Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 159(3): 499-513, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25417103

ABSTRACT

Stromal communication with cancer cells can influence treatment response. We show that stromal and breast cancer (BrCa) cells utilize paracrine and juxtacrine signaling to drive chemotherapy and radiation resistance. Upon heterotypic interaction, exosomes are transferred from stromal to BrCa cells. RNA within exosomes, which are largely noncoding transcripts and transposable elements, stimulates the pattern recognition receptor RIG-I to activate STAT1-dependent antiviral signaling. In parallel, stromal cells also activate NOTCH3 on BrCa cells. The paracrine antiviral and juxtacrine NOTCH3 pathways converge as STAT1 facilitates transcriptional responses to NOTCH3 and expands therapy-resistant tumor-initiating cells. Primary human and/or mouse BrCa analysis support the role of antiviral/NOTCH3 pathways in NOTCH signaling and stroma-mediated resistance, which is abrogated by combination therapy with gamma secretase inhibitors. Thus, stromal cells orchestrate an intricate crosstalk with BrCa cells by utilizing exosomes to instigate antiviral signaling. This expands BrCa subpopulations adept at resisting therapy and reinitiating tumor growth.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/radiotherapy , Exosomes/metabolism , Paracrine Communication , Stromal Cells/metabolism , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Computer Simulation , Drug Resistance, Neoplasm , Female , Humans , Interferons/metabolism , Mice, Nude , Radiation Tolerance , Receptors, Notch/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction , rab GTP-Binding Proteins/metabolism
2.
CA Cancer J Clin ; 67(5): 378-397, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28763097

ABSTRACT

Answer questions and earn CME/CNE Recent decades have seen an unprecedented rise in obesity, and the health impact thereof is increasingly evident. In 2014, worldwide, more than 1.9 billion adults were overweight (body mass index [BMI], 25-29.9 kg/m2 ), and of these, over 600 million were obese (BMI ≥30 kg/m2 ). Although the association between obesity and the risk of diabetes and coronary artery disease is widely known, the impact of obesity on cancer incidence, morbidity, and mortality is not fully appreciated. Obesity is associated both with a higher risk of developing breast cancer, particularly in postmenopausal women, and with worse disease outcome for women of all ages. The first part of this review summarizes the relationships between obesity and breast cancer development and outcomes in premenopausal and postmenopausal women and in those with hormone receptor-positive and -negative disease. The second part of this review addresses hypothesized molecular mechanistic insights that may underlie the effects of obesity to increase local and circulating proinflammatory cytokines, promote tumor angiogenesis and stimulate the most malignant cancer stem cell population to drive cancer growth, invasion, and metastasis. Finally, a review of observational studies demonstrates that increased physical activity is associated with lower breast cancer risk and better outcomes. The effects of recent lifestyle interventions to decrease sex steroids, insulin/insulin-like growth factor-1 pathway activation, and inflammatory biomarkers associated with worse breast cancer outcomes in obesity also are discussed. Although many observational studies indicate that exercise with weight loss is associated with improved breast cancer outcome, further prospective studies are needed to determine whether weight reduction will lead to improved patient outcomes. It is hoped that several ongoing lifestyle intervention trials, which are reviewed herein, will support the systematic incorporation of weight loss intervention strategies into care for patients with breast cancer. CA Cancer J Clin 2017;67:378-397. © 2017 American Cancer Society.


Subject(s)
Breast Neoplasms/epidemiology , Obesity/epidemiology , Adipose Tissue/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/therapy , Comorbidity , Exercise , Female , Humans , Life Style , Obesity/metabolism , Postmenopause , Premenopause , Risk Factors , Weight Gain , Weight Loss
3.
Proc Natl Acad Sci U S A ; 116(14): 7005-7014, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30877256

ABSTRACT

p27 shifts from CDK inhibitor to oncogene when phosphorylated by PI3K effector kinases. Here, we show that p27 is a cJun coregulator, whose assembly and chromatin association is governed by p27 phosphorylation. In breast and bladder cancer cells with high p27pT157pT198 or expressing a CDK-binding defective p27pT157pT198 phosphomimetic (p27CK-DD), cJun is activated and interacts with p27, and p27/cJun complexes localize to the nucleus. p27/cJun up-regulates TGFB2 to drive metastasis in vivo. Global analysis of p27 and cJun chromatin binding and gene expression shows that cJun recruitment to many target genes is p27 dependent, increased by p27 phosphorylation, and activates programs of epithelial-mesenchymal transformation and metastasis. Finally, human breast cancers with high p27pT157 differentially express p27/cJun-regulated genes of prognostic relevance, supporting the biological significance of the work.


Subject(s)
Cell Movement , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Neoplasms/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Cell Adhesion , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p27/genetics , Humans , Neoplasms/genetics , Neoplasms/pathology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-jun/genetics
4.
Psychooncology ; 29(1): 182-194, 2020 01.
Article in English | MEDLINE | ID: mdl-31600424

ABSTRACT

OBJECTIVES: This paper presents the results of a study developed to inform the design of a multigenerational digital lifestyle intervention for overweight/obese women cancer survivors and their families. We followed the first six phases of the Integrate, Design, Assess, and Share (IDEAS) framework. METHODS: Grandmothers with breast, endometrial, or ovarian cancers (n = 46; 66.1 ± 0.9 years old; 34% Hispanic, 33% non-Hispanic black, 33% non-Hispanic white) self-reported their lifestyle behaviors, family structure, mobile device use, and interest in a family-based lifestyle intervention. A randomly selected subset of 21 participants subsequently completed qualitative interviews to understand their family relationships, weight-related challenges, and feedback on intervention prototypes. RESULTS: Participants reported low fruit intake (0.9 ± 0.1 servings/day), moderate vegetable intake (3.0 ± 0.2 servings/day), and high levels of moderate physical activity (990 ± 234 MET-minutes/week). The majority owned a smartphone (93%) and expressed interest in family-based programs (80%) that focused on weight management (91%). Qualitative data were collapsed into seven intervention considerations, including: capitalizing on existing familial support, involving local family who need lifestyle change, tapping into survivors' internal strengths, validating prior weight loss, overcoming barriers to sustained lifestyle change, providing information on cancer risk, and motivating families through reinforcing activities. CONCLUSIONS: Following the IDEAS framework, our next steps are to develop a fully-functioning prototype and conduct a randomized pilot trial to test the feasibility and effects of a digital intervention that empowers racially/ethnically diverse overweight/obese women cancer survivors to improve their physical activity and dietary intake and to lose weight by encouraging healthy lifestyle behaviors in their children and grandchildren.


Subject(s)
Cancer Survivors/psychology , Counseling/methods , Family Relations , Health Education/methods , Healthy Lifestyle , Obesity/psychology , Adult , Aged , Exercise , Female , Health Promotion/methods , Humans , Middle Aged , Obesity/complications , Patient Compliance , Weight Loss
5.
Breast Cancer Res Treat ; 156(2): 405-6, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26993130

ABSTRACT

Erratum to: Breast Cancer Res Treat (2013),138:369­381,DOI 10.1007/s10549-012-2389-6. In the original publication of the article, the Fig. 4c and d were published erroneously. The revised Fig. 4 is given in this erratum.

6.
Mol Cell ; 30(6): 701-11, 2008 Jun 20.
Article in English | MEDLINE | ID: mdl-18570873

ABSTRACT

The cell-cycle effects of mTORC1 are not fully understood. We provide evidence that mTOR-raptor phosphorylates SGK1 to modulate p27 function. Cellular mTOR activation, by refeeding of amino acid-deprived cells or by TSC2 shRNA, activated SGK1 and p27 phosphorylation at T157, and both were inhibited by short-term rapamycin treatment and by SGK1 shRNA. mTOR overexpression activated both Akt and SGK1, causing TGF-beta resistance through impaired nuclear import and cytoplasmic accumulation of p27. Rapamycin or raptor shRNA impaired mTOR-driven p70 and SGK1 activation, but not that of Akt, and decreased cytoplasmic p27. mTOR/raptor/SGK1 complexes were detected in cells. mTOR phosphorylated SGK1, but not SGK1-S422A, in vitro. SGK1 phosphorylated p27 in vitro. These data implicate SGK1 as an mTORC1 (mTOR-raptor) substrate. mTOR may promote G1 progression in part through SGK1 activation and deregulate the cell cycle in cancers through both Akt- and SGK-mediated p27 T157 phosphorylation and cytoplasmic p27 mislocalization.


Subject(s)
Cell Cycle/physiology , Immediate-Early Proteins/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , Enzyme Activation , Homeostasis , Humans , Kinetics , Melanoma , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Recombinant Proteins/metabolism , TOR Serine-Threonine Kinases , Transfection
7.
Annu Rev Med ; 64: 45-57, 2013.
Article in English | MEDLINE | ID: mdl-23121183

ABSTRACT

Obesity is a problem of epidemic proportions in many developed nations. Increased body mass index and obesity are associated with a significantly worse outcome for many cancers. Breast cancer risk in the postmenopausal setting and poor disease outcome for all patients is significantly augmented in overweight and obese individuals. The expansion of fat tissue involves a complex interaction of endocrine factors known as adipokines and cytokines. High cytokine levels in primary breast cancers and in the circulation of affected patients have been associated with poor outcome. This review summarizes the how cytokine production in obese adipose tissue creates a chronic inflammatory microenvironment that favors tumor cell motility, invasion, and epithelial-mesenchymal transition to enhance the metastatic potential of tumor cells. Many of the cytokines associated with a proinflammatory state are not only upregulated in obese adipose tissue but may also stimulate the self-renewal of cancer stem cells. Thus, enhanced cytokine production in obese adipose tissue may serve both as a chemoattractant for invading cancers and to augment their malignant potential. These new mechanistic insights suggest that the current obesity epidemic will presage a significant increase in cancer incidence, morbidity, and mortality in the next few decades.


Subject(s)
Cytokines/metabolism , Neoplasms , Obesity , Body Mass Index , Disease Progression , Global Health , Humans , Incidence , Neoplasms/complications , Neoplasms/epidemiology , Neoplasms/metabolism , Obesity/complications , Obesity/epidemiology , Obesity/metabolism , Risk Factors
8.
Breast Cancer Res Treat ; 144(3): 503-17, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24567196

ABSTRACT

Our goal was to establish primary cultures from dissociation of breast tumors in order to provide cellular models that may better recapitulate breast cancer pathogenesis and the metastatic process. Here, we report the characterization of six cellular models derived from the dissociation of primary breast tumor specimens, referred to as "dissociated tumor (DT) cells." In vitro, DT cells were characterized by proliferation assays, colony formation assays, protein, and gene expression profiling, including PAM50 predictor analysis. In vivo, tumorigenic and metastatic potential of DT cultures was assessed in NOD/SCID and NSG mice. These cellular models differ from recently developed patient-derived xenograft models in that they can be used for both in vitro and in vivo studies. PAM50 predictor analysis showed DT cultures similar to their paired primary tumor and as belonging to the basal and Her2-enriched subtypes. In vivo, three DT cultures are tumorigenic in NOD/SCID and NSG mice, and one of these is metastatic to lymph nodes and lung after orthotopic inoculation into the mammary fat pad, without excision of the primary tumor. Three DT cultures comprised of cancer-associated fibroblasts (CAFs) were isolated from luminal A, Her2-enriched, and basal primary tumors. Among the DT cells are those that are tumorigenic and metastatic in immunosuppressed mice, offering novel cellular models of ER-negative breast cancer subtypes. A group of CAFs provide tumor subtype-specific components of the tumor microenvironment (TME). Altogether, these DT cultures provide closer-to-primary cellular models for the study of breast cancer pathogenesis, metastasis, and TME.


Subject(s)
Breast Neoplasms/pathology , Primary Cell Culture , Animals , Biomarkers, Tumor/metabolism , Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Cycle , Cell Proliferation , Cell Transformation, Neoplastic , Disease Models, Animal , Female , Fibroblasts/pathology , Gene Expression Profiling , Heterografts , Humans , Immunohistochemistry , Mice , Neoplasm Metastasis , Primary Cell Culture/methods , Tumor Burden , Tumor Cells, Cultured , Tumor Stem Cell Assay
9.
Nat Cell Biol ; 9(2): 218-24, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17237771

ABSTRACT

Nutrients and bioenergetics are prerequisites for proliferation and survival of mammalian cells. We present evidence that the cyclin-dependent kinase inhibitor p27(Kip1), is phosphorylated at Thr 198 downstream of the Peutz-Jeghers syndrome protein-AMP-activated protein kinase (LKB1-AMPK) energy-sensing pathway, thereby increasing p27 stability and directly linking sensing of nutrient concentration and bioenergetics to cell-cycle progression. Ectopic expression of wild-type and phosphomimetic Thr 198 to Asp 198 (T198D), but not unstable Thr 198 to Ala 198 (p27(T198A)) is sufficient to induce autophagy. Under stress conditions that activate the LKB1-AMPK pathway with subsequent induction of autophagy, p27 knockdown results in apoptosis. Thus LKB1-AMPK pathway-dependent phosphorylation of p27 at Thr 198 stabilizes p27 and permits cells to survive growth factor withdrawal and metabolic stress through autophagy. This may contribute to tumour-cell survival under conditions of growth factor deprivation, disrupted nutrient and energy metabolism, or during stress of chemotherapy.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Energy Metabolism , Multienzyme Complexes/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases , Cell Line, Tumor , Humans , Phosphorylation , Signal Transduction/physiology
10.
Front Nutr ; 11: 1301427, 2024.
Article in English | MEDLINE | ID: mdl-38660060

ABSTRACT

Background: High glycemic variability (GV) is a biomarker of cancer risk, even in the absence of diabetes. The emerging concept of chrononutrition suggests that modifying meal timing can favorably impact metabolic risk factors linked to diet-related chronic disease, including breast cancer. Here, we examined the potential of eating when glucose levels are near personalized fasting thresholds (low-glucose eating, LGE), a novel form of timed-eating, to reduce GV in women without diabetes, who are at risk for postmenopausal breast cancer. Methods: In this exploratory analysis of our 16-week weight loss randomized controlled trial, we included 17 non-Hispanic, white, postmenopausal women (average age = 60.7 ± 5.8 years, BMI = 34.5 ± 6.1 kg/m2, HbA1c = 5.7 ± 0.3%). Participants were those who, as part of the parent study, provided 3-7 days of blinded, continuous glucose monitoring data and image-assisted, timestamped food records at weeks 0 and 16. Pearson's correlation and multivariate regression were used to assess associations between LGE and GV, controlling for concurrent weight changes. Results: Increases in LGE were associated with multiple unfavorable measures of GV including reductions in CGM glucose mean, CONGA, LI, J-Index, HBGI, ADDR, and time spent in a severe GV pattern (r = -0.81 to -0.49; ps < 0.044) and with increases in favorable measures of GV including M-value and LBGI (r = 0.59, 0.62; ps < 0.013). These associations remained significant after adjusting for weight changes. Conclusion: Low-glucose eating is associated with improvements in glycemic variability, independent of concurrent weight reductions, suggesting it may be beneficial for GV-related disease prevention. Further research in a larger, more diverse sample with poor metabolic health is warranted.Clinical trial registration: ClinicalTrials.gov, NCT03546972.

11.
Nat Commun ; 15(1): 5152, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38886396

ABSTRACT

In many cancers, a stem-like cell subpopulation mediates tumor initiation, dissemination and drug resistance. Here, we report that cancer stem cell (CSC) abundance is transcriptionally regulated by C-terminally phosphorylated p27 (p27pT157pT198). Mechanistically, this arises through p27 co-recruitment with STAT3/CBP to gene regulators of CSC self-renewal including MYC, the Notch ligand JAG1, and ANGPTL4. p27pTpT/STAT3 also recruits a SIN3A/HDAC1 complex to co-repress the Pyk2 inhibitor, PTPN12. Pyk2, in turn, activates STAT3, creating a feed-forward loop increasing stem-like properties in vitro and tumor-initiating stem cells in vivo. The p27-activated gene profile is over-represented in STAT3 activated human breast cancers. Furthermore, mammary transgenic expression of phosphomimetic, cyclin-CDK-binding defective p27 (p27CK-DD) increases mammary duct branching morphogenesis, yielding hyperplasia and microinvasive cancers that can metastasize to liver, further supporting a role for p27pTpT in CSC expansion. Thus, p27pTpT interacts with STAT3, driving transcriptional programs governing stem cell expansion or maintenance in normal and cancer tissues.


Subject(s)
Breast Neoplasms , Cyclin-Dependent Kinase Inhibitor p27 , Hyperplasia , Neoplastic Stem Cells , STAT3 Transcription Factor , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Humans , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Animals , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Female , Phosphorylation , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Hyperplasia/metabolism , Mice , Gene Expression Regulation, Neoplastic , Cell Self Renewal/genetics , Cell Line, Tumor , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Glands, Animal/cytology , Jagged-1 Protein/metabolism , Jagged-1 Protein/genetics
12.
Breast Cancer Res Treat ; 138(2): 369-81, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23430223

ABSTRACT

Oncogenic PI3K/mTOR activation is frequently observed in human cancers and activates cell motility via p27 phosphorylations at T157 and T198. Here we explored the potential for a novel PI3K/mTOR inhibitor to inhibit tumor invasion and metastasis. An MDA-MB-231 breast cancer line variant, MDA-MB-231-1833, with high metastatic bone tropism, was treated with a novel catalytic PI3K/mTOR inhibitor, PF-04691502, at nM doses that did not impair proliferation. Effects on tumor cell motility, invasion, p27 phosphorylation, localization, and bone metastatic outgrowth were assayed. MDA-MB-231-1833 showed increased PI3K/mTOR activation, high levels of cytoplasmic p27pT157pT198 and increased cell motility and invasion in vitro versus parental. PF-04691502 treatment, at a dose that did not affect proliferation, reduced total and cytoplasmic p27, decreased p27pT157pT198 and restored cell motility and invasion to levels seen in MDA-MB-231. p27 knockdown in MDA-MB-231-1833 phenocopied PI3K/mTOR inhibition, whilst overexpression of the phosphomimetic mutant p27T157DT198D caused resistance to the anti-invasive effects of PF-04691502. Pre-treatment of MDA-MB-231-1833 with PF-04691502 significantly impaired metastatic tumor formation in vivo, despite lack of antiproliferative effects in culture and little effect on primary orthotopic tumor growth. A further link between cytoplasmic p27 and metastasis was provided by a study of primary human breast cancers which showed cytoplasmic p27 is associated with increased lymph nodal metastasis and reduced survival. Novel PI3K/mTOR inhibitors may oppose tumor metastasis independent of their growth inhibitory effects, providing a rationale for clinical investigation of PI3K/mTOR inhibitors in settings to prevent micrometastasis. In primary human breast cancers, cytoplasmic p27 is associated with worse outcomes and increased nodal metastasis, and may prove useful as a marker of both PI3K/mTOR activation and PI3K/mTOR inhibitor efficacy.


Subject(s)
Bone Neoplasms/prevention & control , Breast Neoplasms/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Pyridones/pharmacology , Pyrimidines/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Bone Neoplasms/mortality , Bone Neoplasms/secondary , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cytoplasm/metabolism , Disease-Free Survival , Female , Gene Expression , Gene Knockdown Techniques , Humans , Kaplan-Meier Estimate , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Targeted Therapy , Neoplasm Invasiveness , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
13.
Proc Natl Acad Sci U S A ; 106(23): 9268-73, 2009 Jun 09.
Article in English | MEDLINE | ID: mdl-19470470

ABSTRACT

p90 ribosomal S6 kinase (RSK1) is an effector of both Ras/MEK/MAPK and PI3K/PDK1 pathways. We present evidence that RSK1 drives p27 phosphorylation at T198 to increase RhoA-p27 binding and cell motility. RSK1 activation and p27pT198 both increase in early G(1). As for many kinase-substrate pairs, cellular RSK1 coprecipitates with p27. siRNA to RSK1 and RSK1 inhibition both rapidly reduce cellular p27pT198. RSK1 overexpression increases p27pT198, p27-cyclin D1-Cdk4 complexes, and p27 stability. Moreover, RSK1 transfectants show mislocalization of p27 to cytoplasm, increased motility, and reduced RhoA-GTP, phospho-cofilin, and actin stress fibers, all of which were reversed by shRNA to p27. Phosphorylation by RSK1 increased p27pT198 binding to RhoA in vitro, whereas p27T157A/T198A bound poorly to RhoA compared with WTp27 in cells. Coprecipitation of cellular p27-RhoA was increased in cells with constitutive PI3K activation and increased in early G(1). Thus T198 phosphorylation not only stabilizes p27 and mislocalizes p27 to the cytoplasm but also promotes RhoA-p27 interaction and RhoA pathway inhibition. These data link p27 phosphorylation at T198 and cell motility. As for other PI3K effectors, RSK1 phosphorylates p27 at T198. Because RSK1 is also activated by MAPK, the increased cell motility and metastatic potential of cancer cells with PI3K and/or MAPK pathway activation may result in part from RSK1 activation, leading to accumulation of p27T198 in the cytoplasm, p27:RhoA binding, inhibition of RhoA/Rock pathway activation, and loss of actomyosin stability.


Subject(s)
Cell Movement , Intracellular Signaling Peptides and Proteins/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , rhoA GTP-Binding Protein/metabolism , Actins/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p27 , Cytoplasm/metabolism , G1 Phase , Humans , MAP Kinase Signaling System , Phosphorylation , Signal Transduction
14.
Cell Rep ; 41(7): 111672, 2022 11 15.
Article in English | MEDLINE | ID: mdl-36384125

ABSTRACT

Recent work showed that the dominant post-menopausal estrogen, estrone, cooperates with nuclear factor κB (NF-κB) to stimulate inflammation, while pre-menopausal 17ß-estradiol opposes NF-κB. Here, we show that post-menopausal estrone, but not 17ß-estradiol, activates epithelial-to-mesenchymal transition (EMT) genes to stimulate breast cancer metastasis. HSD17B14, which converts 17ß-estradiol to estrone, is higher in cancer than normal breast tissue and in metastatic than primary cancers and associates with earlier metastasis. Treatment with estrone, but not 17ß-estradiol, and HSD17B14 overexpression both stimulate an EMT, matrigel invasion, and lung, bone, and liver metastasis in estrogen-receptor-positive (ER+) breast cancer models, while HSD17B14 knockdown reverses the EMT. Estrone:ERα recruits CBP/p300 to the SNAI2 promoter to induce SNAI2 and stimulate an EMT, while 17ß-estradiol:ERα recruits co-repressors HDAC1 and NCOR1 to this site. Present work reveals novel differences in gene regulation by these estrogens and the importance of estrone to ER+ breast cancer progression. Upon loss of 17ß-estradiol at menopause, estrone-liganded ERα would promote ER+ breast cancer invasion and metastasis.


Subject(s)
Breast Neoplasms , Epithelial-Mesenchymal Transition , Estrone , Snail Family Transcription Factors , Female , Humans , 17-Hydroxysteroid Dehydrogenases , Breast Neoplasms/pathology , Estradiol/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Estrone/metabolism , NF-kappa B , Postmenopause , Snail Family Transcription Factors/genetics , Neoplasm Metastasis
15.
Clin Cancer Res ; 28(9): 1948-1965, 2022 05 02.
Article in English | MEDLINE | ID: mdl-35135840

ABSTRACT

PURPOSE: Although chemotherapies kill most cancer cells, stem cell-enriched survivors seed metastasis, particularly in triple-negative breast cancers (TNBC). TNBCs arise from and are enriched for tumor stem cells. Here, we tested if inhibition of DOT1L, an epigenetic regulator of normal tissue stem/progenitor populations, would target TNBC stem cells. EXPERIMENTAL DESIGN: Effects of DOT1L inhibition by EPZ-5676 on stem cell properties were tested in three TNBC lines and four patient-derived xenograft (PDX) models and in isolated cancer stem cell (CSC)-enriched ALDH1+ and ALDH1- populations. RNA sequencing compared DOT1L regulated pathways in ALDH1+ and ALDH1- cells. To test if EPZ-5676 decreases CSC in vivo, limiting dilution assays of EPZ-5676/vehicle pretreated ALDH1+ and ALDH1- cells were performed. Tumor latency, growth, and metastasis were evaluated. Antitumor activity was also tested in TNBC PDX and PDX-derived organoids. RESULTS: ALDH1+ TNBC cells exhibit higher DOT1L and H3K79me2 than ALDH1-. DOT1L maintains MYC expression and self-renewal in ALDH1+ cells. Global profiling revealed that DOT1L governs oxidative phosphorylation, cMyc targets, DNA damage response, and WNT activation in ALDH1+ but not in ALDH1- cells. EPZ-5676 reduced tumorspheres and ALDH1+ cells in vitro and decreased tumor-initiating stem cells and metastasis in xenografts generated from ALDH1+ but not ALDH1- populations in vivo. EPZ-5676 significantly reduced growth in vivo of one of two TNBC PDX tested and decreased clonogenic 3D growth of two other PDX-derived organoid cultures. CONCLUSIONS: DOT1L emerges as a key CSC regulator in TNBC. Present data support further clinical investigation of DOT1L inhibitors to target stem cell-enriched TNBC.


Subject(s)
Triple Negative Breast Neoplasms , Aldehyde Dehydrogenase 1 Family , Cell Line, Tumor , Histone-Lysine N-Methyltransferase/metabolism , Humans , Neoplastic Stem Cells/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Xenograft Model Antitumor Assays
16.
Breast Cancer Res Treat ; 128(1): 69-78, 2011 Jul.
Article in English | MEDLINE | ID: mdl-20669046

ABSTRACT

Antiestrogen therapies arrest susceptible estrogen receptor (ER)-positive breast cancers by increasing p27. Since Src phosphorylates p27 to promote p27 proteolysis, Src activation observed in up to 40% of ER-positive cancers may contribute to antiestrogen resistance. In this article, we show that treatment with the Src-inhibitor saracatinib (AZD0530) together with ER-blocking drugs increased breast cancer cell cycle arrest via p27. Saracatinib and fulvestrant together more effectively increased p27, reduced Ki67, and impaired MDA-MB-361 xenograft tumor growth in vivo than either of the drugs alone. In contrast, saracatinib monotherapy rapidly gave rise to drug resistance. Since combined ER and Src inhibition delays development of resistance in vivo, these data support further clinical investigation of saracatinib in combination with fulvestrant for women with ER-positive breast cancer. Proteomic analysis revealed striking bypass activation of the mTOR pathway in saracatinib-resistant tumors. mTORC1 activation also arose following long-term culture of ER-positive breast cancer lines in the presence of saracatinib. These data indicate the utility of proteomic analysis of drug-resistant tumors to identify potential means of drug resistance. The use of mTOR kinase inhibitors with saracatinib may subvert drug resistance and prove to be more effective than saracatinib alone.


Subject(s)
Antineoplastic Agents/pharmacology , Benzodioxoles/pharmacology , Estradiol/analogs & derivatives , Estrogen Receptor alpha/antagonists & inhibitors , Quinazolines/pharmacology , Tamoxifen/pharmacology , src-Family Kinases/antagonists & inhibitors , Animals , Breast Neoplasms , Cell Line, Tumor , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Drug Resistance, Neoplasm , Drug Synergism , ErbB Receptors/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Female , Fulvestrant , G1 Phase/drug effects , Humans , Ki-67 Antigen/metabolism , Mice , Mice, Nude , Neoplasms, Hormone-Dependent , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
17.
Nat Med ; 8(10): 1153-60, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12244302

ABSTRACT

Mechanisms linking mitogenic and growth inhibitory cytokine signaling and the cell cycle have not been fully elucidated in either cancer or in normal cells. Here we show that activation of protein kinase B (PKB)/Akt, contributes to resistance to antiproliferative signals and breast cancer progression in part by impairing the nuclear import and action of p27. Akt transfection caused cytoplasmic p27 accumulation and resistance to cytokine-mediated G1 arrest. The nuclear localization signal of p27 contains an Akt consensus site at threonine 157, and p27 phosphorylation by Akt impaired its nuclear import in vitro. Akt phosphorylated wild-type p27 but not p27T157A. In cells transfected with constitutively active Akt(T308DS473D)(PKB(DD)), p27WT mislocalized to the cytoplasm, but p27T157A was nuclear. In cells with activated Akt, p27WT failed to cause G1 arrest, while the antiproliferative effect of p27T157A was not impaired. Cytoplasmic p27 was seen in 41% (52 of 128) of primary human breast cancers in conjunction with Akt activation and was correlated with a poor patient prognosis. Thus, we show a novel mechanism whereby Akt impairs p27 function that is associated with an aggressive phenotype in human breast cancer.


Subject(s)
Active Transport, Cell Nucleus/physiology , CDC2-CDC28 Kinases , Cell Cycle Proteins/metabolism , G1 Phase/physiology , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle Proteins/genetics , Cell Line , Cyclin E/metabolism , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/metabolism , Enzyme Inhibitors/metabolism , Female , Humans , Mitogen-Activated Protein Kinases/metabolism , Mutagenesis, Site-Directed , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Survival Rate , Threonine/metabolism , Transforming Growth Factor beta/metabolism , Tumor Suppressor Proteins/genetics
18.
Theranostics ; 11(8): 3552-3564, 2021.
Article in English | MEDLINE | ID: mdl-33664847

ABSTRACT

Rationale: The clinical use of PI3K inhibitors, such as buparlisib, has been plagued with toxicity at effective doses. The aim of this study is to determine if vitamin C, a potent epigenetic regulator, can improve the therapeutic outcome and reduce the dose of buparlisib in treating PIK3CA-mutated triple negative breast cancer (TNBC). Methods: The response of TNBC cells to buparlisib was assessed by EC50 measurements, apoptosis assay, clonogenic assay, and xenograft assay in mice. Molecular approaches including Western blot, immunofluorescence, RNA sequencing, and gene silencing were utilized as experimental tools. Results: Treatment with buparlisib at lower doses, along with vitamin C, induced apoptosis and inhibited the growth of TNBC cells in vitro. Vitamin C via oral delivery rendered a sub-therapeutic dose of buparlisib able to inhibit TNBC xenograft growth and to markedly block metastasis in mice. We discovered that buparlisib and vitamin C coordinately reduced histone H3K4 methylation by enhancing the nuclear translocation of demethylase, KDM5, and by serving as a cofactor to promote KDM5-mediated H3K4 demethylation. The expression of genes in the PI3K pathway, such as AKT2 and mTOR, was suppressed by vitamin C in a KDM5-dependent manner. Vitamin C and buparlisib cooperatively blocked AKT phosphorylation. Inhibition of KDM5 largely abolished the effect of vitamin C on the response of TNBC cells to buparlisib. Additionally, vitamin C and buparlisib co-treatment changed the expression of genes, including PCNA and FILIP1L, which are critical to cancer growth and metastasis. Conclusion: Vitamin C can be used to reduce the dosage of buparlisib needed to produce a therapeutic effect, which could potentially ease the dose-dependent side effects in patients.


Subject(s)
Ascorbic Acid/administration & dosage , Phosphoinositide-3 Kinase Inhibitors/administration & dosage , Triple Negative Breast Neoplasms/drug therapy , Aminopyridines/administration & dosage , Animals , Apoptosis/drug effects , Cell Line, Tumor , Drug Synergism , Female , Gene Expression Regulation, Neoplastic/drug effects , Histone Code/drug effects , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Targeted Therapy , Morpholines/administration & dosage , Precision Medicine , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
19.
Blood Adv ; 5(23): 5072-5085, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34543391

ABSTRACT

Human germinal center-associated lymphoma (HGAL) is an adaptor protein specifically expressed in germinal center lymphocytes. High expression of HGAL is a predictor of prolonged survival of diffuse large B-cell lymphoma (DLBCL) and classic Hodgkin lymphoma. Furthermore, HGAL expression is associated with early-stage DLBCL, thus potentially limiting lymphoma dissemination. In our previous studies, we demonstrated that HGAL regulates B-cell receptor signaling and cell motility in vitro and deciphered some molecular mechanisms underlying these effects. By using novel animal models for in vivo DLBCL dispersion, we demonstrate here that HGAL decreases lymphoma dissemination and prolongs survival. Furthermore, by using an unbiased proteomic approach, we demonstrate that HGAL may interact with multiple cytoskeletal proteins thereby implicating a multiplicity of effects in regulating lymphoma motility and spread. Specifically, we show that HGAL interacts with tubulin, and this interaction may also contribute to HGAL effects on cell motility. These findings recapitulate previous observations in humans, establish the role of HGAL in dissemination of lymphoma in vivo, and explain improved survival of patients with HGAL-expressing lymphomas.


Subject(s)
Cytoskeletal Proteins , Intracellular Signaling Peptides and Proteins/metabolism , Lymphoma, Large B-Cell, Diffuse , Microfilament Proteins/metabolism , Animals , Cytoskeletal Proteins/genetics , Humans , Mice , Mice, Transgenic , Neoplasm Proteins , Proteomics
20.
J Clin Invest ; 117(8): 2205-15, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17627304

ABSTRACT

Estrogen drives both transcriptional activation and proteolysis of estrogen receptor alpha (ER alpha; encoded by ESR1). Here we observed variable and overlapping ESR1 mRNA levels in 200 ER alpha-negative and 50 ER alpha-positive primary breast cancers examined, which suggests important posttranscriptional ER alpha regulation. Our results indicate that Src cooperates with estrogen to activate ER alpha proteolysis. Inducible Src stimulated ligand-activated ER alpha transcriptional activity and reduced ER alpha t(1/2). Src and ER alpha levels were inversely correlated in primary breast cancers. ER alpha-negative primary breast cancers and cell lines showed increased Src levels and/or activity compared with ER alpha-positive cancers and cells. ER alpha t(1/2) was reduced in ER alpha-negative cell lines. In both ER alpha-positive and -negative cell lines, both proteasome and Src inhibitors increased ER alpha levels. Src inhibition impaired ligand-activated ER alpha ubiquitylation and increased ER alpha levels. Src siRNA impaired ligand-activated ER alpha loss in BT-20 cells. Pretreatment with Src increased ER alpha ubiquitylation and degradation in vitro. These findings provide what we believe to be a novel link between Src activation and ER alpha proteolysis and support a model whereby crosstalk between liganded ER alpha and Src drives ER alpha transcriptional activity and targets ER alpha for ubiquitin-dependent proteolysis. Oncogenic Src activation may promote not only proliferation, but also estrogen-activated ER alpha loss in a subset of ER alpha-negative breast cancers, altering prognosis and response to therapy.


Subject(s)
Breast Neoplasms/metabolism , Estrogen Receptor alpha/biosynthesis , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/biosynthesis , Protein Processing, Post-Translational , src-Family Kinases/metabolism , Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Cell Line, Tumor , Enzyme Activation/drug effects , Estrogens/metabolism , Estrogens/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Prognosis , Proteasome Endopeptidase Complex/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , RNA, Small Interfering/pharmacology , Transcription, Genetic/drug effects , Ubiquitin/metabolism , src-Family Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL