamoxifen to circumvent TAMR in human breast cancer cell lines. METHODS: Two genotypically matched sets of TAM-sensitive (TAMS) and TAM-resistant (TAMR) human breast cancer cell lines were assessed for signal-transduction events with Western blotting, apoptosis induction with Annexin V-FITC/PI assays, and characterization of cholesterol-rich microdomains with fluorescence staining. Critical involvement of selected mediators was determined by using RNA interference and chemical inhibitors. RESULTS: Growth-factor receptors (total and phosphorylated forms of HER-1 and HER-2), their downstream prosurvival mediators pAkt, pmTOR, and pERK1/2, phosphorylated form of estrogen receptor-α (pER-α at Ser-167 and Ser-118, and cholesterol-rich lipid microdomains were highly amplified in TAMR cell lines and enhanced by treatment with TAM. α-TEA disrupted cholesterol-rich microdomains, acted cooperatively with TAM to reduce prosurvival mediators, and induced DR5-mediated mitochondria-dependent apoptosis via an endoplasmic reticulum stress-triggered pro-death pJNK/CHOP/DR5 amplification loop. Furthermore, methyl-ß-cyclodextrin (MßCD), a chemical disruptor of cholesterol rich microdomains, acted cooperatively with TAM to reduce prosurvival mediators and to induce apoptosis. CONCLUSIONS: Data for the first time document that targeting cholesterol-rich lipid microdomains is a potential strategy to circumvent TAMR, and the combination of α-TEA + TAM can circumvent TAMR by suppression of prosurvival signaling via disruption of cholesterol-rich lipid microdomains and activation of apoptotic pathways via induction of endoplasmic reticulum stress.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/metabolism , Cholesterol/metabolism , Membrane Microdomains/drug effects , Tamoxifen/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , Antioxidants/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm , Female , Humans , Membrane Microdomains/metabolism , Signal Transduction/drug effects , Tamoxifen/therapeutic use , alpha-Tocopherol/pharmacology , beta-Cyclodextrins/pharmacology
16.
Prostate ; 71(13): 1420-8, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21360560

ABSTRACT

BACKGROUND: Oxidative burden is strongly implicated in the pathogenesis of age-related diseases, including prostate cancer tumor formation. As omega-3 fatty acids possess known antioxidant properties, we investigated the effects of docosahexaenoic acid (DHA-22:6n-3), one component of fish oil, in modulating the effects of oxidative DNA damage in LNCaP and PacMetUT1 human prostate adenocarcinoma cells and in a normal human prostate cell line, PrEC. METHODS: Cell survival was determined by an inhibition of colony formation assay. DNA double-strand breaks, NF-κB subcellular localization and relative survivin expression levels were determined by immunofluorescence and survivin expression levels confirmed by immunoblot assay. Measurement of NF-κB transcriptional activity was investigated by dual luciferase assay. RESULTS: LNCaP and PacMetUT1 cells pretreated with DHA and pulsed with 32 µM H(2) O(2) exhibit decreased survival compared to PrEC. γ-H2AX foci, indicating DNA double-strand breaks, were associated with translocation of NF-κB into the nucleus, whereas exposure to DHA prior to H(2) O(2) treatment prevented NF-κB translocation. Further, DHA attenuated H(2) O(2) -induced NF-κB transcriptional activity and diminished expression of the downstream target, survivin. CONCLUSIONS: NF-κB is heavily implicated in promoting prosurvival signaling and may be critical for resistance to the chronic oxidative stress observed in the pathogenesis of prostate cancer. Our studies indicate that exposure of cells to physiologically achievable levels of DHA prior to treatment with H(2) O(2) results in decreased cancer cell survival which is associated with nuclear exclusion of NF-κB. We therefore propose that DHA selectively sensitizes prostate cancer cells to growth arrest through attenuation of the NF-κB survival pathway.


Subject(s)
Docosahexaenoic Acids/pharmacology , NF-kappa B/antagonists & inhibitors , Oxidative Stress , Prostatic Neoplasms/drug therapy , Apoptosis , Cell Line, Tumor , Cell Nucleus/metabolism , Humans , Hydrogen Peroxide/pharmacology , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Survivin
17.
Prostate ; 71(13): 1441-54, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21321980

ABSTRACT

BACKGROUND: The skeleton is the most common site of prostate cancer metastasis, which often results in osteoblastic lesions. The role of transforming growth factor-beta (TGFß) signaling in prostate cancer-induced osteoblastic metastasis is not clear. We investigated the role of TGFß signaling in prostate cancer-induced bone metastasis using a novel human prostate cancer cell line, PacMetUT1. METHODS: We injected PacMetUT1/Luc-GFP cells in male nude mice by intracardiac and intratibia injections and then investigated the effect of TGFß signaling abrogation on osteoblastic tumor growth and incidence in vivo by using fluorescence and bioluminescence imaging analysis and quantifying bone and tumor volume by histomorphometry analysis. Osteoclasts were counted using TRAP assay. RESULTS: Osteoblastic bone metastasis in skull, rib, and femur was detected after 10-16 weeks of intracardiac injection of the PacMetUT1 cells. Stable knockdown of TGFß1 with an shRNA resulted in decreased tumor incidence and bone formation when the cells were directly injected into the tibiae. Systemic administration of either a small inhibitor of TGFß type I receptor kinase or a pan TGFß binding protein (BG(E) RII) also decreased bone tumor growth and osteoblastic bone formation in vivo after 7 weeks of treatment. CONCLUSIONS: Our results for the first time indicate that blockade of TGFß signaling in the PacMetUT1 model significantly inhibits osteoblastic bone formation and tumor incidence. Thus, TGFß signaling pathway may be a viable target for the prevention and treatment of prostate cancer-induced bone metastasis.


Subject(s)
Bone Neoplasms/secondary , Osteoblasts/pathology , Prostatic Neoplasms/pathology , Signal Transduction/physiology , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Base Sequence , Bone Neoplasms/prevention & control , Cell Line, Tumor , Cell Proliferation , Humans , Lung Neoplasms/secondary , Male , Mice , Middle Aged , Molecular Sequence Data , Smad2 Protein/metabolism , Transforming Growth Factor beta/physiology
18.
Nutr Cancer ; 63(5): 771-7, 2011.
Article in English | MEDLINE | ID: mdl-21667400

ABSTRACT

Currently, progression of prostate cancer to androgen independence remains the primary obstacle to improved survival. In order to improve overall survival, novel treatment strategies that are based upon specific molecular mechanisms that prolong the androgen-dependent state and that are useful for androgen-independent disease need to be identified. Both epidemiological as well as preclinical data suggest that omega-3 fatty acids are effective primary tumor prevention agents; however, their efficacy at preventing and treating refractory prostate cancer has not been as thoroughly investigated. We used an in vitro model of androgen ablation to determine the effect of treatment with omega-3 fatty acids on the progression to an androgen-independent state. The omega-3 fatty acids docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) were able to prevent progression of LNCaP cells while the omega-6 fatty acid arachidonic acid (AA) actually promoted cell growth under conditions of hormone depletion. These results correlated with a decrease in the expression of the androgen receptor as well as suppression of the Akt/mTOR signaling pathway. Connecting the mechanisms by which omega-3 fatty acids affect phenotypic outcome is important for effective exploitation of these nutrient agents as a therapeutic approach. Understanding these processes is critical for the development of effective dietary intervention strategies that improve overall survival.


Subject(s)
Androgens/metabolism , Fatty Acids, Omega-3/metabolism , Neoplasm Proteins/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Cell Line, Tumor , Cell Proliferation , Clone Cells , Docosahexaenoic Acids/metabolism , Eicosapentaenoic Acid/metabolism , Humans , Male , Phosphorylation , Prostatic Neoplasms/diet therapy , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism
19.
Mol Cancer Res ; 16(5): 869-879, 2018 05.
Article in English | MEDLINE | ID: mdl-29453319

ABSTRACT

Obesity is associated with poor prognosis in triple-negative breast cancer (TNBC). Preclinical models of TNBC were used to test the hypothesis that increased leptin signaling drives obesity-associated TNBC development by promoting cancer stem cell (CSC) enrichment and/or epithelial-to-mesenchymal transition (EMT). MMTV-Wnt-1 transgenic mice, which develop spontaneous basal-like, triple-negative mammary tumors, received either a control diet (10% kcal from fat) or a diet-induced obesity regimen (DIO, 60% kcal from fat) for up to 42 weeks (n = 15/group). Mice were monitored for tumor development and euthanized when tumor diameter reached 1.5 cm. Tumoral gene expression was assessed via RNA sequencing (RNA-seq). DIO mice had greater body weight and percent body fat at termination than controls. DIO mice, versus controls, demonstrated reduced survival, increased systemic metabolic and inflammatory perturbations, upregulated tumoral CSC/EMT gene signature, elevated tumoral aldehyde dehydrogenase activity (a CSC marker), and greater leptin signaling. In cell culture experiments using TNBC cells (murine: E-Wnt and M-Wnt; human: MDA-MB-231), leptin enhanced mammosphere formation, and media supplemented with serum from DIO versus control mice increased cell viability, migration, invasion, and CSC- and EMT-related gene expression, including Foxc2, Twist2, Vim, Akt3, and Sox2 In E-Wnt cells, knockdown of leptin receptor ablated these procancer effects induced by DIO mouse serum. These findings indicate that increased leptin signaling is causally linked to obesity-associated TNBC development by promoting CSC enrichment and EMT.Implications: Leptin-associated signals impacting CSC and EMT may provide new targets and intervention strategies for decreasing TNBC burden in obese women. Mol Cancer Res; 16(5); 869-79. ©2018 AACR.


Subject(s)
Leptin/metabolism , Neoplastic Stem Cells/metabolism , Obesity/metabolism , Triple Negative Breast Neoplasms/genetics , Animals , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Female , Humans , Mice , Neoplastic Stem Cells/pathology , Signal Transduction , Triple Negative Breast Neoplasms/pathology
20.
Med Hypotheses ; 117: 63-68, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30077200

ABSTRACT

Prostate cancer incidence increases with age; along with many other cancers, it could be considered a disease of aging. Prostate cancer screening has led to a significant proportion of men diagnosed with low-grade, low-stage prostate cancer who are now more likely to choose an active surveillance strategy rather than definitive treatments. Definitive treatment, such as surgery and radiation therapy, is useful for high-grade disease; however, because of the low long-term risk of progression of a low-grade disease and side effects of surgery and radiation, these treatments are less commonly used for low-grade disease. While five alpha reductase inhibitors have been shown to reduce the risk of cancer detection on subsequent biopsies for men on active surveillance, no medications have been proven to prevent progression to high-grade disease. mTOR pathways have long been known to influence prostate cancer and are targets in various prostate cancer patient populations. Low-dose mTOR inhibition with rapamycin has shown promise in pre-clinical models of prostate cancer and appear to affect cellular senescence and immunomodulation in the aging population. We hypothesize that low-dose mTOR inhibition could reduce progression of low-grade prostate cancer patients, allowing them to remain on active surveillance.


Subject(s)
5-alpha Reductase Inhibitors/pharmacology , Prostatic Neoplasms/drug therapy , TOR Serine-Threonine Kinases/antagonists & inhibitors , Aged , Animals , Cell Line, Tumor , Cellular Senescence , Disease Progression , Early Detection of Cancer , Glucose Intolerance , Humans , Magnetic Resonance Imaging , Male , Mice , Middle Aged , Models, Theoretical , Prostate/pathology , Prostate-Specific Antigen/blood , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Androgen/metabolism , Sirolimus/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL